lega italiana per la lotta contro la malattia di parkinson le ... - Limpe

lega italiana per la lotta contro la malattia di parkinson le ... - Limpe lega italiana per la lotta contro la malattia di parkinson le ... - Limpe

12.07.2015 Views

LIMPElega italiana per la lotta controla malattia di parkinson le sindromiextrapiramidali e le demenzeWeb site: http://www.parkinson-limpe.it/XXVIII National CongressAlba (Cuneo)7–9 November 2001Experimental Models and Clinical Correlations inExtrapyramidal Diseases and DementiaSELECTED SHORT PAPERSScientific Committee: Bruno Bergamasco, Giovanni Corsini, Mario Manfredi, Stefano Ruggieri,Guglielmo ScarlatoSecretariat LIMPE: Viale dell’Università 30, 00185 RomaTel. and Fax: +39-06-4455618E-mail: limpe@interfree.it

LIMPE<strong><strong>le</strong>ga</strong> <strong>italiana</strong> <strong>per</strong> <strong>la</strong> <strong>lotta</strong> <strong>contro</strong><strong>la</strong> ma<strong>la</strong>ttia <strong>di</strong> <strong>parkinson</strong> <strong>le</strong> sindromiextrapiramidali e <strong>le</strong> demenzeWeb site: http://www.<strong>parkinson</strong>-limpe.it/XXVIII National CongressAlba (Cuneo)7–9 November 2001Ex<strong>per</strong>imental Models and Clinical Corre<strong>la</strong>tions inExtrapyramidal Diseases and DementiaSELECTED SHORT PAPERSScientific Committee: Bruno Bergamasco, Giovanni Corsini, Mario Manfre<strong>di</strong>, Stefano Ruggieri,Guglielmo Scar<strong>la</strong>toSecretariat LIMPE: Via<strong>le</strong> dell’Università 30, 00185 RomaTel. and Fax: +39-06-4455618E-mail: limpe@interfree.it


S50Patient MMSE Short ta<strong>le</strong> Verbal span Gollin Corsi PM38A 27 15.2 3 136 4 18/48B 26 12.5 4 82 5 16/48Patients and <strong>contro</strong>ls adapted rather easily to the virtua<strong>le</strong>nvironment, following all items required in the time needed bythe ex<strong>per</strong>imental procedures. They familiarized with VR interfaceand named objects and described rooms (phases 1 and 2).In Fig. 1, we report the fin<strong>di</strong>ngs concerning the tasks <strong>per</strong>formed.In comparison with <strong>contro</strong>ls, the two <strong>parkinson</strong>ianpatients showed a mild <strong>di</strong>fficulty in pointing tasks, incidentalmemory tasks and orientation. A stronger involvement was seenby the evaluation of the speed of execution: both PD patientswere slower during all trials, especially when they were askedto walk through doors or narrow spaces such as in the bathroom.DiscussionHuman reactions are <strong>di</strong>fferent accor<strong>di</strong>ng to the received information.If there is a <strong>di</strong>rect visual feedback, action can beimme<strong>di</strong>ate. It is possib<strong>le</strong> to quantify the influence of biofeedback,accor<strong>di</strong>ng to the use of an oriented para<strong>di</strong>gm of equipmentand of tests, with the reliab<strong>le</strong> software [5, 6].Activities like cooking or driving a car need an involvement(in terms of quantity and duration) of su<strong>per</strong>ior corticalfunctions, especially those requiring attention, that cannot besufficiently explored by tra<strong>di</strong>tional neuropsycological tests.The two patients examinated in this study are at an earlystage of <strong>di</strong>sease, with a rather brief duration of symptomsand a moderate motor <strong>di</strong>sability. Compared with <strong>contro</strong>ls,400350our fin<strong>di</strong>ngs show a marked <strong>di</strong>fference in the speed score.The virtual narrow space exaggerates this trend, even if inreal life these patients do not manifest any freezing phenomenaor motor blocks. Concerning the other tasks <strong>per</strong>formed,the PD patients show a mild <strong>di</strong>fficulty in pointing tasks, incidentalmemory and orientation.The use of VR instrumental offers new opportunities inneurorehabilitation, by:1. Supporting the tra<strong>di</strong>tional clinical approach by means of thedetection of pre<strong>di</strong>ctive markers of <strong>di</strong>sexecutive <strong>di</strong>sorders, and2. Establishing a rehabilitation protocol, teaching patients toapproach their <strong>di</strong>sabilities, in favor of autonomy, self-efficacy,social integration, and improvement of quality of life.References1. Moline J (1997) Virtual reality for health care: a survey. In:Riva G (ed) Virtual reality in neuro-psycho-physiology. IOS,Amsterdam, pp 3–342. Baati<strong>le</strong> J, Langbein WE, Weaver F, Maloney C, Jost MB (2000)Effect of exercise on <strong>per</strong>ceived quality of life of in<strong>di</strong>vidualswith Parkinson’s <strong>di</strong>sease. J Rehabil Res Dev 37(5): 529–5343. Wolfson L, Judge J, Whipp<strong>le</strong> R, King M (1995) Strength is amaior factor in ba<strong>la</strong>nce, gait, and the occurrence of falls. JGerontol 50:64–674. Stern GM, Lander CM, Lees AJ (1980) Akinetic freezing and trickmovements in Parkinson’s <strong>di</strong>sease. J Neural Transm 16:137–1415. Rovetta A, Lorini F, Canina M (1988) A new project for rehabilitationand psychomotor <strong>di</strong>sease analysis with virtual reality support.Stu<strong>di</strong>es in health technology and informatics 50:180–1856. Shallice T, Burgess PW (1991) Deficits in strategy applicationfollowing frontal lobe damage in man. Brain 114(Pt 2):727–741a300250Time, s20015010050Score1510500BathroomBathroom Food Orientation Home Bathroomentrance entranceLunch tab<strong>le</strong>bCorrect <strong>per</strong>formances, n86420IMTOrientationcFig. 1a-c Results obtained inthe VR session for 10 normalsubjects and 2 Parkinson’s <strong>di</strong>seasepatients. a Speed trials. bPointing tasks. c Number ofcorrect <strong>per</strong>formances on incidentalmemory task (IMT) andorientation task. ■ , normalsubjects; ■ , PD patients


S52from healthy <strong>contro</strong>ls. Our results confirm that striatal DATbin<strong>di</strong>ng is a sensitive measurement of degeneration in thedopaminergic system of PD patients. Moreover, <strong>di</strong>fferenceswere found when comparing putaminal values between earlyand <strong>la</strong>te onset PD.The presence of greater decrements in the putamen thanin the caudate nuc<strong>le</strong>us is consistent with neuropathologicaldata showing that the bulk of nigral loss affects the dopaminergicneurons projecting to the putamen. Our results are alsoconsistent with previous stu<strong>di</strong>es with [ 11 F]fluorodopa/PETand [ 123 I]CIT or [ 123 I]FPCIT SPECT. Considering recentdata suggesting that <strong>di</strong>sease may progress at a slower pace ifpatients are treated early with dopamine agonists, it becomescritical to identify those in<strong>di</strong>viduals as soon as motor symptomsappear [4]. More importantly it may be helpful to evaluatepopu<strong>la</strong>tions at genetic or environmental risk for PD [5].In this vein, neuroimaging markers may prove useful in supportingthe clinical suspicion with a <strong>di</strong>rect demonstration ofdopaminergic denervation.Finally, it is interesting that the patient with autosomalrecessive <strong>parkinson</strong>ism presented with a greater denervationthan other early onset counterparts. Our data suggest thatdopaminergic loss is marked already a few years after symptomsonset in in<strong>di</strong>viduals with early onset <strong>parkinson</strong>ism, particu<strong>la</strong>rlyif genetically determined. These fin<strong>di</strong>ngs may bere<strong>le</strong>vant in the early development of motor complications(dyskinesias and motor fluctuations) in these patients.References1. Feigin A, Antonini A, Benti R et al (2002) Tc-99m ECDSPECT in the <strong>di</strong>fferential <strong>di</strong>agnosis of <strong>parkinson</strong>ism. MovDisord (in press)2. Pirker W, Djamshi<strong>di</strong>an S, Asenbaum S et al (2002)Progression of dopaminergic degeneration in Parkinson’s <strong>di</strong>seaseand atypical <strong>parkinson</strong>ism: a longitu<strong>di</strong>nal beta-CITSPECT study. Mov Disord 17(1):45–533. Antonini A, Moresco RM, Gobbo C (2001) The status ofdopamine nerve terminals in Parkinson’s <strong>di</strong>sease and essentialtremor: a PET study with the tracer [ 11 C]FE-CIT. Neurol Sci22:47–484. Marek K, Seibyl J, Shoulson I et al (2002) Dopamine transporterbrain imaging to assess the effects of pramipexo<strong>le</strong> vs<strong>le</strong>vodopa on Parkinson <strong>di</strong>sease progression. JAMA287(13):1653–16615. Piccini P, Burn DJ, Ceravolo R, Maraganore D, Brooks DJ(1999) The ro<strong>le</strong> of inheritance in spora<strong>di</strong>c Parkinson’s <strong>di</strong>sease:evidence from a longitu<strong>di</strong>nal study of dopaminergicfunction in twins. Ann Neurol 45(5):577–582


Neurol Sci (2002) 23:S53–S54 © Springer-Ver<strong>la</strong>g 2002Body weight, <strong>le</strong>vodopapharmacokinetics and dyskinesiain Parkinson’s <strong>di</strong>seaseG. Arabia 1 • M. Zappia 1,2 () • D. Bosco 2L. Crescibene 2 • A. Bagalà 2 • L. Bastone 2M. Caracciolo 2 • M. Scornaienghi 2 • A. Quattrone 1,21Institute of Neurology, University of Catanzaro, Policlinico MaterDomini, Via T. Campanel<strong>la</strong>, I-88100 Catanzaro, Italy2Institute of Neurological Sciences, National Research Council,Piano Lago <strong>di</strong> Mangone (CS), ItalyAbstract We conducted a pharmacokinetic study in 164patients with spora<strong>di</strong>c Parkinson’s <strong>di</strong>sease (PD) to addressthe re<strong>la</strong>tionship between body weight and <strong>le</strong>vodopa pharmacokinetics.Patients underwent an oral acute <strong>le</strong>vodopa testwith 250 mg <strong>le</strong>vodopa and pharmacokinetic variab<strong>le</strong>s werefurther assessed. P<strong>la</strong>smatic <strong>le</strong>vodopa area under the curve(AUC-l) and body weight were significantly and inverselycorre<strong>la</strong>ted. Women were significantly lighter and more dyskineticthan men, and had greater AUC-l values. Our datasuggest that during long-term treatment, lighter PD patients,especially women, may receive a greater cumu<strong>la</strong>tive dosageof <strong>le</strong>vodopa <strong>per</strong> kilogram of body weight. This could exp<strong>la</strong>ingender <strong>di</strong>fferences for the development of <strong>le</strong>vodopa-inducedpeak-dose dyskinesias observed during the course of the <strong>di</strong>sease.Levodopa is the most widely employed and the most effectivetreatment for Parkinson’s <strong>di</strong>sease (PD). However, 30%-80% of PD patients chronically treated with <strong>le</strong>vodopa ex<strong>per</strong>iencelong-term complications [1], and it has been hypothesizedthat the emergence of <strong>le</strong>vodopa-induced involuntarymovements, such as peak-dose dyskinesias (PDD), may bere<strong>la</strong>ted to the administered cumu<strong>la</strong>tive <strong>le</strong>vodopa dose [2]. Itis well known that changes in body weight may induce substantialvariations in <strong>per</strong>ipheral pharmacokinetics of drugs[3], although this issue has not yet been investigated for <strong>le</strong>vodopa.To address whether body weight may influence thepharmacokinetic parameters of <strong>le</strong>vodopa in PD, we stu<strong>di</strong>edthe p<strong>la</strong>sma <strong>le</strong>vels of <strong>le</strong>vodopa in a <strong>la</strong>rge samp<strong>le</strong> of patientswith PD in response to an acute oral <strong>le</strong>vodopa test.One hundred sixty-four spora<strong>di</strong>c PD patients (91 menand 73 women; mean ± SD age, 65.5±8.9 years) participate<strong>di</strong>n this study. The me<strong>di</strong>an score on the mo<strong>di</strong>fied Hoehn andYahr sca<strong>le</strong> was 2.5 (range, 1 to 5), and mean duration of PDwas 83.7±59.8 months. All patients had received <strong>le</strong>vodopatherapy prior to the study and the me<strong>di</strong>an duration of treatmentwas 51.5 months (range, 3–293 months). Clinical con<strong>di</strong>tionsand <strong>per</strong>ipheral pharmacokinetics of <strong>le</strong>vodopa wereassessed following an oral acute <strong>le</strong>vodopa test. Briefly, the<strong>le</strong>vodopa test consisted of the oral administration of a standarddose of 250 mg <strong>le</strong>vodopa plus 25 mg carbidopa (imme<strong>di</strong>atere<strong>le</strong>ase formu<strong>la</strong>tion). The drug was administered at 8AM, after an overnight fast. The motor con<strong>di</strong>tions wereassessed by the Unified PD Rating Sca<strong>le</strong>, section of MotorExamination (UPDRS-ME). The occurrence of PDD followingacute <strong>le</strong>vodopa was determined as previously described[4], and PDD was recorded as present or absent by the examiningneurologist. Samp<strong>le</strong>s of venous blood for the measurementof p<strong>la</strong>smatic <strong>le</strong>vodopa concentrations were drawnthrough an indwelling catheter imme<strong>di</strong>ately before and 30,60, 120, and 240 minutes after drug intake. P<strong>la</strong>sma samp<strong>le</strong>swere stored imme<strong>di</strong>ately at -70° C and <strong>la</strong>ter assayed for <strong>le</strong>vodopausing high <strong>per</strong>formance liquid chromatography withcoulometric detection [5]. Pharmacokinetic variab<strong>le</strong>s inclu<strong>di</strong>ngpeak concentration in µmol/l (Cmax), time to reach thepeak concentration in hours (Tmax), elimination half-life inhours (T 1/2 ), and area under the curve in µmol/l h (AUC-l)were calcu<strong>la</strong>ted using Kinetica 5.0 software (Innaphase,Champs sur Marne, France). The body weight was recordedfor each patient and was measured in kilograms. Statistica<strong>la</strong>nalysis for <strong>di</strong>fferences in the examined variab<strong>le</strong>s was <strong>per</strong>formedwith Student’s t test, Mann-Whitney U test, and thechi-square test, accor<strong>di</strong>ng to the <strong>di</strong>stribution of data.Corre<strong>la</strong>tions between variab<strong>le</strong>s were stu<strong>di</strong>ed by means ofPearson and Spearman tests, and a multip<strong>le</strong> linear regressionmodel, adjusted for age and sex, was set up to investigate there<strong>la</strong>tion between body weight and AUC-l.The <strong>per</strong>ipheral pharmacokinetic variab<strong>le</strong>s of <strong>le</strong>vodopaafter an acute chal<strong>le</strong>nge with the drug in 164 patients withPD were the following: Tmax, 0.97±0.71 hours; Cmax,2.54±1.44 µmol/l; T 1/2 , 1.51±1.21 hours; AUC-l, 5.61±3.19µmol/l h (Tab<strong>le</strong> 1). The me<strong>di</strong>an body weight was 70.1 kg(range, 34–100 kg), and 65 patients (39.6%) exhibited PDD.Body weight and AUC-l were significantly and inverselycorre<strong>la</strong>ted (r=-0.287, p


S54Tab<strong>le</strong> 1 Clinical characteristics and p<strong>la</strong>smatic <strong>le</strong>vodopa area under the curve (AUC-l) in 164 patients with Parkinson’s <strong>di</strong>sease. Values aremean (SD) un<strong>le</strong>ss otherwise in<strong>di</strong>catedMen (n=91) Women (n=73) p valueAge, years 66.0 (8.9) 64.9 (9.0) NS bBody weight, kg 73.9 (9.4) 65.3 (12.2)


Neurol Sci (2002) 23:S55–S56 © Springer-Ver<strong>la</strong>g 2002Serotonin syndrome:a reported caseT.P. Avarello () • S. CottoneDepartment of Neurology, Vil<strong>la</strong> Sofia Hospital,Pa<strong>le</strong>rmo, ItalyAbstract We describe a patient treated with SSRI and L-dopa, who developed agitation, rigi<strong>di</strong>ty, hy<strong>per</strong>ref<strong>le</strong>xia, rest<strong>le</strong>ssness,autonomic instability, fever and finally death. CSFexamination, MRI of the brain, <strong>la</strong>boratory investigations,except for serum CK, glycemia and WBC, were normal. Hiscon<strong>di</strong>tion was thought to result from an central serotoninactivity. The serotonin syndrome occurs following the use ofserotomimetic agents (serotonin reuptake inhibitors, tricyclicand tetracyclic antidepressants, tryptophan alone or in combinationwith monoamine oxidase inhibitors).Serotonin syndrome mostly occurs following the use of serotomimeticagents, alone or in combination with monoamineoxidase inhibitors (MAOIs). It was first described amongdepressed patients in 1960, by Oates and Sjoerdsma, as aconsequence of the administration of high doses of tryptophanin combination with MAOIs.Signs and symptoms vary but always tend to include agitation,confusion, <strong>di</strong>sorientation, rest<strong>le</strong>ssness, coma,myoclonus, rigi<strong>di</strong>ty, hy<strong>per</strong>ref<strong>le</strong>xia, low grade fever, nausea,<strong>di</strong>arrhea, headache, tachycar<strong>di</strong>a, tachypnea, blood pressurechanges and shivering, even if, rarely, all of them present atone same time. E<strong>le</strong>ctrolyte concentrations, cerebrospinal fluidanalyses and brain imaging seem to be normal, with spora<strong>di</strong>creports of increased serum CK and mild <strong>le</strong>ukocytosis. In 1991Sternbach recognized and recommended specific <strong>di</strong>agnosticcriteria for serotonin syndrome. First, symptoms typicallydevelop soon after initiation of serotomimetic agents, or evenjust after an increase in dosage. Second, patients must reportat <strong>le</strong>ast three of the following signs and symptoms: confusion,agitation, incoor<strong>di</strong>nation, myoclonus, hy<strong>per</strong>ref<strong>le</strong>xia, tremor,<strong>di</strong>arrhea or fever. Furthermore, metabolic <strong>di</strong>sturbances, infections,and substance abuse or withdrawal must be excluded.A neuro<strong>le</strong>ptic agent should not be initiated, or its dosageincreased before any appearance of signs and symptoms.Serotonin syndrome is due to a serotonin excess in the centralnervous system (CNS) at the 5-HT 1A receptors.Dopamine and 5-HT 2 receptors may also be involved. Theserotonin syndrome is usually mild and, if managed withdrug withdrawal and supportive therapy, generally improveswithin hours; however, some patients have <strong>di</strong>ed. Causes ofthese deaths involved car<strong>di</strong>opulmonary arrest and DIC withrenal failure. We report a case of serotonin syndrome in apatient who had been given se<strong>le</strong>ctive serotonin reuptakeinhibitors (SSRI) in association with L-dopa.Case reportIn April 2001, a 72-year-old man came to our first aid stationsome days after the acute onset of limb stiffness, anxiety,trismus and dysphagia. Family history was negative for neurologicor psychiatric illness. He had been affected for yearsby <strong>di</strong>abetes mellitus and hepatitis C. In ad<strong>di</strong>tion, his re<strong>la</strong>tivesreported frequent falls, headaches and <strong>di</strong>zziness, as well asan increasing depression, the who<strong>le</strong> occurring for one year.In fact, just because of that same depression, he had beentreated for a year with SSRI and nortriptyline. During thedays prece<strong>di</strong>ng his admission, he had been administrated L-dopa, as an initial <strong>parkinson</strong>ism had also been detected.The first aid station testing revea<strong>le</strong>d unstab<strong>le</strong> blood pressurevalues, tachypnea, tachycar<strong>di</strong>a and sweating. A genera<strong>le</strong>xamination was conducted and reported normal. Furthermental testings proved poor concentration and moderate agitation.Tone was heavily increased, this concerning <strong>le</strong>gs andtrunk. C<strong>le</strong>ar evidence of limb hy<strong>per</strong>ref<strong>le</strong>xia was reported,without the contemporary joining of Babinski’s sign.Dysmetria was not present and sensation was normal. MRI,CSF and <strong>la</strong>boratory data were normal, except for glycemia,serum CK and whi<strong>le</strong> blood cell count. Saline, <strong>di</strong>azepam andantibiotic therapies were given during the hospitalization.The patient’s neurological status worsened quite rapidly and<strong>le</strong>d to death in a few days.DiscussionThe serotonin syndrome was originally described in animalspretreated with l-tryptophan and given various MAOIs orother serotonin precursors in combination with drugs thatincrease their bioavai<strong>la</strong>bility. The serotonin syndrome isthought to be induced by combined activation of 5-HT1Aand the 5-HT2 receptors. Stimu<strong>la</strong>tion of these receptors inthe dorsal and me<strong>di</strong>an raphe nuc<strong>le</strong>i of the brainstem andspinal cord may be important in the pathophysiology of thesyndrome. The dopaminergic system may be involved. It hasrecently been proposed that this syndrome may be me<strong>di</strong>atedby presynaptic inhibition of dopamine re<strong>le</strong>ase or synthesis,somewhat simi<strong>la</strong>r to neuro<strong>le</strong>ptic malignant syndrome. Onepatient on a simultaneous bromocriptine and <strong>le</strong>vodopa treatmentdeveloped a simi<strong>la</strong>r syndrome. Bromocriptine has beenpostu<strong>la</strong>ted to have some serotoninergic pro<strong>per</strong>ties.Bromocriptine, in ad<strong>di</strong>tion to being a D2-dopamine receptoragonist, activates the serotomimetic system, and l-dopainducedmyoclonus probably has a serotomimetic basis. Thepossib<strong>le</strong> partial ro<strong>le</strong>s of the 5-HT2 receptor and dopamine inthe serotonin syndrome are still unc<strong>le</strong>ar. In the present case,we remark on the extremely negative influence of SSRIs onthe dopaminergic system, probably linked to their interactionwith P450-cytochrome.


S56References1. Sandyk R (1986) L-Dopa induced “Serotonin syndrome” in a<strong>parkinson</strong>ian patient on bromocriptine. J Clin Psychopharmacol6(3):194–1952. Sporer KA (1995) The serotonin syndrome. Implicated drugs,pathophysiology and management. Drug Safety 13(2):94–1043. Bodner RA, Lynch T et al (1995) Serotonin syndrome.Neurology 45:219–2234. Beas<strong>le</strong>y CM et al (1993) Possib<strong>le</strong> monoamine oxidase inhibitorserotonin uptake inhibitor interaction: fluoxetine clinical dataand preclinical fin<strong>di</strong>ngs. J Clin Psychopharmacol 13:312–3205. Sternbach H (1991) The serotonin syndrome. Am J Psychiatry148:705–713


Neurol Sci (2002) 23:S57–S58 © Springer-Ver<strong>la</strong>g 2002Effects of the intrastriata<strong>la</strong>dministration of se<strong>le</strong>ctivedopaminergic agonists on Fosexpression in the rat brainF. B<strong>la</strong>n<strong>di</strong>ni 1 () • R. Fancellu 1,3 • F. Orzi 2G. Conti 2 • R. Greco 1 • C. Tassorelli 1 • G. Nappi 1,41 IRCCS “C. Mon<strong>di</strong>no”, Via Pa<strong>le</strong>stro 3, Pavia, Italy2IRCCS “Neuromed”, Pozzilli (IS), Italy3Università dell’Insubria, Varese, Italy4Institute of Neurological and Mental Disorders, Università <strong>di</strong>Roma “La Sapienza”, Rome, ItalyAbstract In this study, we mapped the cerebral expression ofFos protein following intrastriatal stimu<strong>la</strong>tion of D 1 or D 2receptors, in freely moving animals. Animals treated with theD 1 agonist SKF 38393 showed massive Fos increases in thecerebral cortex, ipsi<strong>la</strong>terally to the injected striatum, whichwere counteracted by systemic administration of D 1 antagonistSCH 23390. Conversely, D 2 agonist quinpiro<strong>le</strong> suppressedcortical expression of Fos, whi<strong>le</strong> systemic administrationof D 2 antagonist eticlopride relieved this blockade. Asfor the basal ganglia, Fos was consistently expressed only inthe injected striatum of rats receiving SKF 38393. Theseresults show that striatal dopamine receptors may p<strong>la</strong>y a ro<strong>le</strong>in the modu<strong>la</strong>tion of cortical activity. They also provide newinformation on a c<strong>la</strong>ss of drugs - the dopamine agonists -whose ro<strong>le</strong> in the therapeutic strategy of Parkinson’s <strong>di</strong>seaseis continuously evolving.Accor<strong>di</strong>ng to a popu<strong>la</strong>r model of basal ganglia functionalorganization, the striatum – the main input nuc<strong>le</strong>us of thecircuit – communicates with the basal ganglia output nuc<strong>le</strong>i,substantia nigra pars reticu<strong>la</strong>ta and me<strong>di</strong>al globus pallidus,through two pathways: a <strong>di</strong>rect pathway, originating fromstriatal neurons expressing D 1 dopamine receptors and anin<strong>di</strong>rect pathway, originating from striatal neurons bearingD 2 receptors [1]. In the model, the two pathways remainfunctionally and anatomically segregated. Therefore, se<strong>le</strong>ctiveactivation of either pathway would <strong>le</strong>ad to oppositechanges in the net output of the basal ganglia circuitry and,consequently, in the signaling to the cerebral cortex. Themodel is currently under partial revision: it has been suggested,for examp<strong>le</strong>, that the two dopamine receptor subc<strong>la</strong>sseswould, in fact, co-o<strong>per</strong>ate, thus limiting the actualsegregation of the striatofugal pathways [2]. Indeed, wehave recently reported that the intra-striatal administrationof se<strong>le</strong>ctive dopamine agonists reduces glucose utilizationin nuc<strong>le</strong>i of both the <strong>di</strong>rect and in<strong>di</strong>rect pathways [3].Given the increasing ro<strong>le</strong> of dopamine agonists in thetherapeutic approach to Parkinson’s <strong>di</strong>sease, we sought tofurther elucidate this issue, by mapping the expression of Fosprotein, a nonspecific marker of neuronal activation encodedby the imme<strong>di</strong>ate-early gene c-fos, in the brain of rats receivinguni<strong>la</strong>teral, intrastriatal infusions of se<strong>le</strong>ctive (D 1 or D 2 )dopamine agonists, combined with systemic administrationof the correspon<strong>di</strong>ng antagonists.Ma<strong>le</strong> Sprague-Daw<strong>le</strong>y rats (270–320 g) were anesthetizedwith thiopental so<strong>di</strong>um (50 mg/kg body weightintra<strong>per</strong>itoneally) and p<strong>la</strong>ced in a stereotaxic apparatus.Through a burr ho<strong>le</strong> in the skull, a 0.4-mm guide cannu<strong>la</strong>(0.7 mm external <strong>di</strong>ameter, Danuso, Italy) was p<strong>la</strong>ced in theright striatum (0.7 mm posterior and 2.6 mm <strong>la</strong>teral withrespect to bregma, and -4.5 mm ventral with respect to dura)and secured to the skull with screws and dental cement(Cookson, Georgia, USA). Three days <strong>la</strong>ter, animalsreceived a systemic intra<strong>per</strong>itonal injection of a D 1 (SCH23390, 0.5 mg/kg) or a D 2 (eticlopride, 1 mg/kg) dopamineantagonist or saline, followed 30 min <strong>la</strong>ter by the intrastriatalinfusion of a D 1 (SKF 38393, 30 mM) or a D 2 (quinpiro<strong>le</strong>, 20mM) agonist. Intrastriatal infusions were carried out using amicroprocessor-<strong>contro</strong>l<strong>le</strong>d syringe pump (KD Scientific,USA), which delivered a total drug volume of 0.5 µl, at a rateof 0.2 µl/min. Two hours <strong>la</strong>ter, animals were deeply anesthetizedwith so<strong>di</strong>um pentobarbital and <strong>per</strong>fused transcar<strong>di</strong>ally.Brains were quickly removed, post-fixed and sectionedon a microtome. Brain coronal sections (50 µm) wereprocessed for the detection of Fos protein, using an immunocytochemicaltechnique previously described [4].Fos expression was quantified using a computerize<strong>di</strong>mage analysis system, equipped with de<strong>di</strong>cated software(NIH Image 1.62, Scion, Frederick, MA, USA). Fos-positiveneurons were counted in all the cerebral areas of interest.Final values – representing the average rea<strong>di</strong>ng from at <strong>le</strong>astthree consecutive sections – were expressed as number ofFos-positive cells/mm 2 .The uni<strong>la</strong>teral, intrastriatal administration of SKF 38393caused massive expression of Fos throughout the ipsi<strong>la</strong>teralcerebral cortex, particu<strong>la</strong>rly in the motor, somatosensory, limbic,visual and au<strong>di</strong>tory areas; <strong>di</strong>screte Fos expression was alsodetected within the injected striatum (Fig. 1). Conversely, theintrastriatal infusion of quinpiro<strong>le</strong> <strong>di</strong>d not induce significant Fosexpression in any of the cerebral areas considered. Systemicpre-treatment with the D 1 antagonist SCH 23390 reduced theFos response to SKF 38393, consistently, in all the cortical areasconsidered, as well as in the injected striatum; the averagereduction was 41.3%. Conversely, systemic administration ofthe D 2 antagonist eticlopride induced a 5- to 50-fold increase incortical Fos expression, ipsi<strong>la</strong>terally to the quinpiro<strong>le</strong> injection.In the striatum, eticlopride pretreatment induced a 20-fol<strong>di</strong>ncrease in Fos expression, contra<strong>la</strong>terally to the injection.


S58∆R/L hemisphere (cells/mm 2 )DiscussionIn this study, we mapped the cerebral expression of Fos proteinafter se<strong>le</strong>ctive stimu<strong>la</strong>tion of dopamine receptors (D 1 orD 2 ) in the striatum of rats. Stimu<strong>la</strong>tion of striatal D 1 receptorscaused massive Fos expression throughout the entire cerebralcortex, as well as in the injected striatum. The phenomenonappeared to be receptor-me<strong>di</strong>ated, since systemic administrationof SCH 23390 reduced Fos expression. Conversely, striata<strong>la</strong>ctivation of D 2 receptors was not associated with Fosexpression; <strong>di</strong>screte cortical (ipsi<strong>la</strong>teral) and striatal (contra<strong>la</strong>teral)Fos expression was only observed when animalswere pretreated with a se<strong>le</strong>ctive D 2 antagonist (eticlopride).From a general point of view, the fact that se<strong>le</strong>ctive stimu<strong>la</strong>tionof D 1 or D 2 receptors elicited opposite responses – interms of Fos expression – is in agreement with previous observations,reporting a stimu<strong>la</strong>tory and an inhibitory ro<strong>le</strong> for D 1and D 2 receptors, respectively [5]. However, the exact mechanismunderlying this response is unc<strong>le</strong>ar. The more obviousexp<strong>la</strong>nation, based on the current model of basal ganglia functionalorganization [1], is that the D 1 -me<strong>di</strong>ated striatal stimu<strong>la</strong>tioncaused a polysynaptic activation of the basal ganglia circuitand, therefore, of the striatotha<strong>la</strong>mocortical circuit, thuseliciting the expression of Fos in cortical neurons. However,except for the injected striatum, Fos was not detected in basalganglia nuc<strong>le</strong>i. Alternatively, cortical activation may haveresulted from <strong>di</strong>rect stimu<strong>la</strong>tion of corticostriatal neurons, viathe stimu<strong>la</strong>tion of presynaptic dopamine receptors. It has beenpreviously shown that systemic administration of dopamineagonists elicits Fos expression in the cerebral cortex [6]. Morerecently, Steiner and Kitai found that such phenomenon isblocked by intrastriatal injection of the D 1 antagonist SCH23390 [7] and that the same effect is observed after striataldopamine denervation [8]. They, therefore, suggested thatactivation of striatal dopamine receptors (particu<strong>la</strong>rly of the D 1subtype) regu<strong>la</strong>tes cortex function. Our results support thisview and add another piece of information to the understan<strong>di</strong>ngof basal ganglia functional organization. These fin<strong>di</strong>ngsalso provide new information on the effects of a c<strong>la</strong>ss of drugs– the dopamine agonists – whose ro<strong>le</strong> in the therapeutic strategyof Parkinson’s <strong>di</strong>sease is continuously evolving.ReferencesFig. 1 Cortical and striatal Fosexpression after intrastriata<strong>la</strong>dministration of SKF 39393(, n = 5) or quinpiro<strong>le</strong> (, n =5), coup<strong>le</strong>d with intra<strong>per</strong>itonealsaline injection. MC, motorcortex; SSC, somatosensorycortex; LC, limbic cortex; VC,visual cortex; AC, au<strong>di</strong>tory cortex;CS, corpus striatum.Values are expressed as the <strong>di</strong>fference(∆) in the number ofFos-positive cells, betweenright (injected) and <strong>le</strong>ft hemispheres;*p


Neurol Sci (2002) 23:S59–S60 © Springer-Ver<strong>la</strong>g 2002Autosomal recessive early onset<strong>parkinson</strong>ism is linked to three loci:PARK2, PARK6, and PARK7V. Bonifati 1,2 () • M.C.J. Dekker 3N. Vanacore 1 • G. Fabbrini 1 • F. Squitieri 4R. Marconi 5 • A. Antonini 6 • P. Brustenghi 7A. Dal<strong>la</strong> Libera 8 • M. De Mari 9 • F. Stocchi 1P. Montagna 10 • V. Gal<strong>la</strong>i 7 • P. Rizzu 2 • J.C. vanSwieten 11 • B. Oostra 2 • C.M. van Duijn 3 • G. Meco 1P. Heutink 2 , and the Italian Parkinson GeneticsNetwork*1 Department of Neurological Sciences, La Sapienza University,Via<strong>le</strong> dell’Università 30, I-00185 Rome, Italy2 Department of Clinical Genetics, Erasmus University, Rotterdam,The Nether<strong>la</strong>nds, E-mail: bonifati@kgen.fgg.eur.nl3 Department of Epidemiology and Biostatistics, ErasmusUniversity, Rotterdam, The Nether<strong>la</strong>nds4 Neurogenetics Unit, IRCCS Neuromed, Pozzilli (IS), Italy5 Division of Neurology, Misericor<strong>di</strong>a Hospital, Grosseto, Italy6 Department of Neurosciences, Istituti Clinici <strong>di</strong> Perfezionamento,Mi<strong>la</strong>n, Italy7 Institute of Neurology, University of Perugia, Perugia, Italy8 Division of Neurology, Boldrini Hospital, Thiene (VI), Italy9 Institute of Neurology, University of Bari, Bari, Italy10 Institute of Neurology, University of Bologna, Bologna, Italy11 Department of Neurology, Erasmus Me<strong>di</strong>cal Centre Rotterdam,The Nether<strong>la</strong>ndsAbstract Autosomal recessive, early onset <strong>parkinson</strong>ism(AREP) is genetically heterogeneous. Mutations in theparkin gene (PARK2 locus, chromosome 6q) account for upto 50% of AREP families. The parkin protein <strong>di</strong>sp<strong>la</strong>ys ubiquitin-ligaseactivity for <strong>di</strong>fferent targets, which accumu<strong>la</strong>tein the brain of patients with parkin defect and might causeneurodegeneration. Two new AREP loci (PARK6 andPARK7) have been recently mapped on chromosome 1p andconfirmed in independent datasets, suggesting that bothmight be frequent. The three AREP forms <strong>di</strong>sp<strong>la</strong>y simi<strong>la</strong>rclinical phenotypes. Recruiting new families will helpcloning the defective genes at PARK6 and PARK7 loci. Thiswill contribute to unraveling the pathogenesis of AREP, an<strong>di</strong>t is also expected to foster our understan<strong>di</strong>ng of mo<strong>le</strong>cu<strong>la</strong>revents underlying c<strong>la</strong>ssic Parkinson’s <strong>di</strong>sease.Clinical and mo<strong>le</strong>cu<strong>la</strong>r genetic stu<strong>di</strong>es have recently <strong>le</strong>d tothe identification of <strong>di</strong>fferent monogenic forms of <strong>parkinson</strong>ism.Three chromosomal loci (PARK2, PARK6, andPARK7) have been so far identified in families with autosomalrecessive, early onset <strong>parkinson</strong>ism (AREP). ThePARK2 locus was initially mapped on the long arm of chromosome6 in Japanese families. The defective gene was <strong>la</strong>teridentified and termed parkin [1].A growing number of mutations in the parkin gene hassince been identified in several families from <strong>di</strong>fferent popu<strong>la</strong>tions[2]. To date, more than 25 Italian families withparkin <strong>di</strong>sease are known (<strong>per</strong>sonal observations and [2]).In ad<strong>di</strong>tion to point mutations, <strong>la</strong>rge genomic rearrangements(<strong>le</strong>a<strong>di</strong>ng to exon de<strong>le</strong>tions and multiplications) are frequentlydetected, in homozygous or heterozygous state, in<strong>di</strong>catingthe importance of gene dosage techniques for a sensitivescreening of parkin. In the <strong>la</strong>rgest study published todate, combining genomic sequencing and exon dosageallowed mutations in parkin gene to be detected in 49% of73 European AREP families [2].In a few families a second mutation is not found evenafter gene dosage, suggesting that other mutations (likely inintronic or regu<strong>la</strong>tory regions of the gene) still escape detection,or, some of the mutations might be sufficient to cause<strong>di</strong>sease in heterozygous state.A recent report of <strong>parkinson</strong>ism and heterozygous parkinmutation in multip<strong>le</strong> generations also suggests that parkinre<strong>la</strong>ted<strong>di</strong>sease might sometimes be dominantly inherited [3].However, pseudo-dominant inheritance could also exp<strong>la</strong>inthe <strong>di</strong>sease in multigeneration families. This phenomenonhas already been documented in three families (one Japaneseand two Italian), once again in<strong>di</strong>cating that parkin genemutations might be frequent in some popu<strong>la</strong>tions [4].The protein encoded by the parkin gene contains an N-terminal domain homologous to ubiquitin, and two RINGfinger domains separated by an IBR (in-between-RING)domain in the C-terminal part. Recent stu<strong>di</strong>es have shownthat parkin protein is an E3-c<strong>la</strong>ss ubiquitin-ligase. Differenttargets for parkin-me<strong>di</strong>ated ubiquitination have recentlybeen described: CDCrel-1, a synaptic protein; synphilin-1,an α-synuc<strong>le</strong>in-interacting protein; the PAEL-receptor, aputative transmembrane polypeptide [5]; and αSp22, a newbrain-specific glycosy<strong>la</strong>ted form of α-synuc<strong>le</strong>in [6].The ubiquitin-ligase activity of parkin is abolished bymutations found in AREP families, and the target proteinsaccumu<strong>la</strong>te in neurons of patients with parkin-<strong>di</strong>sease as aresult of <strong>la</strong>ck of ubiquitin-ligase activity. This, in turn, mightcause neurodegeneration [5, 6].The <strong>di</strong>scovery of an interaction between parkin and thebrain-specific form of α-synuc<strong>le</strong>in has important implicationfor Parkinson’s <strong>di</strong>sease in general, since this reaction mightunderlie the deposition of ubiquitinated α-synuc<strong>le</strong>in in Lewybo<strong>di</strong>es [6]. To the extent that parkin activity is essential forLewy body formation, its defect can exp<strong>la</strong>in the absence ofLewy bo<strong>di</strong>es in the brain of patients with parkin mutations.Two new loci for AREP have recently been identified andtermed PARK6 and PARK7. The two loci are close to eachother on the short arm of chromosome 1, but they are separatedby approximately 25 centiMorgans (PARK7 is moretelomeric).The PARK6 locus was initially identified by a genomewidescan in a <strong>la</strong>rge consanguineous family originating from


S60Sicily (the “Marsa<strong>la</strong>” kindred), with four affected in<strong>di</strong>vidualsin three branches [7]. A maximum two-point LOD scoreof 4.1 was obtained for marker D1S199 (θ = 0). Haplotypeanalysis identified a 12.5 cM homozygous region located at1p35-p36, f<strong>la</strong>nked by markers D1S483 (telomeric) andD1S247 (centromeric).The PARK7 locus was identified in the framework of a<strong>la</strong>rger research program named Genetic Research in Iso<strong>la</strong>tedPopu<strong>la</strong>tions (GRIP) [8]. The subject of the GRIP project is agenetically iso<strong>la</strong>ted popu<strong>la</strong>tion in the southwestern area of TheNether<strong>la</strong>nds. A <strong>la</strong>rge family with multip<strong>le</strong> consanguineousloops was identified with four in<strong>di</strong>viduals affected by AREP intwo branches. A genome-wide scan and linkage analysis usingthe program MAPMAKER-HOMOZ resulted in a maximummultipoint LOD score of 4.3 for adjacent markers on 1p36.Haplotype analysis confirmed the presence in the patients ofan homozygous region of about 16 cM f<strong>la</strong>nked by the markerD1S243 (telomeric) and D1S244 (centromeric).Since the initial linkage reports, evidence for bothPARK6 and PARK7 have been obtained in independentdatasets. Among 28 parkin-negative European families withAREP, 8 supported linkage to PARK6 [9]. In particu<strong>la</strong>r, anew Italian consanguineous family with two affected in<strong>di</strong>vidualsallowed refinement of the PARK6 centromeric border,reducing the critical region to 9 cM f<strong>la</strong>nked by markersD1S483 and D1S2674.We stu<strong>di</strong>ed four AREP families, three of which were consanguineous,and obtained support for PARK7 in three families[10]. In particu<strong>la</strong>r, in one Italian family with threeaffected in<strong>di</strong>viduals born from consanguineous parents, weobtained significant evidence for linkage confirmation (maximummultipoint LOD score 2.5, interval-wide p


Neurol Sci (2002) 23:S61–S62 © Springer-Ver<strong>la</strong>g 2002Perfusion-weighted dynamicsusceptibility (DSC) MRI: basalganglia hemodynamic changes afterapomorphine in Parkinson’s <strong>di</strong>seaseL. Brusa 1,2 • A. Bassi 1 • M. Pierantozzi 1,2 • F. Gau<strong>di</strong>ello S.Frasca 4 • R. Floris 3 • P. Stanzione 1,2 ()1IRCCS Fondazione S. Lucia, Via Ardeatina 306, I-00179 Rome, Italy2Neurology Clinic, Tor Vergata University of Rome, Rome, Italy3Institute of Ra<strong>di</strong>ology, Tor Vergata University of Rome, Rome, Italy4Department of Anesthesiology and Reanimation, Tor Vergata Universityof Rome, Rome, ItalyAbstract Re<strong>la</strong>tive regional blood flow of basal ganglia was stu<strong>di</strong>ed by meansof <strong>per</strong>fusion-weighted dynamic susceptibility (DSC) MRI. Parkinson’s <strong>di</strong>sease(PD) patients showed a significant inter-hemispheric asymmetry due to ahigher <strong>per</strong>fusion in the more affected side, whi<strong>le</strong> normal subjects <strong>di</strong>d not. PDexhibited an abnormal “asymmetry index” in the measured nuc<strong>le</strong>i. A secondDSC-MRI examination <strong>per</strong>formed after subcutaneous apomorphine administration<strong>di</strong>d not show any significant asymmetry in PD patients. DSC-MRI ofbasal ganglia confirms the asymmetry observed in PET stu<strong>di</strong>es of PD patients,suggesting that this method is a promising and low-cost technique in neurodegenerative<strong>di</strong>seases.A pattern of increased <strong>per</strong>fusion in the basal ganglia (BG), re<strong>la</strong>ted to adecreased <strong>per</strong>fusion in the cortical regions, was found by positron emissiontomography (PET) [1]. PET is a very expensive imaging method, whi<strong>le</strong>magnetic resonance imaging (MRI) is a <strong>le</strong>ss expensive imaging techniqueavai<strong>la</strong>b<strong>le</strong> almost in every neurological centre. In the <strong>la</strong>st years severalresearchers attempted to utilise dynamic susceptibility (DSC)-MRI to measureregional cerebral blood flow (rCBF) and in epi<strong>le</strong>psy [2].Our target was to study whether DSC-MRI <strong>per</strong>fusion method maydetect an altered pattern of rCBF in patients with Parkinson’s <strong>di</strong>sease (PD)in comparison to normal subjects and whether this altered pattern may benormalised by apomorphine.Fifteen subjects affected by i<strong>di</strong>opathic PD were enrol<strong>le</strong>d for this study.Twelve normal subjects were included as <strong>contro</strong>ls. Eight of them <strong>per</strong>formeda retest procedure. All included subjects gave their informed consent.PD patients, after at <strong>le</strong>ast 20 days of therapy withdrawal, were submittedto <strong>per</strong>fusion DSC-MRI. Ten of them were retested after apomorphineinjection (2–4.5 mg subcutaneously, motor improvement of at <strong>le</strong>ast 50% onUPDRS section III [6]).MRI was <strong>per</strong>formed in the dark, utilising a 1.5 T MR scanner (Philipsgyroscan ACS-NT) with gra<strong>di</strong>ent strength of 23mT/m, rise time of 0.2 mswith sinusoidal gra<strong>di</strong>ent profi<strong>le</strong> and echo-p<strong>la</strong>nar capabilities; a circu<strong>la</strong>rlypo<strong>la</strong>rized head coil with quadrature was used. T2*-weighted echo-p<strong>la</strong>narsequences were used to obtain DSC-MRI images along the anteroposteriorcommissural (AP-CP) p<strong>la</strong>ne. A dose of 0.4 mmol/kg gadolinium-DTPAwas injected to the subject lying with closed eyes. The bolus <strong>per</strong>fusion datawere processed and converted into parameter maps for re<strong>la</strong>tive rCBF.Regions of interest (ROI) of 15 pixels were manually p<strong>la</strong>ced on the headof caudate nuc<strong>le</strong>us (CN), on the putamen (PU), on the external and internalglobus pallidus (GPe/GPi) separately and on the ventro<strong>la</strong>teral nuc<strong>le</strong>us of tha<strong>la</strong>mus(TH). CU, PU and GPe were localised on the slice p<strong>la</strong>ced 3 mm abovethe AC-PC line, whi<strong>le</strong> TH and GPi were localised on the slice p<strong>la</strong>ced 3 mmbelow the AP-CP line [3]. Moreover, <strong>per</strong>fusion was evaluated in a white parieto-occipitalmatter (WPOM), to <strong>per</strong>form normalisation of the data.Row flow data, calcu<strong>la</strong>ted as the mean of each ROI in the BG nuc<strong>le</strong>iand in the WPOM of each side, were logarithmically transformed, and thennormalised as <strong>per</strong>centage of the value obtained from the ipsi<strong>la</strong>teral WPOM.For asymmetry determination (“contrast” effect), normalised data wereexpressed as a ratio of the contra<strong>la</strong>teral correspon<strong>di</strong>ng nuc<strong>le</strong>us, accor<strong>di</strong>ngto the formu<strong>la</strong>: [(right nuc<strong>le</strong>us - <strong>le</strong>ft nuc<strong>le</strong>us)/(right nuc<strong>le</strong>us + <strong>le</strong>ft nuc<strong>le</strong>us)*100].In PD patients, the contrast was expressed as [(best side - worstside)/(best side + worst side)*100] accor<strong>di</strong>ng to their clinical asymmetry.An in<strong>di</strong>vidual “contrast index” was considered abnormal when excee<strong>di</strong>ngthe mean ± 2 SD of normal subjects.Data analysis was <strong>per</strong>formed with STATISTICA for Windows program.Normalised data were analysed with parametric ANOVAs utilisingthe following main factors: “group” (between factor: PD vs. <strong>contro</strong>l subjects);“treatment” (within factor: before-after drug administration or testretest);“nuc<strong>le</strong>i” (within factor: CU, PU, TH GPe and GPi); “side” (withinfactor: right side or best side vs. <strong>le</strong>ft side or worst side).The who<strong>le</strong> group of PD patients exhibited a significantly (F(1/25)=7.98; p


S62DiscussionOur data show that rCBF in the BG of untreated PD patients is stronglyasymmetric between the WS and the BS, and that this asymmetry is normalisedby apomorphine. No asymmetry was present in <strong>contro</strong>ls in the testretestprocedure. Our study confirm previous PET fin<strong>di</strong>ngs of a re<strong>la</strong>tivelyincreased and asymmetric rCBF in the BG in uni<strong>la</strong>teral or bi<strong>la</strong>teral PDpatients [1]. Interestingly, PET stu<strong>di</strong>es <strong>di</strong>d not show the asymmetrybetween homologous BG regions but only if the rCBF of BG nuc<strong>le</strong>i werecompared to those of several cortical areas. On the contrary, the <strong>le</strong>ss expensiveDSC-MRI technique seems to be ab<strong>le</strong> to reveal the asymmetry byexamining homologous subregions of BG. This was allowed by the higherresolution of MRI in comparison to PET.Apomorphine treatment was ab<strong>le</strong> to normalise the asymmetry of rCBF,probably re<strong>la</strong>ted to a dopamine dep<strong>le</strong>tion-dependent mechanism. The postapomorphinerecovery was due to an increased rCBF in the BS, whi<strong>le</strong> theWS <strong>di</strong>d not change. Dopamine receptor stimu<strong>la</strong>tion in the BG may se<strong>le</strong>c-tively change the rCBF in these regions, thus accounting for this fin<strong>di</strong>ng.However apomorphine also produces re<strong>le</strong>vant vaso<strong>di</strong><strong>la</strong>tation, possiblyaccounting for part of the increase in rCBF observed in the BS. The <strong>la</strong>rgerloss of dopamine in the WS might have produced a maximal increase ofrCBF already in basal con<strong>di</strong>tions. Thus, apomorphine may not be ab<strong>le</strong> toinduce further increase of rCBF in that side.References1. Eidelberg D, Moel<strong>le</strong>r JR, Dhawan V et al (1994) The metabolictopography of Parkinsonism. J Cereb Blood Flow Metab14(5):783–8012. Liu HL, Kochunov P, Hou J et al (2001) Perfusion weighte<strong>di</strong>maging of interictal hypo<strong>per</strong>fusion in temporal lobe epi<strong>le</strong>psyusing FAIR-HASTE: comparison with H(2)(15)O PET measurements.Magn Reson Med 45(3):431–4363. Ta<strong>la</strong>irach J, Tournoux P (1988) Co-p<strong>la</strong>nar stereotaxic at<strong>la</strong>s of thehuman brain. George Thieme, Stuttgart New YorkTab<strong>le</strong> 1 Basal ganglia mean rCBF and mean rCBF “contrast”Basal ganglia mean rCBFBasal ganglia mean rCBF “contrast”CU mean rCBF ± SDCU mean rCBF “contrast” ± SDPRE apomorphine/testPOST apomorphine/retestR/B L/W R/B L/W PRE apo/test POST apo/retestPD n=15 111.6±7.32 121.2±1258 – – -3.45±5.82 –Control n=12 115.1±6.53 116.2±6.12 – – -0.79±241 –PD n=10 112.2±7.35 118.0±5.2 126.3±11.8 126.6±11.0 -2.97±3.75 -0.13±4.27Control n=8 117.3±7.1 117.3±6.9 117.5±764 119.5±6.44 -0.34±2.554 -0.46±2.60PU mean rCBF ± SDPU mean rCBF “contrast” ± SDPRE apomorphine/testPOST apomorphine/retestR/B L/W R/B L/W PRE apo/test POST apo/retestPD n=15 112.1±6.4 122.5 ± 13.4 – – -4.23±5.74 –Control n=12 117.2±6.1 117.2 ± 7.62 – – -0.04±2.21 –PD n=10 112.1±.25 121.2±6.2 126.6±11.1 128.2±11.4 -3.76±4.16 -0.52±3.93Control n=8 117.3±6.08 118.7±5.1 119.1±6.11 119.7±6.26 -0.31±1.92 -0 44±1.31TH mean rCBF ± SDTH mean rCBF “contrast” ± SDPRE apomorphine/testPOST apomorphine/retestR/B L/W R/B L/W PRE apo/test POST apo/retestPD n=15 111.4 ± 7.27 122.8 ± 13.0 – – -4.32 ± 5.51 –Control n=12 115.2 ± 7.12 115.3 ± 7.4 – – 0.22 ± 2.06 –PD n=10 112.7 ± 7.73 120.0 ± 5.79 126.6 ± 12.1 128.1 ± 12.3 -3.22 ± 3.03 -0.51 ± 4.25Control n=8 116.5 ± 6.88 116.1 ± 6.33 119.4 ± 6.23 118.1 ± 9.26 -0.023 ± 2.08 0.67 ± 4.33GPe mean rCBF ± SDGPe mean rCBF “contrast” ± SDPRE apomorphine/testPOST apomorphine/retestR/B L/W R/B L/W PRE apo/test POST apo/retestPD n=15 101.2 ± 4.1 112.1 ± 12.2 – – -4. –Control n=12 106.1 ± 4.6 106.2 ± 7.6 – – -0.04 ± 3.14 –PD n=10 101.6 ± 4.41 110.8 ± 5.74 114.6 ± 12.3 114.2 ± 10.4 -4.23 ± 4.44 0.066 ± 3.99Control n=8 105.1 ± 4.04 107.2 ± 4.73 105.4 ± 3.20 106.1 ± 7.83 -0.94 ± 2.10 -0.16 ± 3.43GPi mean rCBF ± SDGPi mean rCBF “contrast” ± SDPRE apomorphine/testPOST apomorphine/retestR/B L/W R/B L/W PRE apo/test POST apo/retestPD n=15 99.15 ± 12.5 109.5 ± 4.93 – – -4.72 ± 6.62 –Control n=12 100.3 ± 7.4 101.1 ± 5.53 – – -0.14 ± 3.53 –PD n=10 98.6 ± 5.16 107.6 ± 7.11 113.1 ± 13.32 114.5 ± 11.6 -4.53 ± 5.32 -0.78 ± 4.64Control n=8 99.67 ± 4.71 102.1 ± 5.11 100.1 ± 2.79 102.2 ± 9.03 -1.44 ± 2.56 -0.72 ± 4.82RCBF, regional cerebral blood flow; CU, caudate nuc<strong>le</strong>us; TH, ventro <strong>la</strong>teral tha<strong>la</strong>mus; PU, putamen; Gpe, external globus pallidus; Gpi, internal globuspallidus; R, right; L, <strong>le</strong>ft; B, best; W, worst. “contrast”, [(right nuc<strong>le</strong>us- <strong>le</strong>ft nuc<strong>le</strong>us)/(right nuc<strong>le</strong>us + <strong>le</strong>ft nuc<strong>le</strong>us)*100] in normal subjects; “contrast”,[(best side - worst side)/(best side + worst side)*100] in PD (see methods)


Neurol Sci (2002) 23:S63–S64 © Springer-Ver<strong>la</strong>g 2002Frequency, <strong>di</strong>stribution andcharacteristics of progressivesupranuc<strong>le</strong>ar palsy in Italy:preliminary observationsA. Brusa 1 • S. Cammarata 21 Corso Aurelio Saffi 15/4, Genoa, Italy2 Division of Neurology, Galliera Hospital, Genoa, ItalyAbstract We report the number of pathologically provencases of progressive supranuc<strong>le</strong>ar palsy, described in theItalian neurological literature from 1961 until now. A <strong>di</strong>scussionof the <strong>di</strong>agnostic value of downward gaze palsy is made.A comparison with the number of simi<strong>la</strong>r cases described inthe rest of the world and with the number of Parkinsonianpatients who <strong>di</strong>ed in the same region in the correspon<strong>di</strong>ngyear is attempted.Even after nearly 40 years since the <strong>di</strong>scovery of progressivesupranuc<strong>le</strong>ar palsy (PSP) [1–3], the precocious <strong>di</strong>agnosis ofthis <strong>di</strong>sease is <strong>di</strong>fficult. Recently, there is a tendency to giveimportance to the presence of downward supranuc<strong>le</strong>ar gazepalsy [4]. We have already demonstrated, from an examinationof the literature, that this was not the case [5, 6]. Fromthe frequency and the age at onset of downward gaze palsyamong Italian cases, it appears that this symptom is neitherfrequent nor precocious (Tab<strong>le</strong> 1). The number of PSP casesin Italy is small, both in comparison to the number ofpatients described in the rest of the world and to the numberof patients who <strong>di</strong>ed from Parkinson’s <strong>di</strong>sease in the sameregion and in the correspon<strong>di</strong>ng years. The small number ofPSP cases is not due only to the <strong>di</strong>fficulty in obtaining autopsy,because until 1980 also the number of clinical casesdescribed in Italy was <strong>le</strong>ss than the correspon<strong>di</strong>ng number ofcases reported in the rest of the world.We also considered the number of <strong>parkinson</strong>ian patientswho <strong>di</strong>ed in the same region, in the correspon<strong>di</strong>ng year(ISTAT data, avai<strong>la</strong>b<strong>le</strong> only from 1968 to 1997). TheISTAT data also include deaths due to PSP, although fromthis year on, deaths due to PSP will be c<strong>la</strong>ssified separatelyunder the code RF170. Also in this respect no corre<strong>la</strong>tionis useful; <strong>per</strong>haps it will be possib<strong>le</strong> in the field of the clinicalcases, whose number is certainly higher. The <strong>di</strong>fficultyin obtaining <strong>per</strong>mission of autopsy resulted in an underestimationof the frequency of the <strong>di</strong>sease.Tab<strong>le</strong> 1 Pathologically proven cases of PSP reported in the literature for Italy and the rest of the world, in comparsion to <strong>di</strong>rection ofsupranuc<strong>le</strong>ar ocu<strong>la</strong>r palsy at onset and the number of deaths due to <strong>parkinson</strong>ism for the correspon<strong>di</strong>ng regionPSP cases in PSP cases in Italy a Deaths from <strong>parkinson</strong>ismYear rest of world, n n Region Ocu<strong>la</strong>r palsy Same Italian region, n b1961 0 1 Liguria NR 01967 3 1 Liguria Down 01969 7 1 Veneto + Up 81Trentino-Alto A<strong>di</strong>ge1978 3 1 Umbria Up + down 221979 5 1 Liguria Up + down 1071981 5 1 Piedmont Up + down 212– 1 Emilia-Romagna Up 154– 3 Lazio Up 1761983 17 1 Emilia-Romagna Up 1921984 18 6 Emilia-Romagna Up + down 167– 3 Liguria Up, 1; Down, 2 951987 58 1 Liguria NR 1581988 169 1 Lombardy Up + down 3891990 34 2 Liguria NR 1522000 0 3 Veneto + Up + down NATrentino Alto A<strong>di</strong>geNR, not reported; NA, not avai<strong>la</strong>b<strong>le</strong>; a Cases are reported in [7]; b Data from ISTAT


S64References1. Stee<strong>le</strong> JC, Richardson JC, Olszewski J (1964) Progressivesupranuc<strong>le</strong>ar palsy. A heterogeneous degeneration involvingthe brain stem, basal ganglia and cerebellum with verticalgaze and pseudobulbar palsy, nuchal dystonia and dementia.Arch Neurol 10:333–3592. Lees AJ (1987) The Stee<strong>le</strong>-Richardson-Olszewski syndrome(progressive supranuc<strong>le</strong>ar palsy). Mov Disord 2:272–2873. Golbe LI, Davis PH, Schoenberg, BS, Duvoisin RC (1988)Preva<strong>le</strong>nce and natural history of progressive supranuc<strong>le</strong>arpalsy. Neurology 38:1031–10344. Litvan I, Agid Y, Jancovic J, Goetz C, Brandel JP, Lai EC,Wenning G, D’Olhaberriague L, Verny M, Chaudhuri KR,McKee A, Jellinger K, Bartko JJ, Mangone CA, Pearce RKB,(1996) Accuracy of clinical criteria for the <strong>di</strong>agnosis of progressivesupranuc<strong>le</strong>ar palsy (Stee<strong>le</strong>-Richardson-Olszewskisyndrome). Neurology 46:922–9305. Brusa A (1961) Dégénérescence plurisystematisée dunévraxe, de caractère spora<strong>di</strong>que, à debut tar<strong>di</strong>f et evolutionsubaigue. Etude anatomo-clinique d'un cas <strong>di</strong>ffici<strong>le</strong> à c<strong>la</strong>sser.Rev Neurol (Paris) 104:412–4296. Brusa A, Mancar<strong>di</strong> GL, Bugiani O (1980) Progressivesupranuc<strong>le</strong>ar palsy 1979: an overview. Ital J Neurol Sci1:205–2227. Brusa A, Peloso PF (1993) An introduction to progressivesupranuc<strong>le</strong>ar palsy. John Libbey CIC, Rome


Neurol Sci (2002) 23:S65–S66 © Springer-Ver<strong>la</strong>g 2002Frontal intermittent rhythmicdelta activity (FIRDA) in patientswith dementia with Lewy bo<strong>di</strong>es:a <strong>di</strong>agnostic tool?S. Calzetti () • E. Bortone • A. NegrottiL. Zinno • D. ManciaInstitute of Neurology, University of Parma, Parma, ItalyAbstract The accuracy of the clinical <strong>di</strong>agnosis of dementiawith Lewy bo<strong>di</strong>es (DLB) remains poor, especially in earlyphases of the <strong>di</strong>sease, in spite of applying current consensus<strong>di</strong>agnostic criteria. The need for supportive <strong>di</strong>agnostic tools istherefore warranted. In this study EEG recor<strong>di</strong>ngs showed amain pattern of bi<strong>la</strong>teral frontal intermittent rhythmic deltaactivity (FIRDA) in 7 of 10 patients, aged 58–83 years, 8 ofwhom were <strong>di</strong>agnosed as affected by “probab<strong>le</strong>” and 2 by“possib<strong>le</strong>” DLB. Conversely, the same EEG abnormality wasfound only in 2 of 9 age-matched patients, 8 of whom had“probab<strong>le</strong>” and 1 “possib<strong>le</strong>” Alzheimer’s <strong>di</strong>sease, accor<strong>di</strong>ng toNINCDS-ADRDA criteria, taken as <strong>contro</strong>ls. The degree ofcognitive impairment was comparab<strong>le</strong> among the two groupsof patients. If these fin<strong>di</strong>ngs will be confirmed in a <strong>la</strong>rgerseries, FIRDA, even though an aspecific EEG pattern, couldbe of value in improving the <strong>di</strong>agnostic accuracy of DLB.women, aged 58-85 years (me<strong>di</strong>an 73 years): 8 patients had a<strong>di</strong>agnosis of “probab<strong>le</strong>” and 1 had a <strong>di</strong>agnosis of “possib<strong>le</strong>”AD, accor<strong>di</strong>ng to NINCDS-ADRDA criteria [2]. In thesepatients the me<strong>di</strong>an duration of the <strong>di</strong>sease was 2 years (range,2–11 years), and the degree of cognitive impairment was moderatein 2 patients and severe in the remaining 7.EEG recor<strong>di</strong>ngs were <strong>per</strong>formed with a 21-channel Nihon-Kohden apparatus. The e<strong>le</strong>ctrodes were positioned accor<strong>di</strong>ngto the International 10–20 system with bipo<strong>la</strong>r montages.Slowing of the background EEG activity in the frequencyband between 6 and 8 Hz, associated with poor reactivityto eyes closing, was found in all patients with DLB and in 6of 9 patients with AD. Seven of 10 patients with DLBshowed a main pattern of bi<strong>la</strong>teral frontal intermittent rhythmicdelta activity (FIRDA), whereas bi<strong>la</strong>teral theta slowwaves in the temporal regions were found in the remaining 3patients. In contrast, FIRDA pattern occurred in only 2 of 9patients with AD, whereas in the other 7 patients bi<strong>la</strong>teraltheta slow waves in the temporal regions were found.An examp<strong>le</strong> of FIRDA in a patient with DLB is shown inFig. 1.Renewed interest has been recently raised on the ro<strong>le</strong> of e<strong>le</strong>ctroencephalography(EEG) in the <strong>di</strong>agnosis of patients withdegenerative dementias. However, whereas aspecific EEGabnormalities corre<strong>la</strong>ting with the degree of cognitive impairmenthave long been recognized in patients with Alzheimer’s<strong>di</strong>sease (AD), in patients with dementia with Lewy bo<strong>di</strong>es(DLB) a wide spectrum of EEG changes has been reported,even though to date the <strong>la</strong>tter <strong>di</strong>sorder remains poorly investigatedwith this technique. Indeed, the search for a neurophysiologicaldysfunction which proves useful in supporting theclinical <strong>di</strong>agnosis of DLB is strongly warranted, due to therather low <strong>le</strong>vel of accuracy of the currently avai<strong>la</strong>b<strong>le</strong> <strong>di</strong>agnosticcriteria [1], especially in the early phases of the <strong>di</strong>sease.We report the results of a study that investigated the patternsof EEG abnormalities in patients <strong>di</strong>agnosed with “probab<strong>le</strong>”or “possib<strong>le</strong>” DLB and AD.The former group included 10 patients, 7 men and 3women, aged 58–83 years (me<strong>di</strong>an 69.5 years); in 8 of thesepatients a <strong>di</strong>agnosis of “probab<strong>le</strong>” and in the other 2, a <strong>di</strong>agnosisof “possib<strong>le</strong>” DLB was made, accor<strong>di</strong>ng to the previouslymentioned criteria. Duration of the <strong>di</strong>sease ranged between 2and 11 years (me<strong>di</strong>an 3 years), and the degree of cognitiveimpairment was mild in 1, moderate in 3 and severe in 6 of thepatients. The second group comprised 9 patients, 4 men and 550µV|—1 sFig. 1 EEG recor<strong>di</strong>ng showing FIRDA pattern in a patient with DLB.The time constant was 100 msec and the signals were amplified to50µV and filtered up to 35 Hz


S66DiscussionPrevious anedoctical reports have shown a wide spectrum ofEEG changes in DLB, inclu<strong>di</strong>ng aspecific [3] and more definiteabnormal patterns, i.e. FIRDA [3, 4], triphasic waves [5] and<strong>per</strong>io<strong>di</strong>c synchronous <strong>di</strong>scharges [6] or <strong>per</strong>io<strong>di</strong>c or pseudo<strong>per</strong>io<strong>di</strong>csharp wave comp<strong>le</strong>xes [7, 8] resembling those typical ofCreutzfeldt-Jacob’s <strong>di</strong>sease. These fin<strong>di</strong>ngs likely ref<strong>le</strong>ct a clinicopathologicalheterogeneity in the reported case series.The first formal EEG study showed in half of 14 patientswith autoptically verified DLB a pattern of temporal lobe slowwave transients, which were found to be corre<strong>la</strong>ted with a clinicalhistory of loss of consciousness [9]. In a more recent investigation[10], “bursts of bisynchronous, <strong>di</strong>ffuse rhythmic 2- to 3-Hz waves” were reported in an unspecified proportion of 18patients with DLB, whereas in other 6 patients “<strong>per</strong>sistent <strong>di</strong>ffusedelta, bisynchronous spikes or sharp waves and triphasicwaves” were observed. Therefore, to date it remains unc<strong>le</strong>arwhether or not a <strong>di</strong>stinctive abnormal EEG pattern may be recognize<strong>di</strong>n DLB.If our fin<strong>di</strong>ngs will be confirmed in a <strong>la</strong>rger series, the occurrenceof FIRDA in patients with degenerative dementia couldbe regarded as an in<strong>di</strong>cator supporting the <strong>di</strong>agnosis of DLB.References1. McKeith IG, Ga<strong>la</strong>sko D, Kosaka K et al (1996) Consensusguidelines for the clinical and pathologic <strong>di</strong>agnosis ofdementia with Lewy bo<strong>di</strong>es (DLB): report of theConsortium on DLB international workshop. Neurology47:1113–11242. McKhann G, Drachman DA, Folstein M et al (1984) Clinical<strong>di</strong>agnosis of Alzheimer’s <strong>di</strong>sease: report of the NINCDS-ADRDA Work Group under the auspices of the Department ofHealth and Human Services Task Force on AlzheimerDisease. Neurology 34:939–9443. Crystal HA, Dickson DW, Lizar<strong>di</strong> JE et al (1990) Antemortem<strong>di</strong>agnosis of <strong>di</strong>ffuse Lewy body <strong>di</strong>sease. Neurology40:1523–15284. Burkhardt CR, Fil<strong>le</strong>y CM, K<strong>le</strong>inschmidt-DeMasters BK et al(1988) Diffuse Lewy body <strong>di</strong>sease and progressive dementia.Neurology 38:1520–15285. Byrne EJ, Lennox G, Lowe J et al (1989) Diffuse Lewy body<strong>di</strong>sease: clinical features in 15 cases. J Neurol NeurosurgPsychiatry 52:709–7176. Yamamoto T, Imai T (1988) A case of <strong>di</strong>ffuse Lewy body andAlzheimer’s <strong>di</strong>seases with <strong>per</strong>io<strong>di</strong>c synchronous <strong>di</strong>scharges. JNeuropathol Exp Neurol 47:536–5487. Haïk S, Brandel JP, Sazdovitch V et al (2000) Dementiawith Lewy bo<strong>di</strong>es in a neuropathologic series of suspectedCreutzfeldt-Jakob <strong>di</strong>sease. Neurology 55:1401–14048. Tschampa HJ, Neumann M, Zerr I et al (2001) Patients withAlzheimer’s <strong>di</strong>sease and dementia with Lewy bo<strong>di</strong>es mistakenfor Creutzfeldt-Jakob <strong>di</strong>sease. J Neurol NeurosurgPsychiatry 71:33–399. Briel RCG, McKeith IG, Barker WA et al (1999) EEG fin<strong>di</strong>ngsin dementia with Lewy bo<strong>di</strong>es and Alzheimer’s <strong>di</strong>sease.J Neurol Neurosurg Psychiatry 66:401–40310. Barber PA, Varma AR, Lloyd JJ et al (2000) The e<strong>le</strong>ctroencephalogramin dementia with Lewy bo<strong>di</strong>es. Acta NeurolScand 101:53–56


Neurol Sci (2002) 23:S67–S68 © Springer-Ver<strong>la</strong>g 2002Bipo<strong>la</strong>r affective <strong>di</strong>sorder andParkinson’s <strong>di</strong>sease: a rare, insi<strong>di</strong>ousand often unrecognized associationA. Cannas 1 () • A. Spissu 2 • G.L. Floris 1S. Congia 1 • M.V. Sad<strong>di</strong> 1 • M. Melis 1 • M.M. Mascia 1F. Pinna 1 A. Tuveri 1 • P. Sol<strong>la</strong> 1 • A. Milia 1M. Giagheddu 1 • P. Tacconi 11 Institute of Neurology, University of Cagliari, Via Ospeda<strong>le</strong> 46, I-09127, Cagliari, Italy2 Neurological Department, S. Miche<strong>le</strong> Hospital, Cagliari, ItalyAbstract Five patients (4 women) with Parkinson’s <strong>di</strong>sease(PD) and primary major psychiatric <strong>di</strong>sorder (PMPD) meetingDSM-IV criteria for the <strong>di</strong>agnosis of bipo<strong>la</strong>r affective<strong>di</strong>sorder (BAD) were stu<strong>di</strong>ed. Four patients had early onsetPD. Four developed a severe psychiatric <strong>di</strong>sorder a few yearsafter starting dopaminergic therapy in presence of a mildmotor <strong>di</strong>sability and a mild cognitive impairment, with noevidence of cerebral atrophy at CT or MRI. Two patientsdeveloped a c<strong>le</strong>ar manic episode; the other three presented asevere depressive episode (in one case featuring a Cotardsyndrome). None showed previous signs of long term L-dopatreatment syndrome (LTS), hallucinosis or other minor psychiatric<strong>di</strong>sorders. The two manic episodes occurred shortlyafter an increase of dopaminergic therapy and in one caserapid cyclic mood fluctuations were observed. At the onsetof psychiatric symptoms, all patients had an unspecific <strong>di</strong>agnosisof chronic delusional hallucinatory psychosis (CDHP).Recent stu<strong>di</strong>es on the occurrence of major psychiatric <strong>di</strong>sorders(MPD), e.g. delusion with or without hallucinations,major mood <strong>di</strong>sorders, in subjects with Parkinson’s <strong>di</strong>sease(PD) provide interesting cues for <strong>di</strong>scussion. The developmentof a psychiatric illness early in the course of PD anddopaminergic therapy, in re<strong>la</strong>tively young patients withoutdementia or delirium, is of particu<strong>la</strong>r interest [1, 2]. These<strong>di</strong>sorders, which usually occur in an insi<strong>di</strong>ous fashion with anonspecific picture of chronic delusional hallucinatory psychosis(CDHP) in <strong>parkinson</strong>ian patients treated withdopaminergic drugs, sometimes hide a coexisting primarymajor psychiatric <strong>di</strong>sorder (PMPD) and often represent aserious <strong>di</strong>agnostic and therapeutic prob<strong>le</strong>m [3]. Few stu<strong>di</strong>eshave investigated the association between PD and bipo<strong>la</strong>raffective <strong>di</strong>sorders (BAD) [4, 5], although some reports evidentiatedc<strong>le</strong>ar re<strong>la</strong>tionships among mood fluctuations(depression and mania), motor status and brain dopamine<strong>le</strong>vels. In fact, regression of <strong>parkinson</strong>ian akinesia during amanic episode [6], occurrence of a manic episode afteradministration of apomorphine in <strong>parkinson</strong>ian patients withdepression and, vice versa, the development of a <strong>parkinson</strong>iansyndrome in patients with BAD have been reported [7].All patients with i<strong>di</strong>opathic PD who had been referred toour Parkinson Centre over an 8-year <strong>per</strong>iod and who had developeda psychotic picture with insi<strong>di</strong>ous onset and clinicalsymptoms consistent with CDHP were taken into considerationand 5 particu<strong>la</strong>r cases were identified. On careful evaluation ofthe clinical history and psychiatric manifestations of thesepatients, a rarely described association of PD and BAD wasrevea<strong>le</strong>d. BAD was <strong>di</strong>agnosed retrospectively by examining al<strong>la</strong>vai<strong>la</strong>b<strong>le</strong> clinical data, the typical history and DSM-IV criteria.Each patient was examined for sex, age-at-onset and clinicalvariant of PD, <strong>di</strong>sease duration, years of dopaminergictherapy, Hoehn/Yahr (H/Y) stage, cognitive status, GlobalDeterioration Sca<strong>le</strong> (GDS) and Mini Mental StateExamination (MMSE), brain imaging (CT or MRI), previoussign of LTS, age at psychosis onset and treatment, previoushistory of minor psychiatric <strong>di</strong>sturbances (e.g. vivid dreams,hy<strong>per</strong>sexuality, hallucinosis) and interval of onset of psychiatricsymptoms.Patients were four women and one man; three had c<strong>la</strong>ssicvariant of PD and two had an akinetic-rigid syndrome.Four had early onset PD. The mean age at <strong>di</strong>sease onset was47.2 years (range, 45-49).BAD was <strong>di</strong>agnosed in four patients several years after<strong>di</strong>agnosis of PD; only one fema<strong>le</strong> patient had been previouslyaware of BAD. After a few years of dopaminergic therapy(range, 3–5 years), the four patients suddenly developed aclinical picture of CDHP with progressive delusional manifestations,mainly of <strong>per</strong>secutory type in depressive phase, and ofmegalomanic and/or erotomanic type during manic episodes.In florid <strong>per</strong>iods these symptoms were associated with au<strong>di</strong>toryand visual hallucinations linked to mood fluctuations. BADwas thus <strong>di</strong>agnosed in 4 of 5 patients some years after <strong>di</strong>agnosisof PD and initiation of dopaminergic treatment. In thesefour patients, the mean age at <strong>di</strong>agnosis of PD was 48.7 yearsand the mean age at <strong>di</strong>agnosis of BAD was 52.2 years. Theinterval between starting dopaminergic therapy and onset ofthe psychotic <strong>di</strong>sorder consistent with BAD was 3.5 years.The study of risk factors for development of a psychiatric<strong>di</strong>sorder strictly linked to dopaminergic treatment or PD evidentiatedlow motor <strong>di</strong>sability (H/Y stage ranged from 2 to3), non-significant cognitive impairment (mean GDS score,2.4; range, 2-4; mean MMSE, 26.4) and absence of brainatrophy in our patients. These fin<strong>di</strong>ngs were not consistentwith an organic or pharmacotoxic psychiatric <strong>di</strong>sorder.During observation, two patients ex<strong>per</strong>ienced a manicepisode, two had major depression and one had a markedmood depression featuring a Cotard syndrome. Four patientswere affected by BAD with long intervals between episodes,whi<strong>le</strong> one patient showed a rapid-cycling BAD. Our patientsmanifested some interesting characteristics consistent withprevious anecdotal reports: the comp<strong>le</strong>te remission of clinicalsymptoms and signs of PD in manic episodes (twopatients) that <strong>le</strong>d to a rapid and important reduction ofdopaminergic therapy and the development of mania with


S68subacute onset after increase of <strong>per</strong>golide (two patients) or<strong>le</strong>vodopa (one patient).DiscussionTo date, the association between PD and PMPD has been poorlydefined and is still scarcely considered. The need to proceedbeyond anecdotal reports is supported by the important influenceexerted by the correct <strong>di</strong>agnosis of these <strong>di</strong>sorders, in particu<strong>la</strong>ron the e<strong>la</strong>boration of therapeutical strategies. Theassumption that DSM-IV criteria imply both the exclusion oforganic brain illness and use of psychoactive drugs in <strong>di</strong>agnosingPMPD should not <strong>di</strong>scourage from investigating that association,because it is equiva<strong>le</strong>nt to asserting that PD could neverbe manifested in association with PMPD, and this is unreasonab<strong>le</strong>and undemonstrated.The association between BAD and PD has been reportedonly rarely [4, 5]. In our opinion, this is due both to the rarityof the association and to the fact that when a major psychiatric<strong>di</strong>sorder occurs during dopaminergic treatment forPD, it may be incorrectly <strong>la</strong>bel<strong>le</strong>d as drug-induced CDHP;vice versa, when BAD is pre-existent to PD the <strong>la</strong>tter mayeasily be <strong>la</strong>bel<strong>le</strong>d as iatrogenic <strong>parkinson</strong>ism. On the otherhand, the re<strong>la</strong>tionship between mood fluctuation, depressionor mania, and brain dopamine seems to emerge c<strong>le</strong>arly in literature[8, 9]. In particu<strong>la</strong>r, it has been previously observedthat <strong>le</strong>vodopa is ab<strong>le</strong> to induce hypomania or mania inpatients with BAD or PD [10].In our ex<strong>per</strong>ience PMPD in PD tends to occur early duringdopaminergic therapy and is frequently characterized byan unspecified and dramatic picture of CDHP. Early onset ofCDHP in early onset PD inevitably implies two alternative<strong>di</strong>agnoses: “a comorbid psychotic illness (often unrevea<strong>le</strong>dby the patient initially) or an evolving <strong>parkinson</strong>ism-plussyndrome” [2].Our fin<strong>di</strong>ngs demonstrate that the absence of atrophy inneuroimaging and or cognitive impairment in psychometricevaluation provides evidence of the <strong>la</strong>ck of psychiatric organicor drug-induced <strong>di</strong>sorders. Simi<strong>la</strong>rly, the absence of minor psychiatric<strong>di</strong>sorders such as hallucinosis, hy<strong>per</strong>sexuality, vividdreams and confusional episodes (drug-induced phenomena inthe advanced PD), as well as motor complications in LTS supportthis fin<strong>di</strong>ng.Meticulous investigation of family and <strong>per</strong>sonal history isessential for a correct nosological c<strong>la</strong>ssification of CDHP, withrespect for DSM-IV criteria, particu<strong>la</strong>rly when this <strong>di</strong>sorder ismanifested only a few years after the start of dopaminergictherapy in recently <strong>di</strong>agnosed PD patients without cerebra<strong>la</strong>trophy or cognitive impairment. Indeed, in four of our patientsthe interval between the start of dopaminergic therapy andonset of psychiatric <strong>di</strong>sorders ascribed to BAD was <strong>le</strong>ss thanfour years. In the fifth patient BAD symptoms <strong>la</strong>rgely precededthe onset of PD.The observation of a cyclic and bipo<strong>la</strong>r course of a seriousmood <strong>di</strong>sorder in the presence of a typical family history facilitates<strong>di</strong>agnosis of undetected BAD in PD patients. The onsetof PD in a patient suffering from BAD, and vice versa, seriouslycomplicates anti<strong>parkinson</strong> therapy because of the influencethat dopaminergic drugs exert over mood. The treatmentof manic phases and severe depression in patients also affectedby PD presents opposite prob<strong>le</strong>ms with regard to dopaminergictherapy and common prob<strong>le</strong>ms concerning the need to usemood-stabilizing drugs such as carbolithium, carbamazepineand valproate.In our patients, the visual and/or au<strong>di</strong>tory hallucinations inthe manic phase or in Cotard syndrome were treated with atypicalneuro<strong>le</strong>ptics. The Cotard syndrome was successfully treatedwith intravenous clomipramine (50 mg/day for 20 days). Inad<strong>di</strong>tion, the remarkab<strong>le</strong> resolution of <strong>parkinson</strong>ian symptomatologyduring a manic phase in one patient should be mentioned.This occurrence convinced the patient that she was nolonger affected by PD whi<strong>le</strong> a rapid cycling was induced by theadministration of <strong>per</strong>golide and <strong>le</strong>vodopa.The observations of the present study provide evidence forthe underestimation of the association BAD/PD, and emphasizethe need to investigate prob<strong>le</strong>ms proposed by clinical <strong>di</strong>agnosis,physiopathological interpretation and, especially, therapeuticapproach.References1. Graham JM, Grunewald RA, Sagar HJ (1997) Hallucinosis ini<strong>di</strong>opathic Parkinson’s <strong>di</strong>sease. J Neurol Neurosurg Psychiatry63(4):434–3402. Goetz CG, Vogel C, Tanner CM, Stebbins G T (1998) Earlydopaminergic drug-induced hallucinations in <strong>parkinson</strong>ianpatients. Neurology 51:811–8143. K<strong>la</strong>wans HL (1988) Psychiatric side effects during the treatmentof Parkinson’s <strong>di</strong>sease. J Neural Transm Suppl 27:117–1224. Mayeux R (1987) Mental state. Mania and bipo<strong>la</strong>r illness. In:Kol<strong>le</strong>r WC (ed) Handbook of Parkinson’s <strong>di</strong>sease. Dekker,New York, p 1295. Kim E, Zwil AS, McAllister TW, Glosser DS, Stern M, HurtigH (1994) Treatment of organic mood <strong>di</strong>sorders in Parkinson’s<strong>di</strong>sease. J Neuropsychiatry Clin Neurosci 6(2):181–1846. Larmande P, Palisson E, Saikali I, Maillot F (1993)Disappearance of akinesia in Parkinson’s <strong>di</strong>sease during amanic attack. Rev Neurol (Paris) 149(10):557–5587. Scappa S, Taverbaugh P, Ananth J (1993) Episo<strong>di</strong>c tar<strong>di</strong>ve<strong>di</strong>skinesia and <strong>parkinson</strong>ism in bipo<strong>la</strong>r <strong>di</strong>sorder. Can JPsychiatry 38(10):633–6348. Diehl D J, Gershon S (1992) The ro<strong>le</strong> of dopamine in mood<strong>di</strong>sorders. Compr Psychiatry 33(2):115–1209. Przedborski S, Liard A, Hildebrand J (1992) Induction ofmania by apomorphine in a depressed <strong>parkinson</strong>ian patient.Mov Disord 7(3):285–28710. Murphy DL, Bro<strong>di</strong>e HKL, Goodwin FK et al (1971) L-Dopa:regu<strong>la</strong>r induction of hypomania in bipo<strong>la</strong>r manic-depressivepatient. Nature 229:135–136


Neurol Sci (2002) 23:S69–S70 © Springer-Ver<strong>la</strong>g 2002Mo<strong>di</strong>fication of respiratory functionparameters in patients with severeParkinson’s <strong>di</strong>seaseM.F. De Pan<strong>di</strong>s 1 () • A. Starace 1 • F. Stefanelli 2P. Marruzzo 1 • I. Meoli 2 • G. De Simone 2R. Prati 1 • F. Stocchi 31 Department of Neurology, Vil<strong>la</strong> Margherita - SG Moscati,Rehabilitation Institute of Benevento, Italy2 Department of Pneumology, Vil<strong>la</strong> Margherita - SG Moscati,Rehabilitation Institute of Benevento, Italy3 Department of Neurological Sciences, University La Sapienza,Rome, ItalyAbstract Respiratory dysfunction remains one of the most commoncauses of death in patients with complicated Parkinson’s<strong>di</strong>sease (PD). The aim of this study was to investigate pulmonaryfunction in fluctuating PD patients during “on” and “off” statesof the <strong>di</strong>sease. We stu<strong>di</strong>ed 12 fluctuating, non-smoking PDpatients (H&Y stages 3–5) without a history of lung or car<strong>di</strong>ovascu<strong>la</strong>r<strong>di</strong>sease; all patients underwent Hoehn and Yahr sca<strong>le</strong>(H&Y) and Unified Parkinson Disease Rating Sca<strong>le</strong> (UPDRSitems 18–31) to evaluate extrapyramidal impairment, as well aspulmonary function tests (PFT) and arterial blood gas analysesto assess respiratory function. All evaluations were <strong>per</strong>formedduring a stab<strong>le</strong> on state of <strong>di</strong>sease and in an off state produced by12 hours of therapy withdrawal. A restrictive pattern of flow-volumeloop was observed both in on and off states of <strong>di</strong>sease. Inthe off state, we found a significant worsening in both FEV1 andFVC; the FEV1/FVC ratio was unmo<strong>di</strong>fied. These results suggesta restrictive pattern of flow-volume loop in these patients.Pharmacological treatment and <strong>le</strong>vodopa therapy provideclinical benefits and reduced mortality in Parkinson’s <strong>di</strong>sease,yet respiratory dysfunction remains one of the mostcommon causes of death in these patients [1, 2]. A varietyof respiratory prob<strong>le</strong>ms have been reported in the literature:obstructive and restrictive dysfunction, respiratory dysrhythmiasand abnormality of central <strong>contro</strong>l of venti<strong>la</strong>tion[3–8]. Although there is evidence for potential pulmonarydysfunctions, most patients report no respiratory symptomsuntil the final stage of the <strong>di</strong>sease [8]. Our aim was toinvestigate pulmonary function in severe, fluctuating PDpatients during on and off states of the <strong>di</strong>sease, usingspirometry and arterial blood gas analyses.We stu<strong>di</strong>ed 12 consecutive, non-demented, non-smoking,fluctuating PD patients (7 women), recovered in our unit. Themean age of the patients was 67.66±5.46 years, and the durationof <strong>di</strong>sease was between 8 and 25 years (mean, 14.5±0.66 years).The main inclusion criterion was the severity of <strong>di</strong>sease that,accor<strong>di</strong>ng to the H&Y sca<strong>le</strong>, had to be at <strong>le</strong>ast stage 3 (mean4.08±0.66). All patients were on <strong>le</strong>vodopa-substitution therapy(mean dosage, 704.16±145.31 mg/day), with ad<strong>di</strong>tional administrationof add-on DA-agonists for 10 patients and of ICOMTfor 5 patients. Anti-<strong>parkinson</strong>ian drug intake was not changed fora <strong>per</strong>iod of at <strong>le</strong>ast 3 weeks prior to testing. None of the patientshad a history of lung <strong>di</strong>sease or car<strong>di</strong>ovascu<strong>la</strong>r <strong>di</strong>sorders.Patients were subjected to a neurological assessment of<strong>di</strong>sease with Unified Parkinson Disease Rating Sca<strong>le</strong>(UPDRS motor examination items 18–31), pulmonary functiontests (PFT) and arterial blood gas analysis in on and offstates. The off state was reached by a 12-hour withdrawal ofpharmacological therapy. Pulmonary function tests were <strong>per</strong>formedby a dry seal spirometer (Vmax 22 Sensor Me<strong>di</strong>cs,Mi<strong>la</strong>n, Italy) which provided measurements for forced expiratoryvolume in 1 s (FEV1), forced vital capacity (FVC),FEV1/FVC ratio, peak of expiratory flow (PEF), forcedexpiratory flow when 75%, 50% and 25% of FVC remain tobe exha<strong>le</strong>d (FEF75%, FEF50%, FEF25%), and forced expiratoryflow during midd<strong>le</strong> half of the FVC (FEF 25%–75%),accor<strong>di</strong>ng to ATS criteria. To evaluate PaO 2 , PaCO 2 , and pH,all patients underwent an arterial blood gas analysis in thesupine position at the same hour of the day.The results were expressed as mean and standard deviation.A paired t test for parametric data was <strong>per</strong>formed tocompare the results in on and off states. A p value


S70Tab<strong>le</strong> 1 Pulmonary function testing and arterial blood gas analyses in on and off states in 12 fluctuating PD patients. Values are means(SD)On state Off state pFVC 86.50 (26.1) 76.83 (26.74)


Neurol Sci (2002) 23:S71–S72 © Springer-Ver<strong>la</strong>g 2002Visualisation of the subtha<strong>la</strong>micnuc<strong>le</strong>us: a multip<strong>le</strong> sequential imagefusion (MuSIF) technique for <strong>di</strong>rectstereotaxic localisation andposto<strong>per</strong>ative <strong>contro</strong>lM. Egi<strong>di</strong> 1 () • P. Rampini 1 • M. Locatelli 1M. Farabo<strong>la</strong> 2 • A. Priori 3 • A. Pesenti 3 • F. Tamma 4E. Caputo 4 • V. Chiesa 4 • R.M. Vil<strong>la</strong>ni 11 Neurosurgery Clinic, IRCCS Maggiore Policlinico Hospital,Mi<strong>la</strong>n, Italy2 Department of Neurora<strong>di</strong>ology, IRCCS Maggiore PoliclinicoHospital, Mi<strong>la</strong>n, Italy3 Neurology Clinic, IRCCS Maggiore Policlinico Hospital, Mi<strong>la</strong>n,Italy4 Neurology Clinic, S. Paolo Hospital, Mi<strong>la</strong>n, ItalyAbstract A novel multip<strong>le</strong>, sequential image fusion (MuSIF)procedure merging stereotaxic CT with frame<strong>le</strong>ss magneticresonance imaging (MRI) is used since June 2000 to visualiseand <strong>di</strong>rectly localise the subtha<strong>la</strong>mic nuc<strong>le</strong>us (STN) on T2images. In 13 consecutive Parkinson’s cases, intrao<strong>per</strong>ativerecor<strong>di</strong>ng and stimu<strong>la</strong>tion verified bi<strong>la</strong>teral e<strong>le</strong>ctrode imp<strong>la</strong>ntationguided by fused T2 images. In 85% of sides, finalimp<strong>la</strong>ntation opted for visualised target track. Imp<strong>la</strong>nted e<strong>le</strong>ctrodeposition on posto<strong>per</strong>ative T2 images matched p<strong>la</strong>nnedtarget. Clinical follow-up reproduces literature’s best results.This MuSIF technique, effective for <strong>di</strong>rect STN targeting, haspractical advantages: MRI can be <strong>per</strong>formed regard<strong>le</strong>ss ofsurgery time; regu<strong>la</strong>r MR scanning to correct real image <strong>di</strong>stortionis unneeded; and the need for multip<strong>le</strong> localisingtracks is reduced by enabling us to account for each patient’sSTN anatomy.For functional procedures, the subtha<strong>la</strong>mic nuc<strong>le</strong>us (STN) isusually localised through ventriculography or stereotaxicmagnetic resonance imaging (MRI). The former method isinvasive and both are in<strong>di</strong>rect: the STN is not visualised butits position is calcu<strong>la</strong>ted in re<strong>la</strong>tion to visib<strong>le</strong> anatomical<strong>la</strong>ndmarks. Moreover, frequent checks of the MR environmentare needed to correct for real image <strong>di</strong>stortion [1]. Athird method uses image-fusion techniques that correct forimage <strong>di</strong>stortion by matching the MR image to a <strong>di</strong>stortionfreeimage, usually computed tomography (CT) [2].However, only thin T2 spin echo (SE) images allow for sufficientviewing of the STN [3, 4], yet a standard 1.5 T MRscanner produces a limited number of slices, too few to makeT2/CT image fusion reliab<strong>le</strong> for functional purposes.The following multip<strong>le</strong>, sequential image fusion (MuSIF)technique, developed to overcome these technical prob<strong>le</strong>ms,was used on 13 patients with Parkinson’s <strong>di</strong>sease for stereotaxice<strong>le</strong>ctrode imp<strong>la</strong>ntation in 26 STNs from June 2000 toSeptember 2001.The study, <strong>per</strong>formed any day before surgery in thepatient’s best me<strong>di</strong>cation con<strong>di</strong>tion to reduce movement artifacts,acquired frame<strong>le</strong>ss images on a 1.5-T MR scanner(Siemens, Concord, CA). First, a three-<strong>di</strong>mensional (3D)MPRAGE sequence was recorded in the sagittal p<strong>la</strong>ne (1.64mm effective thickness, 7 min duration). Then, a thin-slice (2mm) axial T2 SE sequence was recorded paral<strong>le</strong>l to the AC-PC line (17 min duration). On surgery day, stereotaxic CTwas <strong>per</strong>formed using a GE Light Speed scanner with contiguous1.25-mm slices aligned to the basal ring of the CRWframe (Ra<strong>di</strong>onics, Burlington, MA) and approximately paral<strong>le</strong>lto the AC-PC p<strong>la</strong>ne. Care was taken to include as muchskull as possib<strong>le</strong> in the volume scanned and all <strong>la</strong>ndmarkswere used in subsequent image-fusion procedures.MR and CT image fi<strong>le</strong>s were then loaded into a workstationrunning image-fusion and stereotaxic p<strong>la</strong>nning software(ImageFusion and StereoP<strong>la</strong>n 2.0, Ra<strong>di</strong>onics, Burlington, MA).First, the frame<strong>le</strong>ss MPRAGE study was fused with the stereotaxicCT data using the ImageFusion “bone matching” algorithmuntil visual checking and numeric results showed accuratefusion. Fused images were saved as a new set of MPRAGEimages. Then, the frame<strong>le</strong>ss T2 study was fused onto the fusedMPRAGE image set with the “intensity matching” algorithmand a fused p<strong>la</strong>n of the T2 study was saved, i.e. raw data ratherthan a new image set. Finally, MPRAGE and T2 images thatshared the stereotaxic CT study’s spatial accuracy, stereotaxicvolume and fiducials were avai<strong>la</strong>b<strong>le</strong> for stereotaxic localisationand p<strong>la</strong>nning. The initial target was se<strong>le</strong>cted on transverse T2images inside the hypointensity <strong>la</strong>teral to the red nuc<strong>le</strong>us, at the<strong>le</strong>vel of the anterior boundary of its midd<strong>le</strong> portion. A thinhy<strong>per</strong>intense <strong>la</strong>yer between the STN and the substantia nigra,frequently visib<strong>le</strong> on reformatted coronal images, <strong>per</strong>mittedrefinement of target position. Entry point was set approximately3 cm from the midline, just anterior to the coronal suture.Recor<strong>di</strong>ng and stimu<strong>la</strong>tion were obtained by sing<strong>le</strong> coaxialmicroe<strong>le</strong>ctrodes along one or two simultaneous paral<strong>le</strong>l tracks<strong>di</strong>rected to the calcu<strong>la</strong>ted target. A four-po<strong>le</strong> <strong>le</strong>ad was thenimp<strong>la</strong>nted accor<strong>di</strong>ng to recorded activity and stimu<strong>la</strong>tioneffects. MRI was repeated after imp<strong>la</strong>nting the stimu<strong>la</strong>tor, on thesame sequences as used preo<strong>per</strong>atively. A visual check of theimp<strong>la</strong>nted e<strong>le</strong>ctrode’s actual position with respect to the STNwas easy on T2 images, whi<strong>le</strong> fusing the posto<strong>per</strong>ative studyanew with preo<strong>per</strong>ative p<strong>la</strong>nning allowed the <strong>di</strong>stance betweenimp<strong>la</strong>nted e<strong>le</strong>ctrode and calcu<strong>la</strong>ted target to be measured.The STN was recognisab<strong>le</strong> in all 26 sides. Frame<strong>le</strong>ssMRI <strong>di</strong>stortion (mean scaling factors, 1.4% and 0.7% forMPRAGE and T2 images, respectively) was <strong>le</strong>ss than instereotaxic MRI [1]. The microe<strong>le</strong>ctrode track aimed at theinitially calcu<strong>la</strong>ted target matched recorded STN activity in21 sides and was chosen for final imp<strong>la</strong>ntation in 22 sidesaccor<strong>di</strong>ng to stimu<strong>la</strong>tion effects. Intrao<strong>per</strong>ative cerebral mass<strong>di</strong>sp<strong>la</strong>cement due to CSF <strong>le</strong>akage may partly account for the


S72Pre-o<strong>per</strong>ative Post-o<strong>per</strong>ative Pre-o<strong>per</strong>ativeright target e<strong>le</strong>ctrode artifacts <strong>le</strong>ft targetFig. 1 T2 MR images of the pre-o<strong>per</strong>ative p<strong>la</strong>nned targets and posto<strong>per</strong>ative artifacts of the imp<strong>la</strong>nted e<strong>le</strong>ctrodes in the STN targetad<strong>di</strong>tional paral<strong>le</strong>l track needed in the remaining four STNs,all of which were indeed on the second side o<strong>per</strong>ated. Thefinal position of imp<strong>la</strong>nted e<strong>le</strong>ctrodes in posto<strong>per</strong>ative T2images (Fig. 1) matched p<strong>la</strong>nned target position well: <strong>di</strong>stanceranged from 0.5 to 1.5 mm, which fit very well with theapplication accuracy of stereotaxic frames [5]. Three-monthclinical results reproduce the literature’s best series.DiscussionThis MuSIF technique applied to 1.5 T frame<strong>le</strong>ss MR imagesis highly effective for STN targeting and offers some practica<strong>la</strong>dvantages: MRI can be <strong>per</strong>formed regard<strong>le</strong>ss of surgery time;pre-fusion image <strong>di</strong>stortion is <strong>le</strong>ss than in stereotaxic MRimages; and regu<strong>la</strong>r MR scanner checks to correct for realimage <strong>di</strong>stortion are unneeded. With T2 images, the actua<strong>la</strong>natomy of the given patient’s STN [6] can be taken intoaccount, possibly reducing the need for multip<strong>le</strong> localisingtracks. Targeting a specific portion of the STN holds promiseas a potential ad<strong>di</strong>tional instrument for studying the functionaltopography of the STN. Methods of validating STN targetingtechniques should include posto<strong>per</strong>ative T2 MRI.References1. Burchiel KJ, Nguyen TT, Coombs BD, Szumosky J (1996)MRI <strong>di</strong>stortion and stereotactic neurosurgery using theCosman-Roberts-Wells and Leksell frames. Stereotact FunctNeurosurg 66:123–1362. A<strong>le</strong>xander E III, Kooy HM, van Herk M, Schwartz M,Barnes PD, Tarbell N, Mulkern RV, Holupka EJ, Loeff<strong>le</strong>r JS(1995) Magnetic resonance image-<strong>di</strong>rected stereotactic neurosurgery:Use of image fusion with computerized tomographyto enhance spatial accuracy. J Neurosurg 83:271–2763. Starr P, Vitek JL, DeLong M, Bakay R (1999) Magnetic resonanceimaging-based stereotactic localization of the globuspallidus and subtha<strong>la</strong>mic nuc<strong>le</strong>us. Neurosurgery 44:303-3144. Bejjani BP, Dormont D, Pidoux B, Yelnik J, Damier P,Arnulf I, Bonnet AM, Marsault C, Agid Y, Philippon J,Cornu P (2000) Bi<strong>la</strong>teral subtha<strong>la</strong>mic stimu<strong>la</strong>tion forParkinson’s <strong>di</strong>sease by using three-<strong>di</strong>mensional stereotacticmagnetic resonance imaging and e<strong>le</strong>ctrophysiological guidance.J Neurosurg 92:615–6255. Maciunas RJ, Galloway RL, Latimer JW (1994) The applicationaccuracy of stereotactic frames. Neurosurgery35:682–6956. Van Buren JM, Maccubbin DA (1962) An outline at<strong>la</strong>s of thehuman basal ganglia with estimation of anatomical variants.J Neurosurg 19:811–839


Neurol Sci (2002) 23:S73–S74 © Springer-Ver<strong>la</strong>g 2002Psychophysiological approachin Parkinson’s <strong>di</strong>sease: L-dopa effectson preprogramming and <strong>contro</strong><strong>la</strong>ctivityF. Fattapposta 1 () • F. Pierelli 2 • F. My 1M. Mostarda 1 • S. Del Monte 1 • L. Parisi 1M. Serrao 1 • N. Locuratolo 1 • G. Amabi<strong>le</strong> 11 Clinical Institute of Nervous and Mental Diseases, La SapienzaUniversity, Via<strong>le</strong> dell’Università, 30, I-00185 Rome, Italy2 IRCCS Neuromed, Pozzilli (IS), ItalyAbstract To investigate whether preprogramming(Bereitschaftspotential, BP) and <strong>contro</strong>l activity (skil<strong>le</strong>d <strong>per</strong>formancepositivity, SPP) in a comp<strong>le</strong>x task are sensitive toL-dopa, movement re<strong>la</strong>ted potentials (MRPs) were recorde<strong>di</strong>n 12 non-demented Parkinson’s <strong>di</strong>sease (PD) patients beforeand after acute L-dopa administration, and in 17 <strong>contro</strong>l subjects.After L-dopa administration, the PD patients scored asignificantly higher <strong>per</strong>centage of correct <strong>per</strong>formances(p


S74observed in the off-therapy PD patients group when comparedwith <strong>contro</strong>ls. No <strong>di</strong>fferences were observed betweenon-therapy patients and <strong>contro</strong>ls. During L-dopa therapy, asignificant (p


Neurol Sci (2002) 23:S75–S76 © Springer-Ver<strong>la</strong>g 2002Noradrenergic loss enhances MDMAtoxicity and induces ubiquitin-positivestriatal whorlsM. Ferrucci 1 • M. Gesi 1 • P. Lenzi 1 • P. Soldani 1R. Ruffoli 1 • A. Pel<strong>le</strong>grini 1 • S. Ruggieri 2,3A. Paparelli 1 • F. Fornai 1,3 ()1 Department of Human Morphology and Applied Biology,University of Pisa, Via Roma 55, I-56126 Pisa, Italy2 Department of Neurological Sciences, University of Rome,Rome, Italy3 INM IRCCS Neuromed, Pozzilli (IS), ItalyAbstract Movement <strong>di</strong>sorders involve a number of neurodegenerativecon<strong>di</strong>tions, mostly affecting basal ganglia.Parkinson’s <strong>di</strong>sease (PD) is c<strong>la</strong>ssically defined by the se<strong>le</strong>ctiveloss of dopaminergic neurons in the substantia nigra parscompacta. Administration of specific neurotoxins representsa common tool to reproduce this <strong>le</strong>sion. Among these,amphetamine derivatives act as powerful monoamine neurotoxins,impairing striatal dopamine (DA) axons in mice.Despite the well-investigated effects on striatal DA terminals,only spora<strong>di</strong>c stu<strong>di</strong>es have focused on the potential toxicityof amphetamines towards post-synaptic neurons withinthe striatum. In the present work we found that 3,4-methy<strong>le</strong>ne<strong>di</strong>oxymethamphetamine(MDMA) produces ultrastructura<strong>la</strong>lterations in striatal cells, featuring as membraneouswhorls, positive for ubiquitin and heat shock protein 70.These morphological alterations were enhanced in locuscoeru<strong>le</strong>us-<strong>le</strong>sioned mice.Most degenerative <strong>di</strong>sorders involve the basal ganglia.Parkinson’s <strong>di</strong>sease (PD) is characterized by a se<strong>le</strong>ctive lossof dopaminergic (DA) neuronal cells belonging to thenigrostriatal pathway [1].Damage to striatal DA fibers can be produced either bymicroinfusing monoamine neurotoxins into the substantianigra pars compacta (SNpc), or systemically administeringspecific DA neurotoxins like amphetamine derivatives [2].These compounds are, unfortunately, <strong>la</strong>rgely abused byhumans and produce a robust effect on the DA system,increasing at first striatal DA re<strong>le</strong>ase [3], so <strong>le</strong>a<strong>di</strong>ng to thedamage of the nigrostriatal DA pathway, possibly due to theproduction of free ra<strong>di</strong>cals and reactive oxygen species.Recently, a number of ex<strong>per</strong>imental and clinical stu<strong>di</strong>esdocumented the importance of the locus coeru<strong>le</strong>us (LC) inmodu<strong>la</strong>ting the progression of neurodegenerative <strong>di</strong>sorders;concomitantly, reduction of noradrenergic (NA) neurotransmissionin various brain areas innervated by the LC is found asa constant e<strong>le</strong>ment of the pathobiochemistry of PD. Altogether,these fin<strong>di</strong>ngs strongly suggest that NA loss could have a specificro<strong>le</strong> in sustaining the <strong>di</strong>sease [4]. Among amphetaminederivatives, 3,4-methy<strong>le</strong>ne<strong>di</strong>oxymethamphetamine (MDMA) iswell known for its neurotoxic effects against monoamine axons,whi<strong>le</strong> the effects in striatal GABA cells are <strong>le</strong>ss investigated.In this study we evaluated, at the ultrastructural <strong>le</strong>vel, theeffects of MDMA on striatal neurons of the mouse. In ad<strong>di</strong>tion,we investigated the ro<strong>le</strong> of LC in modu<strong>la</strong>ting the occurrenceof subcellu<strong>la</strong>r alterations in striatal <strong>per</strong>ikarya.Ma<strong>le</strong> C57 b<strong>la</strong>ck mice (C57BL/6J) 9–10 weeks old were<strong>di</strong>vided into five groups of 10 mice each. To one group weadministered intra<strong>per</strong>itoneally the NA neurotoxin DSP-4 (50mg/kg). Three days <strong>la</strong>ter, a subgroup of DSP-4 treated miceand a group of <strong>contro</strong>ls received MDMA hydrochloride(three administrations of 30 mg/kg at 2-h intervals). Oneweek after MDMA administration, one group was sacrificedand the striatum was processed in order to measure catecho<strong>la</strong>minesand their metabolites by HPLC as previouslydescribed [5].Another group of mice was anaesthetized with chloralhydrate (4 ml/kg), <strong>per</strong>fused and processed for e<strong>le</strong>ctronmicroscopy and immunocytochemistry. Thin sections of 50nm were deosmicated in a saturated solution of Na-meta<strong>per</strong>iodatefor 30 min, washed in PBS and incubated in the primaryantibody (Ab-I) for 24 h at 4° C. We used Ab-I anti-ubiquitinand anti-heat shock protein 70 (HSP-70), <strong>di</strong>luted 1:100;secondary antibo<strong>di</strong>es were conjugated with gold partic<strong>le</strong>s of10 nm <strong>di</strong>ameter. Measurement of striatal whorls was carriedout by counting the number of nuc<strong>le</strong>ar or cytosolic whorls outof the total number of neurons under observation. The sameprocedure was followed for each animal from each <strong>di</strong>fferentgroup of treatment. Data are given as <strong>per</strong>centage of whorlsand they were compared using ANOVA with Sheffè’s posthocanalysis. Null hypothesis (H o ) was rejected when p


S76Fig. 1 E<strong>le</strong>ctron microscope micrograph of a cytosolic whorlwithin a striatal cell from MDMA-treated mouse. Multip<strong>le</strong>concentric membranes (arrowhead) and a more e<strong>le</strong>ctrondensecore (*) are evidentDiscussionThis work points out the occurrence of ultrastructural alterationsinto striatal post-synaptic neurons following MDMAtreatment, consisting of whorls of concentric membranes.Ubiquitin immunostaining suggests these formations to befunctionally re<strong>la</strong>ted to the ubiquitin-proteasome pathway, a cellu<strong>la</strong>rsystem which p<strong>la</strong>ys a pivotal ro<strong>le</strong> in proteolysis and whichis involved in the regu<strong>la</strong>tion of cell cyc<strong>le</strong> [6]. Our fin<strong>di</strong>ngs ofubiquitin-positive whorls in association with the smooth endop<strong>la</strong>smicreticulum, as well as their positive staining for thestress-induced cha<strong>per</strong>one-like protein HSP-70, strongly supportthe hypothesis that they represent the morphological profi<strong>le</strong>of an alteration of the ubiquitin-proteasome pathway.Membranous whorls were described within neurons of specificbrain areas in several pathological con<strong>di</strong>tions; in PD typicalneuronal inclusions, Lewy bo<strong>di</strong>es (LB), consist ofeosinophilic material found in the cytop<strong>la</strong>sm. Immunostainingshows ubiquitin as a prominent and constant component of LB.Lewy bo<strong>di</strong>es are also present in other neurodegenerative <strong>di</strong>sorders,such as <strong>di</strong>ffuse LB <strong>di</strong>sease and Alzheimer’s <strong>di</strong>sease.Recently, mutant TorsinA, a novel protein identified asresponsib<strong>le</strong> for the early-onset torsion dystonia, has been foundto stain LB [7]. TorsinA in human brain is mainly expressed inDA neurons of the SNpc; this protein, which contains an ATPbin<strong>di</strong>ngsite, is thought to <strong>per</strong>form cha<strong>per</strong>one functions,involved in the identification of misfolded proteins and theirrefol<strong>di</strong>ng or degradation [7]. In this respect, our fin<strong>di</strong>ng ofHSP-70 immunostaining in striatal whorls might represent anexamp<strong>le</strong> of a cha<strong>per</strong>one mo<strong>le</strong>cu<strong>le</strong> recruited following oxidativedamage induced by MDMA treatment.In conclusion, we hypothesize that whorls represent, atthe morphological <strong>le</strong>vel, an enhancement of a more general,physiological response of the cell to the production of alteredproteins. In particu<strong>la</strong>r, the marked striatal DA re<strong>le</strong>aseobserved after MDMA administration, constantly associatedwith the production of highly reactive species, can oxi<strong>di</strong>ze<strong>la</strong>rge amounts of intracellu<strong>la</strong>r proteins. Moreover, a recentstudy [8] demonstrated that DA inhibits the proteasome pathway,making more critical the oxidative effects of DA on“proteasome-impaired” striatal cells.References1. Hornykievicz O, Kish SJ (1986) Biochemical pathophysiologyof Parkinson’s <strong>di</strong>sease. Adv Neurol 45:19–342. Seiden LS, Ricaurte GA (1987) Neurotoxicity of methamphetamineand re<strong>la</strong>ted drugs. In: Meltzer HY (ed)Psychopharmacology: the third generation of progress.Raven, New York, pp 359–3663. O’Dell SJ, Weihmul<strong>le</strong>r FB, Marshall JF (1991) Multip<strong>le</strong>methamphetamine injections induce marked increases in extracellu<strong>la</strong>rstriatal dopamine which corre<strong>la</strong>te with subsequent neurotoxicity.Brain Res 564:256–2604. Gesi M, Soldani P, Giorgi FS et al (2000) The ro<strong>le</strong> of the locuscoeru<strong>le</strong>us in the development of Parkinson’s <strong>di</strong>sease. NeurosciBiobehav Rev 24:655–6685. Fornai F, Giorgi FS, Gesi M et al (2001) Biochemical effects ofthe monoamine neurotoxins DSP-4 and MDMA in specificbrain regions of MAO-B-deficient mice. Synapse 39:213–2216. De Martino GN, S<strong>la</strong>ughter CA (1999) The proteasome, a novelprotease regu<strong>la</strong>ted by multip<strong>le</strong> mechanisms. J Biol Chem274:22123–221267. Sharma N, Hewett J, Ozelius LJ et al (2001) A close associationof TorsinA and α-synuc<strong>le</strong>in in Lewy bo<strong>di</strong>es. A fluorescence resonanceenergy transfer study. Am J Pathol 159:339–3448. Kel<strong>le</strong>r JN, Huang FF, Dimayuga ER et al (2000) Dopamineinduces proteasome inhibition in neural PC12 cell line. FreeRa<strong>di</strong>c Biol Med 29:1037–1042


Neurol Sci (2002) 23:S77–S78 © Springer-Ver<strong>la</strong>g 2002Clinical pre<strong>di</strong>ctors in Parkinson’s<strong>di</strong>seaseE. Gasparoli • D. Delibori • G. Po<strong>le</strong>sello • L. SantelliM. Ermani • L. Battistin • F. Bracco ()Department of Neurological and Psychiatric Sciences, Universityof Padua, Padua, ItalyAbstract Parkinson’s <strong>di</strong>sease is characterized by heterogeneityof clinical presentations, association of signs and symptoms,rate of progression, and response to therapy. The aim ofthis prospective 5-year study was to evaluate whether clinicalfeatures at onset were pre<strong>di</strong>ctive of the subsequent progression.Two courses were identified which <strong>di</strong>ffered in the characteristicsat onset. Slow course was characterized by earlierage at onset, <strong>la</strong>teralization of motor signs, rest tremor, andabsence of gait <strong>di</strong>sturbance. Rapid course presented older age,<strong>le</strong>ss evident <strong>la</strong>teralization of signs, predominance of bradykinesia-rigi<strong>di</strong>tyand gait <strong>di</strong>sturbance. Our results confirmed thatPD is clinically heterogeneous and specific patterns of onsetseem to be associated with <strong>di</strong>fferent rates of <strong>di</strong>sease progression.Pre<strong>di</strong>ctive models based on these clinical characteristicshave a good sensitivity in in<strong>di</strong>cating a slow <strong>di</strong>sease progressionbut are not reliab<strong>le</strong> in in<strong>di</strong>cating a rapid evolution.Parkinson’s <strong>di</strong>sease (PD) is characterised by heterogeneity ofclinical presentations, association of signs and symptoms,rate of progression of the <strong>di</strong>sease, and response to therapy,suggesting the existence of <strong>di</strong>fferent subgroups, possiblyre<strong>la</strong>ted to <strong>di</strong>fferent underlying processes. In the pre-<strong>le</strong>vodopaera, the clinical data of Hoehn and Yahr [1] revea<strong>le</strong>d a greatheterogeneity of <strong>di</strong>sease progression: 37% of the patientswhose duration of <strong>di</strong>sease was <strong>le</strong>ss than 5 years were in stageIII, whi<strong>le</strong> 34% of patients whose duration of <strong>di</strong>sease was tenyears were in stage I or II. The patients in the same stage hada <strong>la</strong>rge variation of <strong>di</strong>sease duration.Possib<strong>le</strong> tools for monitoring <strong>di</strong>sease progression areevolution of clinical pattern, modality of response to therapy,neuroimaging techniques and genetic research. In the case ofautosomal recessive <strong>parkinson</strong>ism, genetic tests could pre<strong>di</strong>ctthe slow progression with good response to <strong>le</strong>vodopa.Sequential neuroimaging techniques seem to be a useful toolin monitoring the intra-in<strong>di</strong>vidual progression of dopaminergiccell loss in PD but cannot pre<strong>di</strong>ct the kind of progressionwhen <strong>per</strong>formed only at onset. Currently, <strong>di</strong>sease progressionrate can be evaluated with clinical in<strong>di</strong>cators: rate ofimpairment of motor signs and their temporal association,the presence of non-motor signs, the variation in theresponse to therapy, the presence of motor and non-motorcomplications, and comorbi<strong>di</strong>ty.The aim of this study was to evaluate whether clinicalfeatures at onset and their <strong>di</strong>fferent sequence of associationcould be pre<strong>di</strong>ctive of the subsequent rate of <strong>di</strong>sease progression.The pattern of <strong>di</strong>sease progression may define theprognosis and influence therapeutic strategies.A prospective longitu<strong>di</strong>nal study was carried out in 103patients (69 men) with a <strong>di</strong>agnosis of PD accor<strong>di</strong>ng to theParkinson’s Disease Society Brain Research Centre’s clinical<strong>di</strong>agnostic criteria. The exclusion criteria were atypical<strong>parkinson</strong>ism (at onset and follow up) and dementia at onset.At onset the following parameters were recorded: sex, age atonset, main motor sign, <strong>la</strong>teralization of motor signs, associationof signs/symptoms, presence of depression, and presenceof gait <strong>di</strong>sturbance. At the 5-year follow-up, impairmentof motor score was evaluated by Unified Parkinson’sDisease Rating Sca<strong>le</strong> (UPDRS) and cognitive impairmentwas measured by MMSE (Mini-Mental State Examination).Complications of therapy (motor fluctuations and dyskinesia),impairment of postural ref<strong>le</strong>xes, functionally limitinggait <strong>di</strong>sturbance and cumu<strong>la</strong>tive dose of <strong>le</strong>vodopa wererecorded to evaluate the severity of <strong>di</strong>sease progression.Cluster analysis was used to identify two groups ofpatients based on UPDRS motor score, presence of motorfluctuations and dyskinesia at 5 years. This <strong>di</strong>stinction in twogroups was used to give a prognostic meaning to the characteristicsevaluated at onset. To evaluate the prognostic meaningof the variab<strong>le</strong>s at onset, the progress of each componentof progression, the UPDRS motor score and possib<strong>le</strong> re<strong>la</strong>tionshipsbetween each parameter of onset and presence offluctuations and dyskinesias were analyzed. The characteristicssignificantly corre<strong>la</strong>ted with the outcome were used toobtain a pre<strong>di</strong>ctive model using logistic regression.For statistical analysis chi-square tests were used for all<strong>di</strong>cotomic variab<strong>le</strong>s and Student’s t test was used for all continuousvariab<strong>le</strong>s. All variab<strong>le</strong>s were then subjected to clusteranalysis.Two <strong>di</strong>sease courses (slow and rapid) were identified bycluster analysis. Patients with a slow evolution (61%) werecharacterized by earlier age at onset, <strong>la</strong>teralization of <strong>parkinson</strong>iansigns, preva<strong>le</strong>nce of rest tremor and absence of gait<strong>di</strong>sturbance. Patients with rapid progression (39%) had anolder age, absence of <strong>la</strong>teralization of <strong>parkinson</strong>ian signs,predominance of bradykinesia-rigi<strong>di</strong>ty and gait <strong>di</strong>sturbance(Tab<strong>le</strong> 1).The statistical corre<strong>la</strong>tion between the variab<strong>le</strong>s at onsetand the UPDRS motor score after 5 years suggested a significantcorre<strong>la</strong>tion of age at onset (p=0.01), main motor sign(p=0.02), <strong>la</strong>ck of <strong>la</strong>teralization of clinical signs (p=0.001)and presence of gait <strong>di</strong>sturbance (p=0.03). Age at onset(51.0±15.3 years vs. 61.5±9.1) was significantly corre<strong>la</strong>tedwith presence of dyskinesias (p=0.001), whi<strong>le</strong> no other characteristicat onset was significantly corre<strong>la</strong>ted with the <strong>la</strong>terpresence of motor fluctuations.Pre<strong>di</strong>ctive models based on these clinical characteristicshave a good sensitivity (87.3% of correct pre<strong>di</strong>ction) in in<strong>di</strong>catinga slow <strong>di</strong>sease progression but poor sensitivity in in<strong>di</strong>-


S78Tab<strong>le</strong> 1 Characteristics of PD patients at onset, by type of progressionSlow progression (n=63)Rapid progression (n=40)UPDRS motor score a 11.9 (3.9) 25.1 (5.1)Motor fluctuations, % 19.0 52.5Dyskinesias, % 15.8 30.0Age at onset, years a 56.0 (11.0) 63.5 (11.0)*Lack of <strong>la</strong>teralization, % 17.4 45.0*Main motor sign, %Tremor 58.8 42.5Rigi<strong>di</strong>ty 41.2 57.5Depression, % 9.5 12.5Gait <strong>di</strong>sturbance, % 3.2 10.0*p=0.002a Values are mean (SD)cating a rapid evolution (37.5% pre<strong>di</strong>ction of correctresponse).Our results confirm that PD is clinically heterogeneousand suggest that specific patterns of onset may be associatedwith <strong>di</strong>fferent rates of <strong>di</strong>sease progression. Several stu<strong>di</strong>eshave addressed the question of clinical heterogeneity byproposing subgroups <strong>di</strong>stinguished by age at onset, variab<strong>le</strong>progression, family history, patterns of motor symptoms andassociated non-motor fin<strong>di</strong>ngs such as dementia and depression.In these stu<strong>di</strong>es the <strong>di</strong>sease progression was evaluatedconsidering the progression of sing<strong>le</strong> signs, and not thesequential association of signs. Homogeneous pre<strong>di</strong>ctive criteriawere not found and their results show that early <strong>di</strong>seaseonset [2, 3] with tremor dominance corre<strong>la</strong>tes with slow progression,whi<strong>le</strong> older age at onset and presentation withbradykinesia are pre<strong>di</strong>ctive of a more aggressive course [4].Cluster analysis was used to identify subgroups [5]: inour study two <strong>di</strong>fferent groups of patients (slow 60% andrapid 40%) were identified. Our results provide a pre<strong>di</strong>ctivemodel which reliably pre<strong>di</strong>cts only a slow <strong>di</strong>sease progression,but not a rapid evolution. We found that age at onsetwas significantly associated with dyskinesias, whi<strong>le</strong> no othercharacteristic at onset was significantly corre<strong>la</strong>ted with the<strong>la</strong>ter presence of motor fluctuations. This supports thehypothesis of possib<strong>le</strong> <strong>di</strong>fferent pathogenic mechanisms.In the previous stu<strong>di</strong>es [6, 7], the motor complicationswere considered in corre<strong>la</strong>tion with the <strong>di</strong>sease stage, theduration and the kind of exposition to <strong>le</strong>vodopa more thanwith the progression rate of <strong>di</strong>sease.The existence of <strong>di</strong>fferent subtypes of <strong>di</strong>sease is not onlyrevea<strong>le</strong>d by the characteristics at <strong>di</strong>sease onset but also bythe response to therapy. In a long-term study on the efficacyof dopamine agonist vs. <strong>le</strong>vodopa in the treatment of earlypreviously untreated PD patients [8], 35% of patients inmonotherapy remained clinically improved for 4 years,showing that there are patients who respond better to thera-py and/or with a more slowly progressing course of <strong>di</strong>sease.Our results confirm this hypothesis.The possibility of pre<strong>di</strong>cting, at <strong>di</strong>sease onset, the rate ofprogression of symptoms, the quality of therapy responseand <strong>la</strong>tency and severity of motor and non-motor complicationsmay give a instrument to identify the most appropriateinitial therapeutic strategy of the sing<strong>le</strong> subgroups. Thismethod is useful in consideration of both efficacy in pharmacological<strong>contro</strong>l and presence of complications in longtermtherapy.References1. Hoehn MM, Yahr MD (1967) Parkisonism: onset, progression,mortality. Neurology 17:427–4422. Goetz CG et al (1988) Risk factors for progression in Parkinson’s<strong>di</strong>sease. Neurology 38:1841–18443. Jankovic J, McDermott M et al (1990) Variab<strong>le</strong> expression ofParkinson’s <strong>di</strong>sease: a baseline analysis of the DATATOPcohort. Neurology 40:1529–15344. Jankovic J, Kapa<strong>di</strong>a AS (2001) Functional decline inParkinson’s <strong>di</strong>sease. Arch Neurol 58:1611–16155. Graham JM, Sagar HJ (1999) A data-driven approach to thestudy of heterogeneity in i<strong>di</strong>opathic Parkinson’s <strong>di</strong>sease: identificationof three <strong>di</strong>stinct subtypes. Mov Disord 14:10–206. Goetz CG, Stebbins GT, B<strong>la</strong>succi LM (2000) Differential progressionof motor impairment in <strong>le</strong>vodopa-treated Parkinson’s<strong>di</strong>sease. Mov Disord 15:479–4847. Schrag A, Quinn N (2000) Dyskinesias and motor fluctuationsin Parkinson’s <strong>di</strong>sease. A community-based study. Brain123:2297–23058. Rinne UK, Bracco F, Chouza C et al (1988) Early treatment ofParkinson’s <strong>di</strong>sease with cabergoline de<strong>la</strong>ys the onset ofmotor complications. Results of a doub<strong>le</strong>-blind <strong>le</strong>vodopa <strong>contro</strong>l<strong>le</strong>dtrial. Drugs 55[Suppl]:23–30


Neurol Sci (2002) 23:S79–S80 © Springer-Ver<strong>la</strong>g 2002Screening cognitive declinein dementia: preliminary data onthe Italian version of the IQCODEV. Isel<strong>la</strong> • M.L. Vil<strong>la</strong> • L. Fratto<strong>la</strong> • I. Appollonio ()Neurological Department, San Gerardo Hospital, University ofMi<strong>la</strong>n-Bicocca, Monza, ItalyAbstract The IQCODE is a retrospective questionnaire forcaregivers about changes which occurred in a patient’s cognitiveand functional efficiency in the previous 10 years oflife. Previous stu<strong>di</strong>es demonstrated the vali<strong>di</strong>ty of theIQCODE for the screening of dementia simi<strong>la</strong>r to that of tra<strong>di</strong>tionalcognitive screening tests, with the ad<strong>di</strong>tional advantageof allowing the detection of cognitive change, ratherthan just cognitive impairment. The present pa<strong>per</strong> deals withthe preliminary results of the validation of the Italian versionof the questionnaire in a samp<strong>le</strong> of 45 mild to severelydemented patients and 13 patients with mild cognitiveimpairment (MCI), compared to 20 cognitively intact elderlysubjects. The IQCODE demonstrated satisfactory <strong>di</strong>scriminativepower for dementia as well as for MCI and a goodcorre<strong>la</strong>tion with the MMSE.Informant-based instruments have often proved to be aseffective as brief cognitive tests for the screening of dementia[2]. In spite of some limitations (such as the need for acompliant and reliab<strong>le</strong> informant and the in<strong>di</strong>rect nature ofthe assessment), such instruments have two important pro<strong>per</strong>tieswith respect to the tra<strong>di</strong>tional <strong>di</strong>rect neuropsychologica<strong>la</strong>ssessment: better applicability in subjects with particu<strong>la</strong>rlyhigh or low educational and pre-morbid cognitive <strong>le</strong>velsand independence from patients’ compliance.The Informant Questionnaire on Cognitive Decline in theElderly (IQCODE) [1] is a 26-item informant-based questionnairewhich requires caregivers to compare the currentpatient’s cognitive and functional <strong>per</strong>formances with his/her10-year earlier <strong>le</strong>vel of functioning. The IQCODE also has thead<strong>di</strong>tional advantage of allowing measurement of cognitivedecline from previous cognitive <strong>le</strong>vel, which is one of themajor criteria required for the clinical <strong>di</strong>agnosis of dementia.Several stu<strong>di</strong>es have already verified the vali<strong>di</strong>ty of theIQCODE as a screening test for dementia not only in its originalEnglish version, but also in some other <strong>la</strong>nguages [3–5].The present pa<strong>per</strong> deals with the preliminary results of astudy evaluating the <strong>di</strong>scriminative power of the Italian versionof the IQCODE in a clinical samp<strong>le</strong> of cognitivelyimpaired patients compared to normal <strong>contro</strong>ls and of itsconcurrent vali<strong>di</strong>ty with respect to the MMSE.Patients se<strong>le</strong>cted for the study were 45 demented subjectsfulfilling DSM-IV <strong>di</strong>agnostic criteria for dementia [6] and 13subjects affected by mild cognitive impairment (MCI)accor<strong>di</strong>ng to Petersen’s criteria [7]. Twenty in<strong>di</strong>vidualsreferred to our Neuropsychological Unit for subjective cognitivecomp<strong>la</strong>ints, with normal <strong>per</strong>formance at psychometrictesting, were enrol<strong>le</strong>d as <strong>contro</strong>ls. Demented patients werethen <strong>di</strong>vided into three subgroups accor<strong>di</strong>ng to their MMSEscore: MMSE score >18, mildly demented (n=25); MMSEscore = 18–12, moderately demented (n=13); MMSE score


S80Tab<strong>le</strong> 1 Socio-demographic features and MMSE and IQCODE scores of the five study groups compared with one-way ANOVA. Valuesare mean (SD) un<strong>le</strong>ss otherwise in<strong>di</strong>catedMild Moderate SevereControls MCI dementia dementia dementia(n=20) (n=13) (n=25) (n=13) (n=7)Age, years 67.7±4.9 69.7±6.2 69.8±7.3 72.1±6.9 70.1±6.6Ma<strong>le</strong>s, n (%) 55 60 55 60 55Education, years 7.2±3.4 7.0±2.6 6.6±3.7 8.0±5.2 6.7±2.9MMSE 28.7±1.5 7.1±2.5* 22.3±3.2 § 15.1±1.3 § * 9.3±2.2 § *IQCODE 87±7.9 96±11 § * 102.6±15 § 112.1±11 § * 120.9±6 § *§ p


Neurol Sci (2002) 23:S81–S82 © Springer-Ver<strong>la</strong>g 2002Frequency of apraxia of eyelidopening in the general popu<strong>la</strong>tion an<strong>di</strong>n patients with extrapyramidal<strong>di</strong>sordersP. Lamberti • M. De Mari • A. Zenzo<strong>la</strong>M.S. Aniello • G. Defazio ()Department of Neurologic and Psychiatric Sciences, Section ofNeurology, University of Bari, I-70124 Bari, ItalyAbstract We ascertained the preva<strong>le</strong>nce of apraxia of eyelidopening (AEO) in a community located in Puglia, a region ofsouthern Italy. The crude preva<strong>le</strong>nce rate was 59 <strong>per</strong> million(95% confidence interval, 24–173). AEO coexisted withadult onset b<strong>le</strong>pharospasm in 75% of cases, with atypical<strong>parkinson</strong>ism in 25% of cases. Among the overall patientpopu<strong>la</strong>tion seen at our movement <strong>di</strong>sorders clinic from 1987to 1997, AEO was iso<strong>la</strong>ted in 10 otherwise healthy in<strong>di</strong>viduals,associated with adult-onset dystonia in 13 cases, andassociated with a <strong>parkinson</strong>ian syndrome in 9 cases. The frequencyof AEO was 10.8% in the dystonia group, and 2.1%in the overall <strong>parkinson</strong>ian group (Parkinson’s <strong>di</strong>sease,0.7%; progressive supranuc<strong>le</strong>ar palsy, 33.3%). In twopatients with possib<strong>le</strong> progressive supranuc<strong>le</strong>ar palsy, AEOworsened after increasing <strong>le</strong>vodopa dosage or acute apomorphinechal<strong>le</strong>nge and <strong>di</strong>sappeared following <strong>le</strong>vodopa <strong>di</strong>scontinuation.AEO developing in the setting of a <strong>parkinson</strong>iansyndrome may be either <strong>di</strong>sease- or drug-re<strong>la</strong>ted.Apraxia of eyelid opening (AEO) is an important and oftenundetected cause of non-paralytic inability to maintain theeyes open and involuntary <strong>per</strong>sistent eye closure [1]. Thecon<strong>di</strong>tion is thought to be due to a range of <strong>di</strong>sorders involvingmotor activities in the orbicu<strong>la</strong>ris oculi (OO) and <strong>le</strong>vatorpalpebrae su<strong>per</strong>ior (LPS) musc<strong>le</strong>s [2, 3]. AEO may beiso<strong>la</strong>ted in otherwise healthy in<strong>di</strong>viduals [4] or associatedwith extrapyramidal <strong>di</strong>sorders [1] inclu<strong>di</strong>ng dystonia, i<strong>di</strong>opathicParkinson’s <strong>di</strong>sease (IPD) and <strong>parkinson</strong>ism otherthan IPD. No formal epidemiologic study has focused on thefrequency of AEO in the general popu<strong>la</strong>tion, and only a fewstu<strong>di</strong>es have dealt with the frequency of AEO in extrapyramidal<strong>di</strong>sorders.We <strong>di</strong>agnosed AEO accor<strong>di</strong>ng with Lepore andDuvoisin’s clinical criteria [5]. These included: (i) no sign ofongoing orbico<strong>la</strong>ris oculi (OO) contractions such as loweringof the brows beneath the su<strong>per</strong>ior orbital margins(Charcot’s sign); (ii) marked frontalis musc<strong>le</strong> overaction during<strong>per</strong>iod of inability to raise eyelids; (iii) no ocu<strong>la</strong>rmotor/ocu<strong>la</strong>r sympathetic nerve dysfunction or ocu<strong>la</strong>rmyopathy. E<strong>le</strong>ctromyographic examinations of the OO andLPS [3] musc<strong>le</strong>s were not <strong>per</strong>formed since the tests areunwieldy, require ex<strong>per</strong>tise not rea<strong>di</strong>ly avai<strong>la</strong>b<strong>le</strong> in our settingand, finally, seem unnecessary to the <strong>di</strong>agnosis of AEO.The preva<strong>le</strong>nce of AEO in the general popu<strong>la</strong>tion wasstu<strong>di</strong>ed in an area inclu<strong>di</strong>ng 3 neighboring towns(Casamassima, Conversano and Putignano) situated in theprovince of Bari, Puglia, southern Italy. The overall popu<strong>la</strong>tionwas 67 606 on 1 January 1998 (the preva<strong>le</strong>nce day). Inthis area, healthcare services are provided by the ItalianNational Health System through about 50 general practitionersand 3 general hospitals with both neurological and ophthalmologicoutpatient/inpatient departments. Patients seekingme<strong>di</strong>cal attention by general practitioners are usuallysubmitted to physicians of local hospitals or the UniversityHospital of Bari where a tertiary referral movement <strong>di</strong>sorderscenter and an ophthalmologic botulinum toxin unit arelocated. Cases were identified by reviewing records codedfor appropriate <strong>di</strong>agnoses (inclu<strong>di</strong>ng apraxia of eyelid opening,b<strong>le</strong>pharospasm, cranial dystonia or other focal and segmental/multifocaldystonia, i<strong>di</strong>opathic Parkinson’s <strong>di</strong>sease,and secondary <strong>parkinson</strong>ism) of patients atten<strong>di</strong>ng the outpatient/inpatientdepartments of neurology/ophthalmologyof the general and university hospitals from 1987 through1999. Diagnosis of dystonia, IPD and <strong>parkinson</strong>ism otherthan IPD was made accor<strong>di</strong>ng with accepted criteria [6] byex<strong>per</strong>ienced movement-<strong>di</strong>sorder specialists based ondetai<strong>le</strong>d history, physical and neurologic examinations,appropriate <strong>la</strong>boratory and neuroimaging tests, and (for<strong>parkinson</strong>ian patients) the presence of convincing/unconvincingresponse to <strong>le</strong>vodopa.By this approach, we identified 4 cases in the study area,alive at preva<strong>le</strong>nce day. Three fema<strong>le</strong> patients (aged 52, 67and 70 years) suffered from AEO associated with b<strong>le</strong>pharospasm(BSP). None of them reported prior neuro<strong>le</strong>ptictreatment or showed neurological signs other than dystoniaand abnormalities on head imaging stu<strong>di</strong>es. In a ma<strong>le</strong> patientaged 63 years, AEO developed one year after the onset ofpossib<strong>le</strong> progressive supranuc<strong>le</strong>ar palsy <strong>di</strong>agnosed accor<strong>di</strong>ngwith NINDS criteria. Based on this samp<strong>le</strong>, a crude preva<strong>le</strong>ncerate of 59 <strong>per</strong> million was calcu<strong>la</strong>ted (95% confidenceinterval, 24–173).Among the overall patient popu<strong>la</strong>tion seen at our movement<strong>di</strong>sorders clinic from 1987 to 1997, AEO was observe<strong>di</strong>n the following clinical settings: (i) iso<strong>la</strong>ted in otherwisehealthy in<strong>di</strong>viduals (n=10); adult-onset dystonia (4 ma<strong>le</strong>sand 9 fema<strong>le</strong>s aged 42–69), inclu<strong>di</strong>ng b<strong>le</strong>pharospam (n=10),cranial dystonia (n=2) and writer’s cramp (n=1); Parkinson’s<strong>di</strong>sease (two ma<strong>le</strong>s and 1 fema<strong>le</strong> aged 53–73 years); and possib<strong>le</strong>or probab<strong>le</strong> (accor<strong>di</strong>ng to NINDS criteria) progressivesupranuc<strong>le</strong>ar palsy (PSP) (3 ma<strong>le</strong>s and 3 fema<strong>le</strong>s aged 53–74years). Most patients suffered from episodes of involuntarydrooping of the eyelids. Although this feature was notincluded in the criteria used by Lepore and Duvoisin [5],


S82transient failure to sustain lid e<strong>le</strong>vation is now considered afurther manifestation of AEO [1, 2]. In the same <strong>per</strong>iod weobseved 120 patients with adult-onset dystonia, 399 withParkinson’s <strong>di</strong>sease and 33 with possib<strong>le</strong> or probab<strong>le</strong> PSP.Thus, the frequency of AEO was 10.8% in the dystoniagroup, and 2.1% in the overall <strong>parkinson</strong>ism group(Parkinson’s <strong>di</strong>sease, 0.7%; PSP, 33.3%).Clinical and demographic features of patients with iso<strong>la</strong>tedAEO have been described elsewhere [4]. In most patientsincluded in the dystonia group, namely those suffering fromBSP, it was not possib<strong>le</strong> to assess the time e<strong>la</strong>psing betweenAEO and BSP onset. Among <strong>parkinson</strong>ian patients, AEOdeveloped 2–6 years (mean, 4.1; SD, 1.3) following the onsetof the <strong>parkinson</strong>ian syndrome. Two patients, one with IPD, theother with PSP, developed AEO before the introduction of <strong>le</strong>vodopaor other anti<strong>parkinson</strong>ian drugs. When they were treatedwith benserazide/<strong>le</strong>vodopa 50/200 mg thrice daily, neitherPSP nor AEO improved. Seven <strong>parkinson</strong>ian patients developedAEO 1–5 years (mean, 3.2; SD, 1.6) following the introductionof <strong>le</strong>vodopa. In two of such patients (both sufferingfrom possib<strong>le</strong> PSP), AEO worsened after increasing <strong>le</strong>vodopadosage and <strong>di</strong>sappeared when <strong>le</strong>vodopa was <strong>di</strong>scontinued.Later, a dose of apomorphine (APO) widely accepted for acutetests (5 mg subcutaneously preceded by 5 days treatment withdom<strong>per</strong>idone) had no significant effect on limb motor activitybut induced episodes of involuntary drooping of the eyelidsand prolonged (a few minutes) inability to raise eyelids followingvoluntary or involuntary eye closure. APO effect <strong>la</strong>stedapproximately one hour. One of these patients has beendescribed in detail elsewhere [7]. The other was a 64-year-oldma<strong>le</strong> subject suffering from possib<strong>le</strong> PSP for 3 years whodeveloped AEO 3 months following the introduction ofbenserazide/<strong>le</strong>vodopa (50/200 mg daily).DiscussionAlthough the number of cases was small, and confidence intervalwas wide, our rate suggests that AEO may be as preva<strong>le</strong>ntas a number of better known neurological con<strong>di</strong>tions such asHuntington’s <strong>di</strong>sease, myasthenia gravis, and amyotrophic <strong>la</strong>teralsc<strong>le</strong>rosis. Because the study was service-based, the presentrate probably underestimates the con<strong>di</strong>tion. The demographicand clinical features of the identified patients are of limitedvalue because of the small number. However, the average ageat onset in sixth decade, the fema<strong>le</strong> preponderance, and thehigher frequency of AEO in the setting of dystonia are consistentwith the fin<strong>di</strong>ngs of the clinical series.Several reports have emphasized the presence of AEO in<strong>parkinson</strong>ian syndrome [1, 5], mainly PSP, but detai<strong>le</strong>ddescription with particu<strong>la</strong>r reference to the re<strong>la</strong>tionshipsbetween anti<strong>parkinson</strong>ian me<strong>di</strong>cations, especially <strong>le</strong>vodopa,and AEO was <strong>la</strong>cking in most cases. In the present series, AEOdeveloped following the onset of <strong>parkinson</strong>ian syndrome andthe introduction of anti<strong>parkinson</strong>ian therapy inclu<strong>di</strong>ng <strong>le</strong>vodopain 7 of 9 patients. This raises the possibility of a causalre<strong>la</strong>tionship between <strong>le</strong>vodopa and AEO. Supporting thisview, we report two patients in whom AEO worsened afterincreasing <strong>le</strong>vodopa dosage or acute APO chal<strong>le</strong>nge and <strong>di</strong>sappearedfollowing <strong>le</strong>vodopa <strong>di</strong>scontinuation.Few data are avai<strong>la</strong>b<strong>le</strong> on the response of iso<strong>la</strong>ted or dystonia-associatedAEO to drugs interfering with dopaminetransmission. Dewey and Marangore [8] reported two patientswith iso<strong>la</strong>ted AEO and favorab<strong>le</strong> response to <strong>le</strong>vodopa, but theissue of psychogenicity was not comp<strong>le</strong>tely excluded. Werecently described a patient developing iso<strong>la</strong>ted AEO followingprolonged exposure to flunarizine, a drug known to induceextrapyramidal reactions [4]. Sustained remission when thedrug was stopped was in favor of a causal re<strong>la</strong>tionshipbetween iso<strong>la</strong>ted scAEO and flunarizine [4]. Although AEOdeveloping in the context of a <strong>parkinson</strong>ian syndrome wasknown to exist before initiation of <strong>le</strong>vodopa therapy [1], ourobservations are ground for thinking that the con<strong>di</strong>tion may beeither <strong>di</strong>sease- or drug-re<strong>la</strong>ted. The possibility of mo<strong>di</strong>fyingdopaminergic treatment should always be considered whendealing with AEO in patients with <strong>parkinson</strong>ism. If changes inthe drug regimen do not have any benefit, pretarsal botulinumtoxin treatment may be tried [4].References1. Boghen D (1999) Apraxia of eyelid opening: a review.Neurology 48:1491–15032. Aramideh M, Ongerboer de Visser BW et al (1994).E<strong>le</strong>ctromyographic features of <strong>le</strong>vator palpebrae su<strong>per</strong>iorisand orbicu<strong>la</strong>ris oculi musc<strong>le</strong>s in b<strong>le</strong>pharospasm. Brain117:27–383. Elston JS (1992) A new variant of b<strong>le</strong>pharospasm. J NeurolNeurosurg Psychiatry 55:369–3714. Defazio G, Livrea P, Lamberti R et al (1998) Iso<strong>la</strong>ted socal<strong>le</strong>dapraxia of eyelid opening: report of 10 cases and areview of the literature. Eur Neurol 39:204–2105. Lepore FE, Duvoisin RC (1985) Apraxia of eyelid opening: aninvoluntary <strong>le</strong>vator inhibition. Neurology 35:423–4276. Stacy M, Jankovic J (1992) Differential <strong>di</strong>agnosis ofParkinson’s <strong>di</strong>sease and the <strong>parkinson</strong>ism plus syndrome.Neurol Clin 10[Suppl 2]:341–3597. Defazio G, De Mari M, De Salvia R et al (1999) “Apraxia ofeyelid opening” induced by <strong>le</strong>vodopa and apomorphine inatypical <strong>parkinson</strong>ism (possib<strong>le</strong> progressive supranuc<strong>le</strong>arpalsy): a case report. Clin Neuropharmacol 22:292–2948. Dewey RB, Marangore DM (1994) Iso<strong>la</strong>ted eyelid openingapraxia: report of a new <strong>le</strong>vodopa responsive syndrome.Neurology 44:1752–1754


Neurol Sci (2002) 23:S83–S84 © Springer-Ver<strong>la</strong>g 2002Amanta<strong>di</strong>ne in Huntington’s <strong>di</strong>sease:open-<strong>la</strong>bel video-blinded studyC. Lucetti 1 • G. Gambaccini 1 • S. Bernar<strong>di</strong>ni 1G. Dell’Agnello 1 • L. Petrozzi 1 • G. Rossi 2U. Bonuccelli 1 ()1 Department of Neuroscience, University of Pisa, Pisa, Italy2 Epidemiology and Biostatistics Unit, Institute of ClinicalPhysiology, CNR, Pisa, ItalyAbstract Huntington’s <strong>di</strong>sease (HD) is characterized bychorea, cognitive and behavioral changes. Amanta<strong>di</strong>ne, anon-competitive NMDA receptor antagonist, has shown anantidyskinetic effect on <strong>le</strong>vodopa-induced dyskinesias,which are known to have strict pathogenetic analogies withchoreic hy<strong>per</strong>kinesias. The antidyskinetic efficacy of amanta<strong>di</strong>neand its effects on cognitive and behavioural symptomswere evaluated. Eight HD patients received oral amanta<strong>di</strong>ne(100 mg tid) unblinded for a 1-year <strong>per</strong>iod. A significantreduction of dyskinesias was reported (p


S84Interrater agreement between “live” and “video” raterswas 0.78 (“excel<strong>le</strong>nt”agreement beyond the range of chance)(5). No significant changes were observed in the score ofboth neuropsychological tests and psychiatric rating sca<strong>le</strong>safter 6 and 12 months of amanta<strong>di</strong>ne therapy.DiscussionThe results of this open-<strong>la</strong>bel study support the antidyskineticeffect of amanta<strong>di</strong>ne noted by others [5] in the past and,recently, by Verhagen et al. [6] and Bonuccelli et al. [7]. Inrecent years, there has been a growing interest in understan<strong>di</strong>ngthe effect of antiglutamatergic treatments on motor<strong>di</strong>sturbances of HD patients and riluzo<strong>le</strong> and remacemideseem to have some benefit in choreic dyskinesias [8, 9]. Themechanism underlying the antidyskinetic action of amanta<strong>di</strong>neremains elusive. As it was demonstrated that amanta<strong>di</strong>nebrain <strong>le</strong>vels achieved with therapeutic doses are sufficientto block NMDA receptors, we can hypothesize thatthese antiglutamatergic pro<strong>per</strong>ties of amanta<strong>di</strong>ne may wel<strong>le</strong>xp<strong>la</strong>in the effect in choreic hy<strong>per</strong>kinesias. The improvementof dyskinesias might result from a pharmacologicalreduction of glutamatergic transmission preferentially in thetha<strong>la</strong>mocortical pathways as opposed to the other glutamatergicpathway in the subtha<strong>la</strong>mic-pallidal circuits. Thesymptomatic effect of amanta<strong>di</strong>ne in HD patients corroboratethe ro<strong>le</strong> of basal ganglia glutamatergic hy<strong>per</strong>function inthe pathophysiology of choreic dyskinesias.References1. Reiner A, Albin RL, Anderson KD et al (1988) Differentialloss of striatal projection neurons in Huntington’s <strong>di</strong>sease.Proc Natl Acad Sci USA 5:5733–57372. Verhagen Metman L, Del Dotto P, van den Munckof et al(1998) Amanta<strong>di</strong>ne as treatment for dyskinesias and motorfluctuations in Parkinson’s <strong>di</strong>sease. Neurology 5:1323–13263. Del Dotto P, Pavese N, Gambaccini G et al (2001) Intravenousamanta<strong>di</strong>ne improves <strong>le</strong>vodopa-induced dyskinesias: an acutedoub<strong>le</strong>-blind p<strong>la</strong>cebo-<strong>contro</strong>l<strong>le</strong>d study. Mov Disord16(3):515–5204. F<strong>le</strong>iss JL (1981) The measurement of inter-rater agreement.In: F<strong>le</strong>iss JL (ed) Statistical methods for rates and proportions.Wi<strong>le</strong>y, New York, pp 212–2365. Gray MW, Herzberg L, Lerman JA et al (1975) Amanta<strong>di</strong>ne inchorea. Lancet 2(7925):132–1336. Verhagen L, Morris M, Farmer C et al (2001) A doub<strong>le</strong>-blind,p<strong>la</strong>cebo-<strong>contro</strong>l<strong>le</strong>d crossover study of the effect of amanta<strong>di</strong>neon chorea in Huntington’s <strong>di</strong>sease. Neurology 56[Suppl3]:A368 (abstract)7. Bonuccelli U, Lucetti C, Gambaccini G et al (2000)Antidyskinetic effect of intravenous amanta<strong>di</strong>ne inHuntington’s <strong>di</strong>sease. Neurology 54[7 Suppl 3]:P02.064(abstract)8. Rosas HD, Koroshets WJ, Jenkins BG et al (1999) Riluzo<strong>le</strong>therapy in Huntington’s <strong>di</strong>sease. Mov Disord 14:326–3309. Kieburtz K, Feigin A, McDermott M et al (1996) A <strong>contro</strong>l<strong>le</strong>dtrial of remacemide hydrochloride in Huntington’s <strong>di</strong>sease.Mov Disord 11:273–277


Neurol Sci (2002) 23:S85–S86 © Springer-Ver<strong>la</strong>g 2002Parkinson’s <strong>di</strong>sease and reproductivelife eventsE. Martignoni 1,4 () • R.E. Nappi 2 • A. Citterio 1D. Ca<strong>la</strong>ndrel<strong>la</strong> 3 • E. Corengia 1 • A. Fignon 2R. Zangaglia 1 • G. Riboldazzi 3 • C. Pacchetti 1G. Nappi 51Movement Disorders Centre, IRCCS C. Mon<strong>di</strong>no, Institute ofNeurology, Pavia, Italy2Obstetrics and Gynecology Department, IRCCS Policlinico SanMatteo, Pavia, Italy3University of Insubria, Varese, Italy4University A. Avogadro, Novara, Italy5University La Sapienza, Rome, ItalyAbstract Onset, progression and duration of Parkinson’s<strong>di</strong>sease (PD) seem to be simi<strong>la</strong>r in men and women but gender<strong>di</strong>fferences have been suggested concerning clinica<strong>la</strong>spects, such as more severe <strong>di</strong>sease in men and more dyskinesiain women. Taking into account the multip<strong>le</strong> influencesof sex hormones, estrogens in particu<strong>la</strong>r, on basal gangliafunction, the present work compared the characteristicsof reproductive events in PD subjects and in healthywomen, with regard to onset and clinical aspects of the <strong>di</strong>seasewith respect to the mi<strong>le</strong>stones of reproductive life. Atotal of 150 PD women and 200 healthy women matched forage were interviewed about reproductive life and <strong>di</strong>seasecharacteristics (if patients). As a group, the women with PDhad menarche <strong>la</strong>ter than the <strong>contro</strong>ls, but in the normalrange. Menopause was simi<strong>la</strong>r to the <strong>contro</strong>ls for time, type(natural) and onset (slow), but with <strong>le</strong>ss hormonal therapies.Women with PD had fewer children, whi<strong>le</strong> breast fee<strong>di</strong>ngand gynecological <strong>di</strong>seases were comparab<strong>le</strong> to <strong>contro</strong>ls.The characteristics of menses were simi<strong>la</strong>r as far as dysmenorrheaand premenstrual syndrome (PMS). The womenwith PD onset before menopause had a longer <strong>di</strong>sease duration,with a more frequent fluctuating stage, and longertreatment with both <strong>le</strong>vodopa and dopamine agonists. Theyhad more dysmenorrhea and PMS when compared withwomen with PD onset after menopause and <strong>contro</strong>ls.Parkinson’s <strong>di</strong>sease (PD) is a neurodegenerative <strong>di</strong>sorderoccurring in both men and women, although most stu<strong>di</strong>es findPD more common in men. Gender <strong>di</strong>fferences have been suggestedconcerning clinical aspects, such as more severe <strong>di</strong>seasein men and more dyskinesia in women [1], and behaviora<strong>la</strong>nd psychiatric manifestations, such as wandering, verba<strong>la</strong>nd physical abusiveness, hallucinations and delusions [2].There is substantial evidence in<strong>di</strong>cating that estrogen canmodu<strong>la</strong>te dopaminergic activity in the nigrostriatal system.One study suggested that estrogen has an antidopaminergiceffect on motor function in postmenopausal women with PD[3]. Another study in<strong>di</strong>cated that hormone rep<strong>la</strong>cement therapywas associated with <strong>le</strong>ss severe <strong>parkinson</strong>ian symptoms inpostmenopausal women [4], and low-dose estrogen reducedmotor <strong>di</strong>sability in postmenopausal women with PD associatedwith motor fluctuations [5]. In a recent report, no significantdopaminergic effect of estra<strong>di</strong>ol was demonstrated, but progesteroneseemed to have an antidopaminergic effect [6]. In anotherreport, no significant corre<strong>la</strong>tion between the objective orsubjective measures of <strong>parkinson</strong>ism and estrogen and progesterone<strong>le</strong>vels was demonstrated [7]. The aim of this study wasto investigate the possib<strong>le</strong> re<strong>la</strong>tionship between reproductivelife events and the onset of PD, to verify the possib<strong>le</strong> re<strong>la</strong>tionshipbetween <strong>parkinson</strong>ism and the hormonal milieu inwomen. With this purpose, 150 women with PD <strong>di</strong>agnosedaccor<strong>di</strong>ng to the Brain Bank criteria and atten<strong>di</strong>ng the centresfor PD of the Neurological Services of the Ospeda<strong>le</strong> <strong>di</strong> Circoloof Varese and of the IRCCS C. Mon<strong>di</strong>no of Pavia were recruitedfor the study together with 200 women of postmenopausa<strong>la</strong>ge atten<strong>di</strong>ng the Menopause Unit of the Department ofObstetrics and Gynecology of the University of Pavia.The PD women were submitted to a face to face or phoneinterview, designed to investigate the possib<strong>le</strong> re<strong>la</strong>tionshipbetween the <strong>di</strong>sease and reproductive function. The interviewwas prepared with the col<strong>la</strong>boration of gynaecologists, andwas <strong>di</strong>vided into 3 sections: the first one investigated age andeducation <strong>le</strong>vel, age at onset of PD, duration and phase of <strong>di</strong>sease,and pharmacological therapy; the second section investigatedreproductive function, particu<strong>la</strong>rly the characteristics ofmenses, pregnancy, oral contraceptive use and menopause;finally, the <strong>la</strong>st one investigated the re<strong>la</strong>tionship between neurologicalsymptoms and menopause. All the data wereobtained accor<strong>di</strong>ng to the current ru<strong>le</strong>s on confidentiality.The data were col<strong>le</strong>cted into a database (Excel ’97,Microsoft) and analysed with SPSS version 10.0. The qualitativevariab<strong>le</strong>s were compared with chi-square test andquantitative variab<strong>le</strong>s with ANOVA.At first the comparison between the <strong>contro</strong>l and patientgroups was made, followed by the analysis of the patientdata accor<strong>di</strong>ng to the onset before or after menopause(inten<strong>di</strong>ng absence of menstrual b<strong>le</strong>e<strong>di</strong>ng >6 months).Patients with PD were between 42 and 85 years, with a meanage of 65.54 years, whi<strong>le</strong> the mean age of <strong>contro</strong>ls was 57.61years, with a range from 44 to 80. As a group, the womenwith PD had menarche <strong>la</strong>ter than the <strong>contro</strong>ls, although in thenormal range. Menopause in PD patients was simi<strong>la</strong>r to thatin the <strong>contro</strong>ls for time, type (natural in more than 70% of thesubjects), and onset (slow in more than 50% of the women),but with <strong>le</strong>ss hormonal therapies. Women with PD had hadfewer children, but the preva<strong>le</strong>nce of breast fee<strong>di</strong>ng andgynecological <strong>di</strong>seases was simi<strong>la</strong>r to that in the <strong>contro</strong>lwomen. The characteristics of menses were simi<strong>la</strong>r as far asdysmenorrhea and premenstrual syndrome (PMS).


S86In 25.3% of women, the onset of PD was beforemenopause. They had a longer <strong>di</strong>sease duration, with a morefrequent fluctuating stage, and longer treatment with both<strong>le</strong>vodopa and dopamine agonists. They had more dysmenorrheaand PMS when compared with women with PD onsetafter menopause and with <strong>contro</strong>ls.DiscussionThis study compares for the first time the reproductive lifeevents in women with PD and <strong>contro</strong>ls, simi<strong>la</strong>r for age andgeographic and cultural environment. The data agree with anapparently normal step of ferti<strong>le</strong> life span in women with PDonset after menopause, even though PD women had fewerchildren that <strong>di</strong>d the <strong>contro</strong>ls.In PD women with onset before menopause, the re<strong>la</strong>tionshipwith reproductive life events was more evident. Infact, they show more premenstrual symptoms and dysmenorrheaand a premenstrual worsening of motor symptoms.This <strong>la</strong>st observation agrees with a previous report [8], butit seems not specific for the <strong>di</strong>sease as it happenes also invarious other neurological pathologies such as migraine,<strong>per</strong>ipheral neuropathies, depression and spasmo<strong>di</strong>c torticollis[9].References1. Lyons KE, Hubb<strong>le</strong> JP, Troster AI et al (1998) Gender <strong>di</strong>fferencesin Parkinson’s <strong>di</strong>sease. Clin Neuropharmacol 21:118–1212. Fernandez HH, Lapane KL, Ott BR, Friedman JH (2000)Gender <strong>di</strong>fferences in the frequency and treatment of behaviorprob<strong>le</strong>ms in Parkinson’s <strong>di</strong>sease. SAGE Study Group.Systematic Assessment and Geriatric Drug Use viaEpidemiology. Mov Disord 15:490–4963. Kol<strong>le</strong>r WC, Barr A, Biary N (1982) Estrogen treatment of dyskinetic<strong>di</strong>sorders. Neurology 32:547–5494. Saunders-Pullman R, Gordon-Elliot J, Parides M et al (1999)The effect of estrogen rep<strong>la</strong>cement on early Parkinson’s <strong>di</strong>sease.Neurology 52:1417–14215. Tsang KL, Ho SL, Lo SK (2000) Estrogen improves motor<strong>di</strong>sability in <strong>parkinson</strong>ian postmenopausal women with motorfluctuations. Neurology 54:2292–22986. Strijks E, Kremer JA, Horstink MW (1999) Effects of fema<strong>le</strong>sex steroids on Parkinson’s <strong>di</strong>sease in postmenopausalwomen. Clin Neuropharmacol 22:93–977. Kompoliti K, Comel<strong>la</strong> CL, Jaglin JA et al (2000) Menstrualre<strong>la</strong>tedchanges in motoric function in women withParkinson’s <strong>di</strong>sease. Neurology 55:1572–15748. Quinn ND, Marsden CD (1986) Menstrual-re<strong>la</strong>ted fluctuationsin Parkinson’s <strong>di</strong>sease. Mov Disord 1:85–879. Tarsy D, Thulin PC, Herzog AG (2001) Spasmo<strong>di</strong>c torticollisand reproductive function in women. Parkinsonism andRe<strong>la</strong>ted Disorders


Neurol Sci (2002) 23:S87–S88 © Springer-Ver<strong>la</strong>g 2002Magnetic resonance re<strong>la</strong>xometryin Parkinson’s <strong>di</strong>seaseF. Mon<strong>di</strong>no 1 () • P. Filippi 2 • U. Maglio<strong>la</strong> 2S. Duca 31 Department of Neurology, S. Croce Hospital, Cuneo, Italy2 Department of Neurology, S. Luigi Hospital, Turin, Italy3 Neurora<strong>di</strong>ology, Koelliker Hospital, Turin, ItalyAbstract A central ro<strong>le</strong> of iron in the pathogenesis of i<strong>di</strong>opathicParkinson’s <strong>di</strong>sease (PD), due to its increase in substantianigra pars compacta dopaminergic neurons and itscapacity to enhance production of toxin reactive oxygen ra<strong>di</strong>cals,has been <strong>di</strong>scussed for many years. Nuc<strong>le</strong>ar magneticresonance (NMR) re<strong>la</strong>xation is considered an objective andnoninvasive method of measuring regional iron concentrations.By means of this technique we investigated both <strong>contro</strong>lsand PD patients.The brain shares with other organs the need for a constantand rea<strong>di</strong>ly avai<strong>la</strong>b<strong>le</strong> supply of iron and has a simi<strong>la</strong>r arrayof proteins necessary for iron transport, storage and regu<strong>la</strong>tion.However, unlike other organs, the brain p<strong>la</strong>ces demandson iron avai<strong>la</strong>bility that are regional, cellu<strong>la</strong>r and age sensitiveand has developed mechanisms to maintain the cellu<strong>la</strong>riron homeostasis that is crucial for the viability of neurons.In a number of common neurodegenerative <strong>di</strong>sorders,there appears to be a loss of these homeostatic mechanismswith following excess accumu<strong>la</strong>tion of iron in the brain.Presently the mechanisms involved in the <strong>di</strong>sturbances ofiron metabolism in PD remain obscure, but numerous stu<strong>di</strong>esusing a variety of analytic techniques demonstrated thatiron <strong>le</strong>vels are increased in PD, primarily within the substantianigra pars compacta. Laser microprobe (LAMMA) stu<strong>di</strong>escapab<strong>le</strong> of provi<strong>di</strong>ng subcellu<strong>la</strong>r localization in<strong>di</strong>cate thatiron normally accumu<strong>la</strong>tes within neurome<strong>la</strong>nin granu<strong>le</strong>s ofdopaminergic neurons in the substatia nigra and that iron <strong>le</strong>velswithin neurome<strong>la</strong>nin granu<strong>le</strong>s are significantly increase<strong>di</strong>n PD [1]. Information <strong>per</strong>taining to the avai<strong>la</strong>bility of ferritinin the brain of PD patients would be useful, as ironbound to ferritin is re<strong>la</strong>tively unreactive and unlikely toinduce tissue damage. There are conflicting reports in the literature,with in<strong>di</strong>cations that ferritin is both increased [2] anddecreased [3] in PD. These <strong>di</strong>fferences probably ref<strong>le</strong>ct <strong>di</strong>fferencesin the isoform of ferritin that was stu<strong>di</strong>ed. Morerecently, no increase in neuronal isoferritin was detectedwhen specific brain isoforms of ferritin were evaluated [4].Since the introduction in clinical practice of NMR at 1.5tes<strong>la</strong>, a basal ganglia hypointensity has been described inspin echo (SE) T2-weighted images. Drayer et al. [5] andRut<strong>le</strong>dge et al. [6] observed that in T2-weighted imagesobtained at high field intensity MRI, the <strong>di</strong>stribution of lowsignal intensity in the basal ganglia and other areas of thebrain corresponded roughly with the ferric iron (Fe +++ ) <strong>di</strong>stributiondemonstrated by Perls’ stain in brain sections [6].Iron deposits would create local inhomogeneities in the magneticfield which, in turn, would result in loss of the T2 signal.So, if T2 hypointensity of extrapyramidal nuc<strong>le</strong>i is dueto iron presence in these regions, we expect a further reductionin signal intensity when there is iron accumu<strong>la</strong>tion.NMR stu<strong>di</strong>es have reported several patterns of decreasedsignal intensity in the basal ganglia and in substantia nigra ofpatients with PD, using a visual analysis of T2-weighte<strong>di</strong>mages [6, 7]. The estimation of T2 re<strong>la</strong>xation time might bemore reliab<strong>le</strong> than visual analysis in <strong>di</strong>fferentiating healthyvolunteer subjects from PD patients. The aim of this studywas to evaluate the T2 re<strong>la</strong>xation time of the extrapyramidalnuc<strong>le</strong>i in PD patients and in healthy subjects.For this study, 25 PD patients (age range, 39–76 years;mean age, 63 years) were se<strong>le</strong>cted. PD <strong>di</strong>agnosis was based onclinical criteria inclu<strong>di</strong>ng the presence of bradykinesia withtremor or rigi<strong>di</strong>ty or postural instability, asymmetric symptomsonset, good response to L-dopa treatment, and absence ofsigns or symptoms of atypical or secondary <strong>parkinson</strong>ism.They had a mean <strong>di</strong>sease duration of 8.4±5.5 years and wereevaluated accor<strong>di</strong>ng to subscores I and III of UnifiedParkinson’s Disease Rating Sca<strong>le</strong> (UPDRS) showing a cognitive-<strong>per</strong>formancesscore of 4.0±2.6 and a motor examinationscore of 28.92±13.55. All patients were taking anti-PD me<strong>di</strong>cations:L-dopa or an oral dopaminergic agonist or both.Twenty-seven age-mached healthy <strong>contro</strong>l participants(age range, 46–90 years; mean age, 66 years) were also stu<strong>di</strong>ed.Se<strong>le</strong>ction criteria were no history of psychiatric or neurologic<strong>di</strong>sease, no abnormal signs at general me<strong>di</strong>cal andneurologic examinations, and a previous normal neuroimaging(CT or NMR).Informed consent was obtained from each <strong>per</strong>son.Subjects were stu<strong>di</strong>ed with an MR Gyroscan (Philips)scanner o<strong>per</strong>ating at 1.5 T. After conventional sagittal T1-weighted scout images were acquired, a trial set of axialspin-echo sequences with a short TR, 600/20 (TR/TE) and along TR, 2500/20, 80 (TR/first-echo TE, second-echo TE)were were taken paral<strong>le</strong>l to the orbitomeatal p<strong>la</strong>ne, as determinedon the sagittal section.The re<strong>la</strong>xometric study was <strong>per</strong>formed by using two 5-mm sing<strong>le</strong> sections: one inclu<strong>di</strong>ng the head of caudatenuc<strong>le</strong>us, putamen and globus pallidus, the other inclu<strong>di</strong>ngthe midbrain at the <strong>le</strong>vel of substantia nigra and nuc<strong>le</strong>usruber. A 5-mm section thickness was chosen as the bestcompromise to maintain good signal-to-noise ratios and reasonab<strong>le</strong>imaging times (which cannot be prolonged toomuch in patients with PD).At each of these <strong>le</strong>vels, a multiecho sequence (eightechoes; echo times 30–240 ms) was applied, and mean T2


S88values for the following regions of interest (ROIs) were calcu<strong>la</strong>tedon each hemisphere: frontal cortex, temporal cortex,frontal white matter, head of caudate, globus pallidus, anteriorand posterior putamen, nuc<strong>le</strong>us ruber, substantia nigrapars compacta and pars reticu<strong>la</strong>ta. ROIs were irregu<strong>la</strong>r andwere drawn by hand on the screen around the outline of thecorrespon<strong>di</strong>ng structures. Frontal and temporal cortex andfrontal white matter ROIs were p<strong>la</strong>ced on the p<strong>la</strong>ne at thebasal ganglia <strong>le</strong>vel. In each subject, T2 values derived fromthe correspon<strong>di</strong>ng ROIs on the right and <strong>le</strong>ft brain hemisphereswere poo<strong>le</strong>d. T2 re<strong>la</strong>xation time measurements were<strong>per</strong>formed by work station “Easy Vision”.Using an unpaired the two-tai<strong>le</strong>d t test, no significant <strong>di</strong>fferencebetween groups was found in T2 values on temporalcortex, frontal white matter, head of caudate, globus pallidus,anterior and posterior putamen, red nuc<strong>le</strong>us and substantianigra (neither pars compacta nor pars reticu<strong>la</strong>ta). The only significant<strong>di</strong>fference was found in T2 values of the frontal cortexthat were significantly shorter in the PD patient group onboth right (p=0.0089) and <strong>le</strong>ft (p=0.0367) sides. Moreover,both groups showed a significant decline of T2 signal with agein frontal cortex (p


Neurol Sci (2002) 23:S89–S90 © Springer-Ver<strong>la</strong>g 2002Quetiapine versus clozapine:a preliminary report of comparativeeffects on dopaminergic psychosis inpatients with Parkinson’s <strong>di</strong>seaseL. Morgante 1 () • A. Epifanio 1 • E. Spina 2A.E. Di Rosa 1 • M. Zappia 3 • G. Basi<strong>le</strong> 4P. La Spina 1 • A. Quattrone 31 Department of Neuroscience, University of Messina, PoliclinicoUniversitario, Via Conso<strong>la</strong>re Va<strong>le</strong>ria, I-98125 Messina, Italy2 Institute of Pharmacology, University of Messina, Italy3 Department of Neuroscience, University of Catanzaro, Catanzaro,Italy4 Department of Geriatric Me<strong>di</strong>cine, University of Messina,Messina, ItalyAbstract This study investigated the efficacy and safety ofquetiapine versus clozapine in <strong>parkinson</strong>ian patients withdopaminergic psychosis. All patients fulfilling the inclusioncriteria were randomly assigned to receive either quetiapine orclozapine. The duration of the trial was 12 weeks. The severityof psychosis was assessed using the BPRS and the ClinicalGlobal Impression Sca<strong>le</strong>-Severity subsca<strong>le</strong> (CGI-S). TheUPDRS III was used to monitor the progression of PD duringthe study <strong>per</strong>iod. Twenty patients, 10 on clozapine, and 10 onquetiapine, comp<strong>le</strong>ted the study. The psychopathological state,as assessed by the BPRS and by the CGI-S, improved significantly(p


S90Tab<strong>le</strong> 1 Demographic and clinical features of the 20 patients comp<strong>le</strong>ting the study. Values are means (SD). Differences between treatmentgroups were not significantQuetiapineClozapine(n = 10) (n = 10)Dosage, mg/day 90.0 (51.7) 27.5 (14.2)Age, years 69 (12) 68 (11)Duration of illness, months 112 (59) 121 (60)Hoehn & Yahr stage 3.1 (0.5) 3.2 (0.6)L-Dopa dosage, mg/day 675 (188) 705 (123)BPRS totalBaseline 37.7 (6.9) 38.2 (6.2)Endpoint 28.4 (5.2)** 27.5 (4.7)**CGI-SBaseline 3.6 (0.7) 3.8 (0.8)Endpoint 2.1 (0.6)** 1.9 (0.6)**UPDRSIIIBaseline 53.3 (9.7) 57.8 (11.2)Endpoint 54.9 (11.5) 55.7 (14.9)*AIMSBaseline 7.9 (2.2) 7.7 (2.3)Endpoint 6.3 (1.3)* 5.9 (1.6)*** Statistically significant <strong>di</strong>fference between endpoint and baseline p


Neurol Sci (2002) 23:S91–S94 © Springer-Ver<strong>la</strong>g 2002Incidence of RBD and hallucinationin patients affected by Parkinson’s<strong>di</strong>sease: 8-year follow-upM. Onofrj 1 () • A. Thomas 1 • G. D’Andreamatteo 1D. Iacono 1 • A.L. Luciano 1 • A. Di Rollo 1R. Di Mascio 2 • E. Ballone 2 • A. Di Iorio 31 Department of Oncology and Neuroscience; Neurophysiopathology,G. D’Annunzio University, Ospeda<strong>le</strong> Civi<strong>le</strong> <strong>di</strong> Pescara,Via Fonte Romana, I-65124 Pescara, Italy2 Institute of Statistics, G. D’Annunzio University, Chieti, Italy3 Department of Me<strong>di</strong>cine and Ageing, Geriatric Unit, Via deiVestini, Chieti, Italy;Abstract We describe the 8-years follow-up of 80 patientsaffected by i<strong>di</strong>opathic, L-dopa-responsive Parkinson’s <strong>di</strong>sease.All patients were evaluated at baseline and during thefollow-up with visual evoked potential, P300 event re<strong>la</strong>tedpotentials and polysomnography. The patients and their re<strong>la</strong>tivescompi<strong>le</strong>d s<strong>le</strong>ep and hallucination questionnaires.Statistical analysis was <strong>per</strong>formed to evaluate if visua<strong>la</strong>bnormalities, abnormal P300 recor<strong>di</strong>ngs or s<strong>le</strong>ep <strong>di</strong>sturbanceswere linked to the development and hallucinations.Our results show that abnormal vision and abnormal P300<strong>di</strong>d not corre<strong>la</strong>te with the incidence of hallucinations.However, the presence of REM s<strong>le</strong>ep behavioral <strong>di</strong>sorder(RBD) was significantly re<strong>la</strong>ted to the development of hallucinations,independently of age, gender or duration of <strong>di</strong>seasebut dependent on the amount of dopaminoagonist treatment.Many stu<strong>di</strong>es have reported visual abnormalities with e<strong>le</strong>ctroretinogramand visual evoked potential (VEP) alterations inParkinson’s <strong>di</strong>sease (PD) [1–3], showing that visual abnormalitiesare due to dopamine deficiency and re<strong>la</strong>ted to the <strong>per</strong>ceptionof the spatial frequency of stimuli [4]. Oddly enough, withthe exception of one study based on color and contrast <strong>per</strong>ception[5], visual abnormalities were not stu<strong>di</strong>ed in re<strong>la</strong>tion to theoccurrence of the well known “visual” complication of PD, i.e.visual hallucinations. Visual hallucinations are attributed toabnormalities of the ascen<strong>di</strong>ng cholinergic and serotoninergicbrainstem (and tha<strong>la</strong>mic) pathways involved in the <strong>contro</strong>l ofs<strong>le</strong>ep-waking state, or to the defective visual processingaccompanied by abnormal cortical re<strong>le</strong>ase phenomenon [6].The renovated approach to s<strong>le</strong>ep stu<strong>di</strong>es in PD and<strong>parkinson</strong>ism has evidenced that a peculiar <strong>di</strong>sturbance ofrapid eye movement (REM) s<strong>le</strong>ep phase, consisting of theloss of normal musc<strong>le</strong> inhibition during REM s<strong>le</strong>ep andenacting of dreams, i.e. REM s<strong>le</strong>ep behavior <strong>di</strong>sorder(RBD), is present in i<strong>di</strong>opathic PD [7], in some <strong>parkinson</strong>ismstates and in Lewy body <strong>di</strong>sease (LBD). The presence ofRBD has been associated with the occurrence of daytimehallucinations [8]. In order to understand whether hallucinationsin PD are re<strong>la</strong>ted to visual or s<strong>le</strong>ep <strong>di</strong>sturbances, we<strong>per</strong>formed an 8-year follow-up of 80 patients affected byi<strong>di</strong>opathic, L-dopa responsive PD. All patients were evaluatedfor VEP alterations and contrast sensitivity, underwentvisual P3 event re<strong>la</strong>ted potential (ERP) recor<strong>di</strong>ngs andpolysomnography, and responded to s<strong>le</strong>ep and hallucinationsquestionnaires, in order to assess the possib<strong>le</strong> corre<strong>la</strong>tionwith the onset or severity of hallucinations.Consecutive patients with probab<strong>le</strong> PD and their caregiverswho were followed in the movement <strong>di</strong>sorders outpatientsoffices were invited to participate in the study. Initialse<strong>le</strong>ctions included 166 patients. At the end of the study,eight years after admission, 80 patients were considered tobe affected by i<strong>di</strong>opathic PD because, despite occurrence ofwearing-off or simi<strong>la</strong>r phenomenon, they were (1) still ex<strong>per</strong>iencinga benefit from dopaminomimetic therapies that hadbeen increased by 1.5- to 3-times the amount administered atadmission, (2) cognitive and behavioral functions were notaltered during dopaminomimetic treatment, and (3) tremorwas a consistent, although variab<strong>le</strong>, feature in off <strong>per</strong>iods.E<strong>le</strong>ctroretinography (ERG) and polysomnography were<strong>per</strong>formed, and VEPs, contrast sensitivity and visual P300ERPs were recorded accor<strong>di</strong>ng to methods described indetail elsewhere [4, 9–11]. All e<strong>le</strong>ctrophysiological recor<strong>di</strong>ngswere <strong>per</strong>formed at admission, before any treatment regimenwas initiated in 52 patients, and 24 hours after the <strong>la</strong>stL-dopa, dopaminoagonist or anticholinergic treatment in theother patients. Recor<strong>di</strong>ngs were repeated with the samemethod at the end of the study. Criteria for VEP abnormalitywere set by the mean <strong>la</strong>tency + 2 standard deviations (SD),for contrast sensitivity abnormality on 2 dB loss of 2–5cyc<strong>le</strong>s <strong>per</strong> degree (cpd) spatial frequency, for ERG abnormalityon the ratio below 50% of 1 vs 3 cpd evoked ERG, forvisual P3 ERP abnormality on the mean <strong>la</strong>tency + 1.5 SD. Atadmission, patients and family members were interviewedon their s<strong>le</strong>ep behavior and s<strong>le</strong>ep history using a simp<strong>le</strong> s<strong>le</strong>epquestionnaire, respecting the minimal ICSD (1997) criteriafor RBD in agreement with the spouse’s report [8], develope<strong>di</strong>n our center: the questionnaire is proposed to patientsand family members and investigates items such as qualityof s<strong>le</strong>ep, me<strong>di</strong>cation, vocalization, movements during s<strong>le</strong>ep,nightmares, dream enactment, hallucinations when awakingfrom nightmares and frequency of movements during s<strong>le</strong>ep.Patients and family members were asked with a questionnaire,part of the present state examination [12], about hallucinationsand, if present, about the kind of hallucinations.Questionnaires were administered once every year untilthe end of the study. Polysomnography in night s<strong>le</strong>ep was<strong>per</strong>formed whenever the s<strong>le</strong>ep questionnaire suggested RBDand at the end of the study. All patients were evaluated at thebeginning of the study with the Hoehn/Yahr sca<strong>le</strong> [13],UPDRS [14] and MMSE [15].Total daily intakes of L-dopa and dopaminoagonist were


S92compared with the other parameters; for dopaminoagonistsan equiva<strong>le</strong>nt bromocriptine dose was calcu<strong>la</strong>ted as 10 mgbromocriptine = 1 mg <strong>per</strong>golide, 1 mg pramipexolo, 5 mgropiniro<strong>le</strong> or 1.5 mg cabergoline.Systematic <strong>di</strong>fferences between hallucinating and non-hallucinatingpatients were evaluated using χ 2 test, Mantel-Haenzel tests for linear associations and Fisher’s exact test.Factors independently associated with hallucinatory statuswere identified using logistic regression models (CATMODprocedure).We forced in the logistic model two variab<strong>le</strong>s (visua<strong>la</strong>bnormalities and P300), also if the univariated analysisreached a p value greater than 0.10. Our intention was todemonstrate the ro<strong>le</strong> that both visual abnormalities and P300p<strong>la</strong>y on hallucinatory status development. The saturatedlogistic model also contained the variab<strong>le</strong>s age and sex.Adjusted odds ratio (OR) and 95% confidence interval (CI)were calcu<strong>la</strong>ted from the estimated coefficients in the model.To evaluate the hierarchically best model, the con<strong>di</strong>tionalmaximum likelihood ratio (CMLR) was <strong>per</strong>formed, with orwithout interactions between main pre<strong>di</strong>ctors. The Hosmer-Lemeshow test χ 2 (8 df), goodness of fit, was also <strong>per</strong>formed.All analyses were <strong>per</strong>formed using SAS package[16].The RBD questionnaire was validated in comparisonwith polysomnography recor<strong>di</strong>ngs: all patients c<strong>la</strong>ssified ashaving RBD by questionnaire had polysomnographic evidenceof RBD, only 2 patients c<strong>la</strong>ssified as non-RBD by thequestionnaire had 1 or 2 episodes of RBD evidenced bypolysomnography (sensitivity, 100%; specificity, 96.3%).Accor<strong>di</strong>ng to the Hoehn/Yahr sca<strong>le</strong> at baseline, 44patients were at stage 1, 14 patients at stage 1.5, 10 patientsat stage 2, and 12 patients at stage 2.5. After 8 years, the stagingof the same study popu<strong>la</strong>tion was as follows: 20 patientsat stage 2, 14 patients at stage 2.5, 39 patients at stage 3, and7 patients at stage 4.At baseline, visual abnormalities were found in 28 PDpatients (Tab<strong>le</strong> 1). The chronic dopaminergic treatment in“on” state reduced visual abnormalities to normal limits inthe majority of patients and at the end of the study visua<strong>la</strong>bnormalities were observed in “off” and “on” state in only27 patients. Linear regressions of VEP <strong>la</strong>tencies and amplitudesvs. duration and stage of Parkinson’s <strong>di</strong>sease <strong>di</strong>d notreach statistical significance. The preva<strong>le</strong>nce of hallucinationswas below 40% both in patients with and without visua<strong>la</strong>bnormalities (not significant at χ 2 ). A de<strong>la</strong>y of P300 wasrecorded in 2 patients at baseline and in 15 patients during“on” state and 27 patients during “off” state at the 8-year fol-Tab<strong>le</strong> 1 Preva<strong>le</strong>nce of visual and P300 abnormalities, RBD and hallucinations in 80 patients with i<strong>di</strong>opathic L-dopa-responsive Parkinson’s<strong>di</strong>sease. Values are number of patientsBaseline 3 years 6 years 8 yearsOn Off On OffVisionNormal 52 66 67 59 58 53Abnormal 28 14 13 21 22 27P300Normal 78 71 69 59 65 53Abnormal 2 9 11 21 15 27RBD 5 9 23 – 27 –Hallucinations 5 8 19 – 31 –RBD, REM s<strong>le</strong>ep behavioral <strong>di</strong>sorderTab<strong>le</strong> 2 Preva<strong>le</strong>nce of visual and P300 abnormalities and RBD in patients with hallucinations, identified from 80 patients with i<strong>di</strong>opathicParkinson’s <strong>di</strong>sease and followed for 8 years. Value are number of patientsBaseline (n=5) 3 years (n=8) 6 years (n=19) 8 years (n=31)VisionNormal 3 6 11 19Abnormal 2 2 7 12P300Normal 5 7 13 24Abnormal 0 1 5 7RBD 3 8 19 27RBD, REM s<strong>le</strong>ep behavior <strong>di</strong>sorder


S93low-up. P300 de<strong>la</strong>y was <strong>di</strong>stributed equally in patients withand without hallucinations, (NS at χ 2 ). CMLR excludedP300 as a main pre<strong>di</strong>ctor.Throughout the follow-up observation, 27 patients werec<strong>la</strong>ssified to be clinically affected by RBD (questionnaireplus PSG).At the <strong>la</strong>st visit, only 4 patients describing hallucinationswere c<strong>la</strong>ssified among the normal s<strong>le</strong>e<strong>per</strong>s (n=53), whi<strong>le</strong> 27PD patients had RBD and ex<strong>per</strong>ienced hallucinations independentlyof age or gender. There was a higher incidence ofhallucinations in stages 3 and 4 PD patients, not reaching statisticalsignificance (p


S94State: a practical method for gra<strong>di</strong>ng the cognitive state ofpatients for clinician. J Psychiatry Res 12:189–19816. – (2002) SAS 8.1 re<strong>le</strong>ase. SAS Institute Inc, Cary, NC, USA17. Lhermitte J (1929) Sydrome de <strong>la</strong> calotte du pedoncu<strong>le</strong>cérébral. Les troub<strong>le</strong>s psychosensoriels dans <strong>le</strong>s <strong>le</strong>sions dumésocépha<strong>le</strong>. Rev Neurol (Paris) 2:1359–136518. Gel<strong>le</strong>r TJ, Bellur SN (1987) Pedunco<strong>la</strong>r hallucinosis: magneticresonance imaging confirmation of mesencephalic infartionduring life. Ann Neurol 21:602–60419. Ball B (1882) De l’insanité dans <strong>la</strong> paralysie agitante.Encépha<strong>le</strong> 2:22–3220. Parant V (1883) La paralysie agitante examinée comme causede folie: Ann Med Psychol (Paris) 10:45–6321. McKee AC, Levine DN, Kowall NW, Richardson EP (1990)Peduncu<strong>la</strong>r hallucinosis associated with iso<strong>la</strong>ted infarctionof the substantia nigra pars reticu<strong>la</strong>ta. Ann Neurol27:500–50422. Moskovitz C, Moses H, K<strong>la</strong>wans HL (1978) Levodopainducedpsychosis: a kindling phenomenon. Am J Psychiatry135:669–67523. Goetz CG, Vogel C, Tanner CM, Stebbins GT (1998) Earlydopaminergic drug-induced hallucinations in <strong>parkinson</strong>ianpatients. Neurology 51:811–81424. Arnulf I, Bonnet AM, Damier P, Bejjani BP, Seilhean,Derenne JP, Agid Y (2000) Hallucination, REM s<strong>le</strong>ep andParkinson’s <strong>di</strong>sease. Neurology 55:281–288


Neurol Sci (2002) 23:S95–S96 © Springer-Ver<strong>la</strong>g 2002Cerebrospinal fluid <strong>le</strong>vels ofbiomarkers and activity ofacetylcholinesterase (AChE) andbutyrylcholinesterase in AD patientsbefore and after treatment with<strong>di</strong>fferent AChE inhibitorsL. Parnetti 1 () • S. Amici 1 • A. Lanari 1C. Romani 2 • C. Antognelli 2 • N. Andreasen 3L. Minthon 4 • P. Davidsson 5 • H. Pottel 6K. B<strong>le</strong>nnow 5 • V. Gal<strong>la</strong>i 11 Department of Neuroscience, Perugia University, Perugia, Italy2 Department of Genetics and Mo<strong>le</strong>cu<strong>la</strong>r Biology, PerugiaUniversity, Perugia, Italy3 Department of Rehabilitation, Piteå, Sweden4 Department of Psychiatry, Mälmo University Hospital, Mälmo,Sweden5 Department of Neurochemistry, Göteborg University, Göteborg,Sweden6 Innogenetics, Ghent, BelgiumAbstract In order to evaluate the biochemical effects oflong-term treatment with inhibitors of acetylcholinesterase(AChE) in patients with Alzheimer’s <strong>di</strong>sease (AD), we measuredthe activities of AChE and butyrylcholinesterase(BuChe) and the concentrations of β-amyloid (1–42), τ andphosphory<strong>la</strong>ted τ proteins in the cerebrospinal fluid (CSF). Atotal of 91 patients suffering from probab<strong>le</strong> AD of mild tomoderate degree were treated for 6 months with donepezil(n=59), ga<strong>la</strong>ntamine (n=15), rivastigmine (n=10), or p<strong>la</strong>cebo(n=7). AChE activity in CSF was significantly increasedafter treatment with donepezil and ga<strong>la</strong>ntamine; the oppositewas observed in the rivastigmine-treated group. Untreatedpatients <strong>di</strong>d not show any AChE activity variation. BuChE<strong>di</strong>d not show any change in any of the groups stu<strong>di</strong>ed. Meanvalues of β-amyloid(1–42), total τ and phosphory<strong>la</strong>ted τ also<strong>di</strong>d not vary significantly. We conclude that AChE inhibitorsinduce <strong>di</strong>fferent effects on CSF AChE activity, whi<strong>le</strong> otherCSF biomarkers are not significantly affected by treatment.Acetylcholinesterase (AChE) colocalizes with β-amyloid inneuritic p<strong>la</strong>ques and acce<strong>le</strong>rates the assembly of amyloid β-peptides into fibrils deposited in the brain of patients withAlzheimer’s <strong>di</strong>sease (AD). Conversely, the β-amyloid proteinregu<strong>la</strong>tes AChE expression, assembly and glycosy<strong>la</strong>tionin cell cultures, transgenic mice and Alzheimer brain, thuscreating a vicious circ<strong>le</strong> <strong>le</strong>a<strong>di</strong>ng to an increased accumu<strong>la</strong>tionof β-amyloid. AChE inhibitors have been suggested toenhance the re<strong>le</strong>ase of nonamyloidogenic solub<strong>le</strong> derivativesof amyloid precursor proteins (APPs) in vitro and in vivo andpossibly to slow down formation of amyloidogenic compoundsin brain [1–3]. Therefore, inhibition of AChE activitymight influence APP processing and β-amyloid deposition.With respect to the potential influence of AChEinhibitors on τ protein, there is a recent in vitro study showingthat these drugs are ab<strong>le</strong> to modu<strong>la</strong>te phosphory<strong>la</strong>tionand <strong>le</strong>vels of τ protein in SH-SY5Y cells through an interactionwith nicotinic receptors [4]. Accor<strong>di</strong>ng to these premises,the biochemical effects of AChE inhibitors can be monitoredby means of biological markers in the cerebrospinalfluid (CSF), e.g. AChE and butyrylcholinesterase (BuChE)activities and the concentrations of β-amyloid and total andphosphory<strong>la</strong>ted τ proteins. Therefore, we carried out anexplorative study by measuring activities of AChE andBuChE and concentrations of β-amyloid(1–42), τ and phospho-τproteins in CSF of AD patients before and after longtermtreatment with <strong>di</strong>fferent AChE inhibitors.We stu<strong>di</strong>ed 91 patients suffering from probab<strong>le</strong> AD ofmild to moderate degree, recruited in Malmö and Piteå,Sweden and in Perugia, Italy. A total of 84 subjects weretreated with one of three AChE inhibitors for 6 months: 59patients received donepezil, 15 were treated with ga<strong>la</strong>ntamine,and 10 received rivastigmine. As <strong>contro</strong>l group, 7 ADpatients enrol<strong>le</strong>d in a previous doub<strong>le</strong>-blind p<strong>la</strong>cebo <strong>contro</strong>l<strong>le</strong>dclinical trial who underwent lumbar puncture beforeand 6 months after treatment with p<strong>la</strong>cebo were included.AChE and BuChE activities in the CSF were measured spectrophotometrically.β-Amyloid(1–42), τ and phospo-τ [5]were determined using a specifically constructed sandwichELISA (Innogenetics, Ghent, Belgium).All the patients (or their nearest re<strong>la</strong>tives) gave informedconsent to participating in the study. At the time of enrollmentin the study, none of the patients were being treatedwith any drug interfering with cognitive functions. For statistica<strong>la</strong>nalysis, normal <strong>di</strong>stributions were tested using theShapiro-Wilk test; if normality was rejected, non-parametrictests were employed. Subgroup comparisons were doneusing the t test, after Bonferroni correction for multip<strong>le</strong> comparisons.Treatment with donepezil caused a significant and dosere<strong>la</strong>te<strong>di</strong>ncrease of CSF AChE activity as opposite to comp<strong>le</strong>telyunmo<strong>di</strong>fied BuChE activity. None of the other CSFmarkers (β-amyloid(1–42), total τ, phospho-τ) showed a significantchange after treatment.Simi<strong>la</strong>rly to that observed for donepezil, treatment withga<strong>la</strong>ntamine caused a significant increase of AChE activityin CSF, whi<strong>le</strong> BuChE activity remained unchanged. No variationswere observed in the concentrations of the other biochemicalmarkers.Although only 10 patients were treated with rivastigmine,a significant reduction of AChE activity was documente<strong>di</strong>n this group. BuChE activity and <strong>le</strong>vels of the otherbiomarkers <strong>di</strong>d not show major variations.No significant variations were observed in any of the biochemicalparameters stu<strong>di</strong>ed in the 7 <strong>contro</strong>l subjects.


S96With respect to the effect on AChE activity, the drugs testedbehaved <strong>di</strong>fferently: donepezil and ga<strong>la</strong>ntamine caused amarked (donepezil>ga<strong>la</strong>ntamine) increase; rivastigmineinduced a significant decrease. These fin<strong>di</strong>ngs are in agreementwith two recent reports [6, 7]. The <strong>di</strong>fferent mechanismsof action of these drugs might exp<strong>la</strong>in this result: donepezi<strong>la</strong>nd ga<strong>la</strong>ntamine are reversib<strong>le</strong> inhibitors, whi<strong>le</strong> rivastigmineis a pseudo-irreversib<strong>le</strong> inhibitor, implying that in the processof inactivating AChE a c<strong>le</strong>avage of the parent mo<strong>le</strong>cu<strong>le</strong> takesp<strong>la</strong>ce. Donepezil caused a strong and dose-dependent up-regu<strong>la</strong>tionof the enzyme activity, probably due to its non-competitive(i.e. non-compensatory) action; ga<strong>la</strong>ntamine has acompetitive action, which does not depend on the absoluteconcentration of the drug but more on the re<strong>la</strong>tionship with thesubstrate concentration. None of the drugs tested influencedBuChE activity. This was expected for donepezil and, to a <strong>le</strong>ssextent, also for rivastigmine [8], whi<strong>le</strong>, to our know<strong>le</strong>dge, nodata are avai<strong>la</strong>b<strong>le</strong> for ga<strong>la</strong>ntamine re<strong>la</strong>tive to human CSF stu<strong>di</strong>es.The other CSF biomarkers for AD - β-amyloid(1–42), τand phospho-τ - <strong>di</strong>d not show any significant change aftertreatment.In conclusion, this study showed that: (i) AChE inhibitorsinduced <strong>di</strong>fferent effects on AChE activity in the CSF and, at<strong>le</strong>ast for donepezil, the effect was dose-dependent; (ii) the biochemica<strong>le</strong>ffects of these drugs were detected in CSF and <strong>di</strong>fferenttreatments were <strong>di</strong>stinguished; (iii) other CSF biomarkersof AD were not significantly affected by treatment withAChE inhibitors. The possibility to detect in CSF the biochemicalprocesses taking p<strong>la</strong>ce in the central nervous systemof AD patients treated with anti-dementia drugs confirms theimportance of this approach for a better know<strong>le</strong>dge of thepathophysiology of the <strong>di</strong>sease and will allow us to demonstratethe actual impact of the new therapeutic strategies (e.g.anti-β-secretase drugs, anti-amyloid vaccine) aimed at interferingwith the pathogenetic events of the <strong>di</strong>sease.Acknow<strong>le</strong>dgements We are indebted to Dr. Eugeen Vanmeche<strong>le</strong>nfor his scientific and technical advice.References1. Mori F, Lai CC, Fusi F, Giacobini E (1995) Cholinesteraseinhibitors increase secretion of APPs in brain cortex.Neuroreport 6:633–6362. Inestrosa NC, Alvarez A, Perez CA et al (1996)Acetylcholinesterase acce<strong>le</strong>rates assembly of amyloid-β-peptidesinto Alzheimer’s fibrils: possibi<strong>le</strong> ro<strong>le</strong> of the <strong>per</strong>ipheralsite of the enzyme. Neuron 16:881–8913. Saez-Va<strong>le</strong>ro J, Sberna G, McLean CA, Small DH (1999)Mo<strong>le</strong>cu<strong>la</strong>r isoform <strong>di</strong>stribution and glycosy<strong>la</strong>tion of acetylcholinesteraseare altered in brain and cerebrospinal fluid ofpatients with Alzheimer’s <strong>di</strong>sease. J Neurochem72:1600–16084. Hellstrom-Lindahl E, Moore H, Nordberg A (2000) Increased<strong>le</strong>vels of tau protein in SH-SY5Y cells after treatment withcholinesterase inhibitors and nicotinic agonists. J Neurochem74:777–7845. Parnetti L, Lanari A, Amici S et al (2001) CSF phosphory<strong>la</strong>tedtau is a possib<strong>le</strong> marker for <strong>di</strong>scriminating Alzheimer’s <strong>di</strong>seasefrom dementia with Lewy bo<strong>di</strong>es. Neurol Sci 22:77–786. Davidsson P, B<strong>le</strong>nnow K, Andreasen N, Eriksson B, MinthonL, Hesse C (2001) Differential increase in cerebrospinal fluidacetylcholine esterase after treatment with acetylcholineesterase inhibitors in patients with Alzheimer’s <strong>di</strong>sease.Neurosci Lett 16:157–1607. Amici S, Lanari A, Romani R et al (2001) Cerebrospinal fluidacetylcholinesterase activity after long term treatment withdonepezil and rivastigmine. Mech Ageing Dev122:2057–20628. Polinsky RJ (1998) Clinical pharmacology of rivastigmine: anew generation acetylcholinesterase inhibitor for the treatmentof Alzheimer’s <strong>di</strong>sease. Clin Therapeutics 20:634-647


Neurol Sci (2002) 23:S97–S98 © Springer-Ver<strong>la</strong>g 2002Cytogenetic alterationsin lymphocytes of Alzheimer’s <strong>di</strong>seaseand Parkinson’s <strong>di</strong>sease patientsL. Petrozzi 1 • C. Lucetti 1 • R. Scarpato 2G. Gambaccini 1 • F. Trippi 2 • S. Bernar<strong>di</strong>ni 1P. Del Dotto 1 • L. Migliore 1 • U. Bonuccelli 1 ()1 Department of Neuroscience, Clinical Neurology, MovementDisorders Unit, University of Pisa, Via Roma 67, I-56126 Pisa,Italy2 Department of Science for the Study of Human Beings andEnvironment, University of Pisa, Pisa, ItalyAbstract We investigated the presence of cytogenetic alterationsin <strong>per</strong>ipheral blood lymphocytes of Alzheimer’s <strong>di</strong>sease(AD) and Parkinson’s <strong>di</strong>sease (PD) patients. Detectionof spontaneous structural and/or numerical chromosomedamage has been assessed by micronuc<strong>le</strong>us (MN) assay coup<strong>le</strong>dwith fluorescence in situ hybri<strong>di</strong>zation (FISH). Thecytogenetic investigation was <strong>per</strong>formed on 22 AD patients,18 PD patients, and 20 <strong>contro</strong>ls. The spontaneous frequenciesof micronuc<strong>le</strong>i (MN) in human lymphocytes of both ADand PD patients were significantly higher than in <strong>contro</strong>ls.The majority of MN was composed of who<strong>le</strong> chromosomesin AD patients, whi<strong>le</strong> a preva<strong>le</strong>nce of MN arising from chromosomebreakage was observed in PD patients. Differentmo<strong>le</strong>cu<strong>la</strong>r mechanisms underlie cytogenetic alterationsobserved in <strong>per</strong>ipheral lymphocytes of AD and PD patients.There is increasing interest in evaluating, at <strong>per</strong>ipheral <strong>le</strong>vel,the presence of chromosome damage in somatic cells ofpatients with neurodegenerative <strong>di</strong>seases. Cytogenetic stu<strong>di</strong>esin cultured cells of Alzheimer’s <strong>di</strong>sease (AD) patientsshowed an increase in aneuploid metaphases [1, 2], but notalways confirmed [3]. E<strong>le</strong>vated <strong>le</strong>vels of micronuc<strong>le</strong>i (MN)in <strong>per</strong>ipheral lymphocytes [4] and fibrob<strong>la</strong>sts [5] were alsoreported in AD patients. No cytogenetic study has been <strong>per</strong>formeduntil now in Parkinson’s <strong>di</strong>sease (PD) patients. Theaim of this study was to confirm the presence of chromosomemalsegregation in lymphocytes of AD patients, and toassess, in the same cell system, the spontaneous <strong>le</strong>vel ofchromosome alteration in PD patients.The cytogenetic investigation was <strong>per</strong>formed on twogroups of patients: 22 untreated AD patients (4 men and 18women of mean age 68.8 years; SD, 7.0) <strong>di</strong>agnosed accor<strong>di</strong>ngto the National Institute of Neurological andCommunicative Disorders and Stroke-Alzheimer’s Diseaseand Re<strong>la</strong>ted Disorders Association criteria; 18 untreated PDpatients (3 men and 15 women of mean age 65.5 years; SD,10.5), <strong>di</strong>agnosed accor<strong>di</strong>ng to UK Parkinson’s DiseaseSociety Brain Bank criteria. We also enrol<strong>le</strong>d 20 healthy subjectsinclu<strong>di</strong>ng 4 men and 16 women of mean age 67.1 years(SD, 6.6), matched for age, sex and smoking habits to thepatient groups. No previous exposure to toxic metals orrecent X-ray examination was reported, and all patients weredrug-free for at <strong>le</strong>ast two months before the examination.We used the MN assay for detecting the presence of chromosomedamage in cytocha<strong>la</strong>sin β-blocked binuc<strong>le</strong>ated lymphocytes[6]. MN are chromatin structures rea<strong>di</strong>ly visib<strong>le</strong> inthe cytop<strong>la</strong>sm of interphase binuc<strong>le</strong>ated cells. The applicationof fluorescence in situ hibri<strong>di</strong>zation (FISH) techniquewith an alphoid DNA probe specific for the centromere of allhuman chromosomes allowed us to <strong>di</strong>scriminate betweenMN arising from chromosomal fragments (C-MN, not incorpore<strong>di</strong>nto the daughter nuc<strong>le</strong>i at the anaphase because <strong>la</strong>ckingof the centromere) and MN that derive from <strong>la</strong>gging chromosomes(C+MN) during the anaphase stage (chromosome loss).The average spontaneous MN frequency in human lymphocytesof both AD and PD patients was higher comparedwith that of <strong>contro</strong>ls (p


S98DiscussionHigh <strong>le</strong>vels of spontaneous MN were observed in lymphocytesof AD and PD patients. Our results in<strong>di</strong>cate the presenceof cytogenetic alterations, at <strong>per</strong>ipheral <strong>le</strong>vel, in boththese neurodegenerative <strong>di</strong>seases. However, the applicationof FISH analysis revea<strong>le</strong>d that the majority of MN in ADpatients was composed preferentially of who<strong>le</strong> chromosomes.The presence of higher <strong>per</strong>centage of C+MN in ADpatients in<strong>di</strong>cates a more frequent involvement of aneuploidyin the origin of spontaneous MN, supporting thehypothesis that microtubu<strong>le</strong> impairment might be associatedwith the <strong>di</strong>sease. Altered microtubu<strong>le</strong> stability may regu<strong>la</strong>tecritical neuronal functions, <strong>per</strong>haps re<strong>la</strong>ted to <strong>le</strong>ngth, synapticcomp<strong>le</strong>xity and/or neuronal p<strong>la</strong>sticity.The high <strong>per</strong>centage of MN containing acentric fragmentsin lymphocytes of PD patients suggests that MN aremainly due to chromosome (chromatid) breakage. Activeoxygen ra<strong>di</strong>cals are known to induce chromosomal aberrations,and evidence of oxidative damage are reported at<strong>per</strong>ipheral <strong>le</strong>vel in PD patients [7, 8]. With this in mind, wecan hypothesize that the chromosomal breakage observed insomatic cells of PD patients might be re<strong>la</strong>ted to an abnormallyhigh oxidative stress.Finally, our fin<strong>di</strong>ngs in<strong>di</strong>cate that, although both neurological<strong>di</strong>seases share some common clinical and etiologicalfeatures, <strong>di</strong>fferent mo<strong>le</strong>cu<strong>la</strong>r mechanisms underlie theobserved chromosome damage in AD and PD patients.References1. Ward BE, Cook RH, Robinson A, Austin JH (1979) Increasedaneuploidy in Alzheimer <strong>di</strong>sease. Am Med Genet 3:137-1442. Nordenson I, Adolfsson R, Beckman G, Bucht G, Winb<strong>la</strong>d B(1980) Chromosomal abnormality in dementia of Alzheimertype. Lancet I:481-4823. Kormann-Bortolotto MH, Arruda Cardoso Smith M de, NetoJT (1993) Alzheimer’s <strong>di</strong>sease and ageing: a chromosoma<strong>la</strong>pproach. Gerontology 39:1-64. Migliore L, Testa, A, Scarpato R, Pavese N, Petrozzi L,Bonuccelli U (1997) Spontaneous and induced aneuploidy in<strong>per</strong>ipheral blood lymphocytes of Alzheimer’s <strong>di</strong>sease patients.Hum Genet 101:299-3055. Trippi F, Botto N, Scarpato R, Petrozzi L, Bonuccelli U,Latorraca S, Sorbi S, Migliore L (2001) Spontaneous an<strong>di</strong>nduced chromosome damage in somatic cells of spora<strong>di</strong>c andfamilial Alzheimer’s <strong>di</strong>sease patients. Mutagenesis 16:323-3276. Fenech M (1993) The cytokinesis-block micronuc<strong>le</strong>us technique:a detai<strong>le</strong>d description of the method and its applicationto genotoxicity stu<strong>di</strong>es in human popu<strong>la</strong>tions. Mutation Res285:35-447. Kilinç A, Yalçin A, Yalçin D, Taga Y, Emerk K (1988)Increased erytrocyte susceptibility to lipid <strong>per</strong>oxidation inhuman Parkinson’s <strong>di</strong>sease. Neurosci Lett 87:307-3108. B<strong>la</strong>n<strong>di</strong>ni F, Martignoni E, Ricotti R, <strong>di</strong> Jeso F, Nappi G(1999) Determination of hydroxyl fre ra<strong>di</strong>cal formation inhuman p<strong>la</strong>te<strong>le</strong>ts using high-<strong>per</strong>formance liquid chromatographywith e<strong>le</strong>ctrochemical detection. J Chromatogr B732:213-220


Neurol Sci (2002) 23:S99–S100 © Springer-Ver<strong>la</strong>g 2002Control<strong>le</strong>d-re<strong>le</strong>ase transderma<strong>la</strong>pomorphine treatment for motorfluctuations in Parkinson’s <strong>di</strong>seaseL. Priano 1 () • G. Albani 1 • S. Calderoni 1S. Baudo 1 • L. Lopiano 2 • M. Rizzone 2 • V. Astolfi 2R. Cavalli 3 • M.R. Gasco 3 • F. Fraschini 4B. Bergamasco 2 • A. Mauro 1,21Division of Neurology and Neurorehabilitation, IRCCS IstitutoAuxologico Italiano, Casel<strong>la</strong> Posta<strong>le</strong> 1, I-28044 Intra (VB), Italy2Department of Neurosciences, University of Turin, Turin, Italy3 Department of Pharmacological Sciences and Technology,University of Turin, Turin, Italy4 Department of Pharmacology, University of Mi<strong>la</strong>n, Mi<strong>la</strong>n, ItalyAbstract This study evaluated the efficacy in Parkinson’s <strong>di</strong>sease(PD) of a new pharmacologic preparation of apomorphineincluded in microemulsions and administered by transdermalroute, which provides a constant re<strong>le</strong>ase of the drug forseveral hours (Apo-TD). Twenty-one PD patients with motorfluctuations were treated with L-dopa alone, with L-dopa plusoral dopamine-agonists, or with L-dopa plus Apo-TD. Apo-TDimproved UPDRS-III and tapping test scores in “off” con<strong>di</strong>tions,and reduced duration of “off” <strong>per</strong>iods; no improvementin “on” con<strong>di</strong>tions occurred. We conclude that Apo-TD showsits efficacy particu<strong>la</strong>rly by reducing “off” <strong>per</strong>iod duration and<strong>di</strong>sability rather than improving motor <strong>per</strong>formances in “on”con<strong>di</strong>tions and therefore it seems a promising treatment forun<strong>contro</strong>l<strong>le</strong>d “off” phases in PD patients.Several strategies have been proposed to <strong>contro</strong>l “off” <strong>per</strong>iodsin advanced Parkinson’s <strong>di</strong>sease, but at present these remain amajor prob<strong>le</strong>m. Apomorphine is a well-known potent, shortactingagonist on D1 and D2 dopamine receptors, used by subcutaneousor sublingual route for the treatment of “off” <strong>per</strong>iods.However, its clinical use is limited by its short half-life ofapproximately 30 minutes and local side effects. In order toexploit the favourab<strong>le</strong> pharmacodynamic characteristics ofapomorphine and overcome the limits, we stu<strong>di</strong>ed a peculiarpharmacologic preparation of apomorphine, <strong>di</strong>ssolved inmicroemulsions, which may be administered by an epicutaneous-transdermalroute (Apo-TD) and may provide a constantre<strong>le</strong>ase of the drug for several hours, depen<strong>di</strong>ng on fewvariab<strong>le</strong>s (concentration of the drug and application area overthe skin), as demonstrated in vitro with hair<strong>le</strong>ss mouse skin [1].We se<strong>le</strong>cted 21 consecutive patients with i<strong>di</strong>opathicParkinson’s <strong>di</strong>sease, accor<strong>di</strong>ng to the following inclusion criteria:age between 55 and 75 years, stages III-IV Hoehn-Yahr,presence of long-term L-dopa syndrome characterized by“wearing-off” or pre<strong>di</strong>ctab<strong>le</strong> “off” <strong>per</strong>iods; and positiveresponse to subcutaneous apomorphine test without severeside effects. Mean age was 59.5 years and mean duration ofillness was 7.9 years with a mean Hoehn-Yahr stage of 3.4;mean daily L-dopa dosage was 645 mg. Patients were evaluatedon T0 (L-dopa therapy + oral dopamine agonists), on T1 (Ldopaalone) and on T2 (L-dopa + Apo-TD). UPDRS–part IIIand tapping test at regu<strong>la</strong>r intervals from 8 a.m. until 10 p.m.were <strong>per</strong>formed and mean score values in “off” and “on” con<strong>di</strong>tionswere calcu<strong>la</strong>ted. A <strong>di</strong>ary recor<strong>di</strong>ng duration of “off”and “on” <strong>per</strong>iods was also obtained. Apo-TD (30 mg) wasapplied to a 100 cm 2 area over the anterior part of the chestusing a bandage, and <strong>le</strong>ft for 12 hours from 8 a.m. until 8 p.m.Differences between motor <strong>per</strong>formances during APO-TDtreatment (T2 evaluation) and motor <strong>per</strong>formances obtained onT1 and T0 evaluations are summarized in Tab<strong>le</strong> 1.Mean UPDRS-III and tapping test scores obtained in“on” con<strong>di</strong>tions in T2 and T0 were not significantly <strong>di</strong>fferent.In “off” con<strong>di</strong>tions, mean UPDRS-III score on T2 waslower and tapping test score was higher compared to thoseon T0, but these <strong>di</strong>fferences <strong>di</strong>d not reach statistical significance.Conversely mean duration of “off” <strong>per</strong>iods on T2(3.1±1.2 h) was shorter compared to that on T0 (4.6±2.6 h)and this <strong>di</strong>fference was statistically significant (p=0.02).Mean UPDRS-III and mean tapping test scores in “on”con<strong>di</strong>tions in T2 were not statistically <strong>di</strong>fferent from thesame scores in T1. On the contrary mean UPDRS-III scorein “off” con<strong>di</strong>tion was lower in T2 compared to T1 (respectively30.1±10.7 vs. 36.6±9.8; p


S100Tab<strong>le</strong> 1 Clinical evaluation during L-dopa + Apo-TD treatment (T2) compared to L-dopa therapy (T1) and L-dopa + dopamine agoniststherapy (TO). Values are mean (SD)T0 T1 T2“On” “Off” “On” “Off” “On” “Off”UPDRS-III score 12.6 (8.9) 32.3 (8.4) 13.3 (5.1) 36.6 (9.8) 11.8 (7.6) 30.1 (10.7)*Tapping test score 18.3 (12.6) 7.7 (3.8) 17.4 (13.5) 6.3 (3.1) 20.1 (12.9) 8.7 (3.5)**p


Neurol Sci (2002) 23:S101–S102 © Springer-Ver<strong>la</strong>g 2002Movement-re<strong>la</strong>ted modu<strong>la</strong>tionof neural activity in human basalganglia and its L-DOPA dependency:recor<strong>di</strong>ngs from deep brain stimu<strong>la</strong>tione<strong>le</strong>ctrodes in patients with Parkinson’s<strong>di</strong>seaseA. Priori 1 () • G. Foffani 1,2 • A. Pesenti 1A. Bianchi 2 • V. Chiesa 3 • G. Baselli 2 • E. Caputo 3F. Tamma 3 • P. Rampini 1 • M. Egi<strong>di</strong> 1 • M. Locatelli 1S. Barbieri 1 • G. Scar<strong>la</strong>to 1†1 Department of Neurological Sciences, IRCCS Ospeda<strong>le</strong>Maggiore, Policlinico, University of Mi<strong>la</strong>n, Pa<strong>di</strong>glione Ponti, ViaF. Sforza 35, 20122 Mi<strong>la</strong>n, Italy2 Department of Bioengineering, Politecnico of Mi<strong>la</strong>n, Italy3 Neurology Clinic, San Paolo Hospital, Mi<strong>la</strong>n, ItalyAbstract Through e<strong>le</strong>ctrodes imp<strong>la</strong>nted for deep brain stimu<strong>la</strong>tion inthree patients (5 sides) with Parkinson’s <strong>di</strong>sease, we recorded the e<strong>le</strong>ctrica<strong>la</strong>ctivity from the human basal ganglia before, during and aftervoluntary contra<strong>la</strong>teral finger movements, before and after L-DOPA.We analysed the movement-re<strong>la</strong>ted spectral changes in the e<strong>le</strong>ctroencephalographicsignal from the subtha<strong>la</strong>mic nuc<strong>le</strong>us (STN) and fromthe internal globus pallidus (GPi). Before, during and after voluntarymovements, signals arising from the human basal ganglia containedtwo main frequencies: a high β (around 26 Hz), and a low β (around 18Hz). The high β (around 26 Hz) power decreased in the STN and GPi,whereas the low β (around 18 Hz) power decrease was consistentlyfound only in the GPi. Both frequencies changed their power with aspecific temporal modu<strong>la</strong>tion re<strong>la</strong>ted to the <strong>di</strong>fferent movement phases.L-DOPA specifically and se<strong>le</strong>ctively influenced the spectral powerchanges in these two signal bands.This is a preliminary report of a study aimed to test the ro<strong>le</strong> of thesubtha<strong>la</strong>mic nuc<strong>le</strong>us (STN) and the globus pallidus internus (GPi)in human motor <strong>contro</strong>l by exploring their movement-re<strong>la</strong>ted spectralchanges on e<strong>le</strong>ctroencephalography (EEG) recorded throughdeep brain stimu<strong>la</strong>tion (DBS) e<strong>le</strong>ctrodes and their dopaminergicdependence in free-moving patients with Parkinson’s <strong>di</strong>sease (PD).After local ethical committee approval, the study was conducted in3 patients (5 sides) with i<strong>di</strong>opathic PD undergoing stereotactic proceduresfor bi<strong>la</strong>teral imp<strong>la</strong>ntation of e<strong>le</strong>ctrodes (3389 quadripo<strong>la</strong>r <strong>le</strong>ad,Medtronic, Minneapolis, USA) for chronic DBS in the STN. Onepatient had also bi<strong>la</strong>teral imp<strong>la</strong>ntation in the GPi. The precise e<strong>le</strong>ctrodep<strong>la</strong>cement was verified posto<strong>per</strong>atively by magnetic resonance imaging(MRI). The methods have been reported elsewhere [1].The task involved the repetitive execution of brisk voluntary secondfinger extensions. The inter-trial interval was >8 s. The mean numberof non-corrupted trials <strong>per</strong> side was 115±30 (mean±SD). For eachside we recorded the surface e<strong>le</strong>ctromyographyc signal using Ag/AgCl9-mm <strong>di</strong>ameter e<strong>le</strong>ctrodes over the extensor in<strong>di</strong>cis musc<strong>le</strong> as a triggersignal; EEG signals were recorded from the contra<strong>la</strong>teral STN e<strong>le</strong>ctrodes.In patient 1, the EEG signal was also recorded from the contra<strong>la</strong>teralGPi nuc<strong>le</strong>i.The variab<strong>le</strong>s extrapo<strong>la</strong>ted were the spectral power and central frequencyvalues during the trial (in the time window between -4 s and +4s with respect to the EMG onset) in two EEG rhythms of interest: lowβ (mean central frequency 18 Hz) and high β (mean central frequency26 Hz). The spectral power was expressed as a <strong>per</strong>cent (%) increase ordecrease in instant power with respect to the mean power measured atrest (4 s to 2.4 s before EMG onset). The same procedure for recor<strong>di</strong>ngand estimating variab<strong>le</strong>s was repeated 30–40 minutes after patientsreceived a 100-mg dose of a fast-acting L-DOPA preparation (MadoparDis<strong>per</strong>sibi<strong>le</strong>, Roche) (number of non-corrupted trials <strong>per</strong> side, 99±7).Before and after L-dopa administration the three movement phases,preparation, execution and recovery (after the movement ended)elicited specific neural activity patterns in the ST and GPi (Tab<strong>le</strong> 1).The changes in the basal ganglia neural activity started duringmovement preparation (about 0.5 s before EMG onset), and reachedtheir minimum or maximum values during movement execution. Powerin the high β rhythm decreased (STN, 5 nuc<strong>le</strong>i, min value,-66.4%±22.4%; GPi, 2 nuc<strong>le</strong>i, min values, -83.5%, -91.6%). At thesame time, whereas power in the low β rhythm decreased also in theGPi (min values, -66.6%, -69,9%), it varied widely in the STN. Inpatient 1, it decreased but only slightly (2 nuc<strong>le</strong>i, min values -31.0%,-20.7%), in patient 2 (the most bradykinetic) it increased markedly (1nuc<strong>le</strong>us, max value +288.1%), and in patient 3 it decreased markedly(2 nuc<strong>le</strong>i, min values, -82.2%, -64.8%).During movement recovery, the power spectrum of the neural activityreturned to values at rest. In ad<strong>di</strong>tion, the high β rhythm in the STNand the low β rhythm in the GPi both showed a post-movement powerincrease (5 STN nuc<strong>le</strong>i, max value, +100.9%±70.4%; 2 GPi nuc<strong>le</strong>i, maxvalues, +88.1%, +172.9%) about 1–2 s after movement onset.L-DOPA, though inducing some dyskinesias, clinically improvedall the patients and also <strong>le</strong>d to changes in the movement re<strong>la</strong>ted neura<strong>la</strong>ctivity of the basal ganglia.During movement preparation and execution, L-DOPA slightly,though not significantly, reduced the high β power decrease in the STN(5 nuc<strong>le</strong>i, min value before L-DOPA, -66.4%±22.4%; after L-DOPA, -57.5%±14.0%) and in the GPi (1 nuc<strong>le</strong>us, min value before L-DOPA, -91.6%; after L-DOPA, -88.6%). Conversely, it had variab<strong>le</strong> effects onthe low β rhythm. In patient 1 it reduced the GPi power decrease (1nuc<strong>le</strong>us, min value before L-DOPA, -69.9%; after L-DOPA, -51.8%)but intensified the power decrease in the STN (1 nuc<strong>le</strong>us, min valuebefore L-DOPA, -20.7%; after L-DOPA, -43.0%). In patient 2, itremoved the <strong>la</strong>rge power increase in the STN observed before L-DOPA, and substituted it with a more “normal” pattern of powerdecrease (1 nuc<strong>le</strong>us, before L-dopa, +288.1%; after L-DOPA, -66.9%).In patient 3, it reduced the STN power decrease (2 nuc<strong>le</strong>i, before L-DOPA, -82.2%, -64.8%; after L-DOPA, -21.6%, -39.7%).A more consistent effect observed was that L-dopa significantlyreduced the power rebounds during movement recovery in the highβ rhythm in the STN (5 nuc<strong>le</strong>i, max value before L-DOPA,+100.9%±70.4%; after L-DOPA, +49.5%±40.0%; paired t test,p


S102Tab<strong>le</strong> 1 Movement-re<strong>la</strong>ted modu<strong>la</strong>tion of e<strong>le</strong>ctrical activity of the human basal ganglia before and after the administration of L-dopa.Changes in high and low b power were recorded from e<strong>le</strong>ctrodes imp<strong>la</strong>nted for deep brain stimu<strong>la</strong>tion in patients with Parkinson’s <strong>di</strong>seaseSTNGPiBaseline After L-DOPA Baseline After L-DOPA(5 nuc<strong>le</strong>i) (5 nuc<strong>le</strong>i) (2 nuc<strong>le</strong>i) (1 nuc<strong>le</strong>us)Movement preparation and executionHigh β Decreased No change Decreased No changeLow β Increased or Increased or Decreased IncreaseddecreaseddecreasedRecoveryHigh β Increased Decreased Rest power <strong>le</strong>vel No changeLow β Rest No change Increased Decreasedpower <strong>le</strong>velSTN, subtha<strong>la</strong>mic nuc<strong>le</strong>us; GPi, internal globus pallidus. Note that changes after L-DOPA are re<strong>la</strong>tive to baselineDiscussionOur preliminary observations show that e<strong>le</strong>ctrodes for DBS canopen a unique window on the basal ganglia scenario in humans. Themain fin<strong>di</strong>ng is the high β (around 26 Hz) power decrease in the STNand in the GPi, whereas the low b (around 18 Hz) decreases consistentlyonly in the GPi, but not in the STN. EEG power decreases areinterpreted as neural activation [2].In the c<strong>la</strong>ssic basal ganglia model [3, 4], the STN excites the GPi.A coherent power decrease in the same frequency range in both theSTN and GPi suggests increased output activity in the STN, excitingthe GPi and, consequently, inhibiting the tha<strong>la</strong>mocortical circuit.Hence, the high β (26 Hz) power decrease observed in this studymight ref<strong>le</strong>ct movement-re<strong>la</strong>ted activity in the in<strong>di</strong>rect pathway with afinal inhibitory effect on the tha<strong>la</strong>mocortical circuit.The second <strong>di</strong>stinctive fin<strong>di</strong>ng in this study is the re<strong>la</strong>tively specificlow β (around 18 Hz) GPi power decrease. This 18-Hz GPipower decrease was not corre<strong>la</strong>ted with STN power changes in thesame frequency and may have primarily ref<strong>le</strong>cted striatal input, whichhas an inhibitory effect on the GPi. The net result would be to reducethe output inhibitory activity of the GPi, thereby reducing inhibitionon the tha<strong>la</strong>mocortical circuit. From this point of view, the 18-Hz GPipower decrease may represent activity on the <strong>di</strong>rect pathway. In theGpi, L-DOPA reduced the decrease in the low β power, which meansa reduction in the activity of a structure with inhibitory action and,hence, a net <strong>di</strong>sinhibition of the final target of the system (i.e. the tha<strong>la</strong>mus).Thus L-DOPA faclitated the action of the <strong>di</strong>rect pathway.If the STN belongs to the in<strong>di</strong>rect pathway and if the 18 Hz activityref<strong>le</strong>cts the <strong>di</strong>rect pathway, one could wonder why some of this -specu<strong>la</strong>tively - <strong>di</strong>rect pathway-re<strong>la</strong>ted activity spreads also to the STN.A possibility is that the two pathways are not comp<strong>le</strong>tely segregated,particu<strong>la</strong>rly in Parkinson’s <strong>di</strong>sease. There are several possib<strong>le</strong> shunts,but one likely possibility is the pedunculopontine nuc<strong>le</strong>us whichreceives input from the GPi and sends an output to the STN [4].Whether these <strong>la</strong>rge power increases re<strong>la</strong>ted to movement arepathological is unc<strong>le</strong>ar. STN increases in the low β rhythm duringmovement preparation and execution may be important in bradykinesia,as the clinical and e<strong>le</strong>ctrical characteristics in Patient 2 suggest. Anincrease in both low and high β rhythms during movement recoverysuggests impaired recovery of basal ganglia function after movement.Our observation that L-DOPA reduced the post-movement powerrebound could exp<strong>la</strong>in the drug’s clinical benefit on sequential movementsthat are impaired in Parkinson’s <strong>di</strong>sease [5]. Without L-DOPA,the abnormally <strong>la</strong>rge increase in post-movement power may engage thecircuit thus preventing the system from accomplishing the rapid powerdecrease required for the next movement. In contrast, after L-DOPA theabsence of a <strong>la</strong>rge post-movement rebound might render the systemmore quickly and easily excitab<strong>le</strong>, thereby increasing the rate ofsequential movements.In conclusion, we specu<strong>la</strong>te that the human basal ganglia motor circuitprobably comprises various subcircuits, tuned onto <strong>di</strong>fferent frequencies.These subcircuits extend to the anatomical structures c<strong>la</strong>ssicallythought to belong to the <strong>di</strong>rect or in<strong>di</strong>rect pathways: a subsystemo<strong>per</strong>ating in the high β range (around 26 Hz) predominantly <strong>di</strong>stribute<strong>di</strong>n structures of the in<strong>di</strong>rect pathway, and a <strong>di</strong>fferent subsystem o<strong>per</strong>atingin the low β range (around 18 Hz) predominantly <strong>di</strong>stributed in theGPi, a structure of the <strong>di</strong>rect pathway. Large power increases duringmovement preparation and execution or during recovery seem toref<strong>le</strong>ct a pathological instability of the basal ganglia circuit, especiallyre<strong>la</strong>ted to a bradykinetic clinical con<strong>di</strong>tion. L-DOPA removes theabnormal power increases, probably by stabilizing the basal gangliacircuit and reducing the abnormal movement-re<strong>la</strong>ted synchronization.Acknow<strong>le</strong>dgments We thank Mrs. S. Gar<strong>la</strong>schi, Mr. G. Gherar<strong>di</strong>,and Mrs. M. Pastori for their assistance during recor<strong>di</strong>ngs. Thestudy was supported by grant ICS 140.1/R.F.00.195.References1. Foffani G, Priori A, Rohr M et al (2002) Event-re<strong>la</strong>ted desynchronization(ERD) in the human subtha<strong>la</strong>mus and internalglobus pallidus. J Physiol 359P:39P2. Pfurtschel<strong>le</strong>r G, Lopes da Silva FH (1999) Event re<strong>la</strong>tedEEG/MEG synchronization and desynchronization: basic princip<strong>le</strong>s.Clin Neurophysiol 110:1842–18573. A<strong>le</strong>xander GE, Crutcher MD, DeLong MR (1990) Basal gangliatha<strong>la</strong>mocorticalcircuits: paral<strong>le</strong>l substrates for motor, oculomotor,‘prefrontal’ and ‘limbic’ functions. Prog Brain Res 85:119–1464. F<strong>la</strong>herty AW, Graybiel AM (1994) Anatomy of the basal ganglia.In: Marsden CD, Fahn S (eds) Movement <strong>di</strong>sorders 3.Butterworth-Heinemann, Oxford, pp 3–275. Benecke R, Rothwell JC, Dick JP, Day BL, Marsden CD (1987)Simp<strong>le</strong> and comp<strong>le</strong>x movements off and on treatment in patientswith Parkinson’s <strong>di</strong>sease. J Neurol Neurosurg Psychiatry 50:296–303


Neurol Sci (2002) 23:S103–S104 © Springer-Ver<strong>la</strong>g 2002High-frequency e<strong>le</strong>ctrical stimu<strong>la</strong>tionof the subtha<strong>la</strong>mic nuc<strong>le</strong>usin Parkinson’s <strong>di</strong>sease: kineticand kinematic gait analysisM. Rizzone 1,3 () • M. Ferrarin 2 • A. Pedotti 2B. Bergamasco 3 • E. Bosticco 3 • M. Lanotte 4P. Perozzo 3 • A. Tavel<strong>la</strong> 3 • E. Torre 3 • M. Recalcati 2A. Melcarne 4 • L. Lopiano 31 U.O.A. of Neurology, ASL 19, Asti, Italy2 Department of Neurosciences, University of Turin, Turin, Italy3 U.O.A. of Neurosurgery, Azienda Ospedaliera CTO-CRF M.Ade<strong>la</strong>ide, Turin, Italy4 Bioengineering Centre, Don Carlo Gnocchi Foundation,Politecnico of Mi<strong>la</strong>n, Mi<strong>la</strong>n, ItalyAbstract In the advanced phase of Parkinson’s <strong>di</strong>sease (PD),gait <strong>di</strong>sturbances represent one of the main causes of <strong>di</strong>sability.Several stu<strong>di</strong>es demonstrated that high-frequency e<strong>le</strong>ctricalstimu<strong>la</strong>tion (HFS) of the subtha<strong>la</strong>mic nuc<strong>le</strong>us (STN) significantlyimproves the motor symptoms of PD. This study wasfinalised to quantitatively analyze the effect of STN HFS ongait of PD patients, through a three-<strong>di</strong>mensional gait analysissystem. Ten PD patients were stu<strong>di</strong>ed, with and without STNHFS. The results demonstrated that STN HFS significantlyimproves all the main gait parameters in PD patients.Gait <strong>di</strong>sorders p<strong>la</strong>y an important ro<strong>le</strong> in the determination ofthe <strong>di</strong>sability in patients with Parkinson’s <strong>di</strong>sease (PD). Themain characteristics of PD gait are slowness and shuffling,with a shortening of the stride <strong>le</strong>ngth and a reduction ofvelocity despite the maintenance of a normal cadence. Therange of motion at the joint <strong>le</strong>vel is tipically reduced, with aloss of the synergistic arm movements [1, 2]. Moreover, asevere incapability to start walking and sudden freezing arecharacteristics of the advanced phase of PD. These symptomsare due to an alteration of basal ganglia function re<strong>la</strong>tiveto the loss of dopaminergic neurons in the substantianigra pars compacta. In the advanced phase of PD, it is oftenimpossib<strong>le</strong> to improve motor symptoms despite the bestpharmacological treatment. Therefore, the surgical approachin these advanced phases has received increasing interest.Bi<strong>la</strong>teral high-frequency stimu<strong>la</strong>tion (HFS) of the globuspallidus internus or subthalmic nuc<strong>le</strong>us (STN) significantlyimproves PD symptoms through a functional, reversib<strong>le</strong>inhibition of the hy<strong>per</strong>active targets [3–5]. Neverthe<strong>le</strong>ss, afew stu<strong>di</strong>es have focused on the effects of HFS on PDpatients’ gait. In this study, we analysed the effects of bi<strong>la</strong>teralHFS of the STN on PD gait, using three-<strong>di</strong>mensional(3D) gait analysis techniques.We stu<strong>di</strong>ed 9 patients (5 woman, 4 men) with i<strong>di</strong>opathicPD and 9 age- and sex-matched <strong>contro</strong>ls. The mean age of thePD patients was 59.3 years, the mean <strong>di</strong>sease duration was16.6 years (SD 5.7) and the mean Hoehn and Yahr off-stagescore was 3.6 (SD 0.7). All patients were bi<strong>la</strong>terally imp<strong>la</strong>ntedfor HFS of the STN at <strong>le</strong>ast three months before the study,as previously described [5]. All patients and <strong>contro</strong>ls gavewritten informed consent to the study and the protocol wasapproved by the ethics committee. All the patients were stu<strong>di</strong>e<strong>di</strong>n two con<strong>di</strong>tions, without stimu<strong>la</strong>tion and without me<strong>di</strong>cation(Stim-off/Med-off) and with bi<strong>la</strong>teral stimu<strong>la</strong>tion andwithout me<strong>di</strong>cation (Stim-on/Med-off), in order to evaluatethe effects of the HFS alone on gait. All anti<strong>parkinson</strong>iandrugs were stopped at <strong>le</strong>ast 12 hours before the study. Thestimu<strong>la</strong>tion parameters used during the test were the sameused for chronic stimu<strong>la</strong>tion (monopo<strong>la</strong>r catho<strong>di</strong>c stimu<strong>la</strong>tion;mean amplitude 3.1 volts (SD, 0.4); mean rate 144.4 Hz (SD18.0); pulse width 60 or 90 µs).Three-<strong>di</strong>mensional kinematic and kinetic gait analyseswere <strong>per</strong>formed as previously described [6, 7] using an optoe<strong>le</strong>ctronicsystem (ELITE System, BTS, Corsico, Italy) [8]to measure the 3D coor<strong>di</strong>nates of retroref<strong>le</strong>ctive markers anda dynamometric force p<strong>la</strong>tform (Kist<strong>le</strong>r, Winterthur,Switzer<strong>la</strong>nd) for ground reaction force (GRF) detection.Four video cameras, with a sampling rate of 50 Hz, werep<strong>la</strong>ced postero<strong>la</strong>terally of the subject. The working volume(2x3x1 m 3 ) was calibrated by means of a precision grid. Theacquired markers’ coor<strong>di</strong>nates were <strong>di</strong>gitally low-pass filteredto estimate the optimal cut-off frequency and minimize theresidual noise (resultant cut-off frequency within the range of3–7 Hz). Nineteen markers (10 mm <strong>di</strong>ameter) were glued onthe subjects’bony <strong>la</strong>ndmarks: the fifth metatarsal heads, the <strong>la</strong>teralmal<strong>le</strong>oli, the <strong>la</strong>teral femoral condy<strong>le</strong>s, the posterior su<strong>per</strong>ioriliac spines, the acromions, the <strong>la</strong>teral humeral condy<strong>le</strong>s, theulnar prominences, the base of the occipital bone, the sacrum,the seventh thoracic vertebra and the seventh cervical vertebra.The force p<strong>la</strong>tform (60x40 cm 2 ) was embedded in the floorwithin the calibrated volume and in the midd<strong>le</strong> of a 10-m pathway.Ground reaction forces were samp<strong>le</strong>d at 50 Hz.Marker coor<strong>di</strong>nates and ground reactions were processedusing specific analysis programs [7] to compute spatio-temporalgait parameters, joint and body segment kinematics,joint moment and powers, and body baricenter kinematics.For the analysis of stride <strong>le</strong>ngth, step <strong>le</strong>ngth and meangait velocity were expressed as the <strong>per</strong>centage of body height(%h and %h/s respectively) and the <strong>per</strong>centage of height <strong>per</strong>second (% /s), respectively. Ground reaction components,joint moments and joint powers were normalised by <strong>di</strong>vi<strong>di</strong>ngby body weight (N/kg, Nm/kg and W/kg, respectively). Forthe analysis we considered the following parameters: thespatiotemporal gait parameters (stride <strong>le</strong>ngth, cadence, meangait velocity, <strong>per</strong>centage of the stance phase on the strideduration); the range of amplitude during the gait cyc<strong>le</strong> of thehip, knee and ank<strong>le</strong> joint ang<strong>le</strong>s, of the pelvis orientation inthe frontal p<strong>la</strong>ne, of trunk <strong>la</strong>teral f<strong>le</strong>xion and trunk torsion;the maximal values within the gait cyc<strong>le</strong> of the hip and ank<strong>le</strong>


S104joint moments and powers; the mean forward inclination ofthe trunk in the sagittal p<strong>la</strong>ne. All variab<strong>le</strong>s were averagedamong the two sides. Two-tai<strong>le</strong>d Student’s t-test for pairedmeasures was applied to examine <strong>di</strong>fferences between thetwo con<strong>di</strong>tions (Stim-off/Med-off and Stim-on/Med-off).Unpaired measures tests were used for the comparisonsbetween patients and <strong>contro</strong>ls. The minimum 0.05 <strong>le</strong>vel ofsignificance was adopted.The mean gait velocity of patients in Stim-off/Med-offwas significantly reduced with respect to that of <strong>contro</strong>ls(34.5±14.6 %h/s vs. 69.1±10.3 %h/s), mainly for the reductionof the stride <strong>le</strong>ngth (41.5%±11.9% h vs. 77.0%±6.9% h)with litt<strong>le</strong> reduction of cadence (96.4±18.3 steps/min vs.107.3±7.7 steps/min) and litt<strong>le</strong> increase in stance phase(65.4%±4.8% vs. 59.4%±1.8%). In the Stim-on/Med-offcon<strong>di</strong>tion, there was a significant increase of both the meangait velocity and the mean stride <strong>le</strong>ngth (52.5±16.0% h/s and59.9±10.6 %h/s, respectively), whi<strong>le</strong> the cadence increase(103.1±16.1 steps/min) and the stance phase reduction(61.7%± 3.8%) were not significant.All subjects walked in the Stim-on/Med-off con<strong>di</strong>tionwith a <strong>la</strong>rger range of motion (ROM) of all lower limb jointswith respect to the Stim-off/Med-off con<strong>di</strong>tion (hip ang<strong>le</strong>ROM, 39.1°±7.6° vs. 29.3°±8.2°; knee ang<strong>le</strong> ROM,47.0°±7.7° vs. 37.0°±10.3°; ank<strong>le</strong> ang<strong>le</strong> ROM, 18.8°±3.9°vs. 13.7°±3.3). In the Stim-on/Med-off con<strong>di</strong>tion, the rangeof pelvic obliquity increased with respect to the Stimoff/Med-offcon<strong>di</strong>tion (7.6°±3.6° vs. 5.9°±2.3°). Also thetrunk <strong>la</strong>teral f<strong>le</strong>xion and torsion increased (8.0°±3.2° vs.5.9°±1.5° and 8.5°±4.5° vs. 5.0°±2.3°, respectively), whi<strong>le</strong>the forward inclination of the trunk significantly decreasedtowards <strong>contro</strong>ls values (11.2°±5.1° vs. 15.1°±8.1°).For the kinetic variab<strong>le</strong>s, the peak values of moments andpowers at hip and ank<strong>le</strong> joints increased in all patients whenthe Stim-on/Med-off was on compared to the Stim-off/Medoff(hip moment peak: 0.955 Nm/kg ± 0.338 vs. 0.846 Nm/kg± 0.269; ank<strong>le</strong> moment peak: 1.135 Nm/kg ± 0.262 vs. 1.082Nm/kg ± 0.220; hip power peak: 1.570 W/kg ± 0.843 vs.0.893 W/kg ± 0.487; ank<strong>le</strong> power peak: 1.241 W/kg ± 0.567vs. 0.710 W/kg ± 0.408).DiscussionIn this study, we analysed the effects of HFS of the STN onPD patients’ gait using a 3D system. The data col<strong>le</strong>cted inthe Stim-off/Med-off con<strong>di</strong>tion confirmed, as observed inother stu<strong>di</strong>es, that PD gait is slow and with short steps,whi<strong>le</strong> the cadence and the duration of the stride sub-phasesare quite simi<strong>la</strong>r to <strong>contro</strong>ls; these fin<strong>di</strong>ngs validate theadopted protocol of analysis.The main effect of the STN HFS is the <strong>la</strong>rge increase instride <strong>le</strong>ngth, which is mostly responsib<strong>le</strong> for the increase ingait velocity, whi<strong>le</strong> the increase in cadence and the reductionof stance phase duration were not significant. In the Stimon/Med-offcon<strong>di</strong>tion, the angu<strong>la</strong>r excursion at lower limbjoints, reduced in PD patients, was increased, with a <strong>la</strong>rgervariation at hip <strong>le</strong>vel; also the abnormal forward inclination ofthe trunk and the reduced range of amplitude of trunk torsionand <strong>la</strong>teral f<strong>le</strong>xion, typical of PD patients were improved.These changes observed in the Stim-on/Med-off con<strong>di</strong>tionsuggest that STN HFS is capab<strong>le</strong> of significantlyimproving PD gait, <strong>le</strong>a<strong>di</strong>ng toward a more physiologicalmechanism of walking speed modu<strong>la</strong>tion. More stu<strong>di</strong>es,with a comparison of the effects of stimu<strong>la</strong>tion and <strong>le</strong>vodopa,are needed to better investigate the ro<strong>le</strong> of basal gangliain gait mechanisms.References1. Blin O, Ferrandez AM, Serratrice G (1990) Quantitativeanalysis of gait in Parkinson patients: increased variability ofSL. J Neurol Sci, 98:91–972. Pedersen SW, Oberg B, Larsson L-E, Lindval B (1997) Gaitanalysis, isokinetic musc<strong>le</strong> strength measurement in patientswith Parkinson’s <strong>di</strong>sease. Scand J Rehabil Med 29:67–743. Benabid AL, Kous<strong>di</strong>e A, Benazzouz A, Pial<strong>la</strong>t B, Krack P,Limousin-Dowsey P, Lebas JF, Pol<strong>la</strong>k P (2001) Deep brainstimu<strong>la</strong>tion for Parkinson’s <strong>di</strong>sease. Adv Neurol 86:405–4124. Hal<strong>le</strong>tt M, Litvan I (1999) Evaluation of surgery forParkinson’s <strong>di</strong>sease: a report of the Therapeutics andTechnology Assessment Subcommittee of the AmericanAcademy of Neurology. The Task Force on Surgery forParkinson’s Disease. Neurology 53:1910–19215. Lopiano L, Rizzone M, Bergamasco B, Tavel<strong>la</strong> A, Torre E,Perozzo P, Va<strong>le</strong>ntini MC, Lanotte M (2001) Deep brain stimu<strong>la</strong>tionof the subtha<strong>la</strong>mic nuc<strong>le</strong>us: clinical effectiveness andsafety. Neurology 56:552–5546. Pedotti A, Frigo C (1992) Quantitative analysis of locomotionfor basic research and clinical application. Funct Neurol Suppl7(4):47–567. Ferrarin M, Lopiano L, Rizzone M, Lanotte M, BergamascoB, Recalcati M, Pedotti A (2002) Quantitative analysis of gaitin Parkinson’s <strong>di</strong>sease: a pilot study on the effects of bi<strong>la</strong>teralsubthalmic stimu<strong>la</strong>tion. Gait and posture (in press)8. Ferrigno G, Pedotti A (1985) ELITE: a <strong>di</strong>gital de<strong>di</strong>cated hardwaresystem for movement analysis via real-time TV signalprocessing. IEEE Trans Biom Eng 32(11):943–949


Neurol Sci (2002) 23:S105–S106 © Springer-Ver<strong>la</strong>g 2002Cognitive and psychiatriccharacterization of patientswith Huntington’s <strong>di</strong>sease and theirat-risk re<strong>la</strong>tivesP. Soliveri 1 • D. Monza 1 • S. Piacentini 1 • D. Pari<strong>di</strong> 1C. Nespolo 1 • C. Gel<strong>le</strong>ra 2 • C. Mariotti 2A. Albanese 1 • F. Girotti 1 ()1 Department of Neurology I, C. Besta National NeurologicalInstitute, Via Celoria 11, I-20133 Mi<strong>la</strong>n, Italy2 Biochemical and Genetics Laboratory, C. Besta NationalNeurological Institute, Mi<strong>la</strong>n, ItalyAbstract We examined cognitive and psychiatric <strong>di</strong>sturbancesin patients with Huntington’s <strong>di</strong>sease (HD) in comparison to atrisk asymptomatic subjects. Cognitive and psychiatric sca<strong>le</strong>sand an HD motor sca<strong>le</strong> were administered to 40 HD patients,17 pre-symptomatic HD gene carriers (AR + ) and 28 non genecarriers (AR - ). HD patients <strong>di</strong>d worse than AR + and AR - in allmotor, cognitive and psychiatric measures, whi<strong>le</strong> AR + and AR -subjects <strong>di</strong>d not <strong>di</strong>ffer between each other. HD patients hadhigh scores for negative psychiatric symptoms, but there wasno corre<strong>la</strong>tion between illness duration and psychiatric or cognitive<strong>per</strong>formance. In HD, <strong>di</strong>sease course and symptomatologyare heterogeneous and negative psychiatric symptoms arecommon.IntroductionHuntington’s <strong>di</strong>sease (HD) is an autosomal dominant neurodegenerative<strong>di</strong>sease characterised by motor, cognitive andpsychiatric <strong>di</strong>sturbances. Motor and cognitive impairmenthave been fairly extensively investigated, whi<strong>le</strong> behavioura<strong>la</strong>nd psychiatric aspects have been neg<strong>le</strong>cted. However, the<strong>la</strong>tter have a major impact on the lives of patients and theirre<strong>la</strong>tives, and render <strong>di</strong>sease management prob<strong>le</strong>matic.Psychiatric <strong>di</strong>sturbances are particu<strong>la</strong>rly frequent (40% to70%) at all <strong>di</strong>sease stages [1]. Some stu<strong>di</strong>es in<strong>di</strong>cate that psychiatric<strong>di</strong>sturbances are often a prelude to the onset of choreaand cognitive decline [2]. However, this fin<strong>di</strong>ng was not confirmedon pre-symptomatic gene mutation carriers, who as agroup, <strong>di</strong>d not have psychiatric or cognitive impairment [3].The aim of this study was to examine cognitive and psychiatric<strong>di</strong>sturbances in HD patients in comparison to agroup of asymptomatic offspring of HD patients who weretherefore at risk (AR) for the <strong>di</strong>sease. We also sought corre<strong>la</strong>tionsbetween motor, cognitive and behavioural alterationsin patients with HD.Patients and methodsWe stu<strong>di</strong>ed 85 adults (39 men) from 35 HD families: 40 HDpatients (24 men) <strong>di</strong>agnosed from family history, chorea, or characteristicimpairment of voluntary movement on the QuantifiedNeurological Sca<strong>le</strong> (QNE) for HD, and confirmed by the genetictest for HD; 17 asymptomatic at risk subjects (7 men) who testedpositive for the pathological HD gene (AR + ); and 28 asymptomaticat risk subjects (8 men) who tested negative (AR - ).The neuropsychological tests administered were the MiniMental State Examination (MMSE) to assess global cognitive function;the Raven Progressive Matrices 1947 (PM47) to investigatedeductive reasoning and visuo-<strong>per</strong>ceptive ability; the Short Ta<strong>le</strong>Test to assess long term verbal memory; the Visual Search Testwhich examines focused attention; the Benton Visual OrientationLine Test to assess orientation and spatial <strong>per</strong>ception; the VerbalFluency Test (phonemic) to assess strategic word searching and theNelson simplified form of the Wisconsin Card Sorting Test to assesscategorization and set shifting abilities. All except the Nelson Test– for which a correction is not avai<strong>la</strong>b<strong>le</strong> – were corrected for ageand education.The psychiatric sca<strong>le</strong>s were: the Brief Psychiatric Rating Sca<strong>le</strong>(BPRS) a screening test for psychiatric <strong>di</strong>sturbances; the HamiltonAnxiety Sca<strong>le</strong> (HAM-A) which evaluates anxiety, the HamiltonPsychiatric Rating Sca<strong>le</strong> for Depression (HAM-D) whose score is acombination of subjective depressive feelings and the examiner’sjudgement, the Sca<strong>le</strong> for the Assessment of Positive Symptoms(SAPS) which quantifies positive schizophrenic symptoms (hallucinations,delusions, bizarre behaviour and <strong>di</strong>sturbances ofthought), the Sca<strong>le</strong> for the Assessment of Negative Symptoms(SANS) which quantifies negative schizophrenic symptoms (emotionalf<strong>la</strong>tness, abulia, apathy, retiring social behaviour, impairedattention, etc).The genetic test, <strong>per</strong>formed in all subjects, determines the numberof CAG repeats (pathological value >36) on the IT 15 gene ofchromosome 4. The statistical tests used were one-way ANOVAwith post-hoc Scheffè comparison as appropriate, and Pearson’scorre<strong>la</strong>tion test.ResultsPatients with chorea were significantly older than the AR +and AR - subjects; mean duration of education was higher inthe AR + group. CAG expansion <strong>le</strong>ngth <strong>di</strong>d not <strong>di</strong>ffer betweenpatients and AR + subjects (Tab<strong>le</strong> 1).As expected, patients <strong>di</strong>d significantly worse than AR +and AR - in all cognitive tests (p


S106Tab<strong>le</strong> 1 Demographic data and psychiatric scores in HD patients, at-risk carriers (AR + ) and at-risk non carriers (AR - )HD AR + AR - p(n=40) (n=17) (n=28)Men, n (%) 24 (60) 7 (14) 8 (29) –Age, years 43.0 (11.7) 30.2 (7.2) 31.6 (12.3)


Neurol Sci (2002) 23:S107–S108 © Springer-Ver<strong>la</strong>g 2002CAG mutation effect on rateof progression in Huntington’s <strong>di</strong>seaseF. Squitieri () • M. Cannel<strong>la</strong> • M. SimonelliNeurogenetics Unit, IRCCS Neuromed, Pozzilli (IS), ItalyAbstract Huntington’s <strong>di</strong>sease (HD) is progressively invalidatingand caused by a CAG expanded mutation. We testedthe effect of the mutation <strong>le</strong>ngth on the rate of progression ina cohort of 80 patients clinically followed-up and geneticallycharacterized. Two patients presenting an infanti<strong>le</strong> andaggressive HD form starting under 10 years had over 90repeats; the other patients <strong>di</strong>d not show any influence of theCAG expanded number on the rate of progression. In conclusion,the CAG expanded repeat affects the <strong>di</strong>sease progressiononly at a very up<strong>per</strong> pathological range and in rarecases initiating very early in the life, whi<strong>le</strong> it does not seemto affect in any way the severity of the phenotype in mostHD patients. Other factors affecting the motor symptom progression,other than the expanded repeats, therefore have tobe investigated.Huntington <strong>di</strong>sease’s (HD), dominantly transmitted andcaused by a CAG expanded mutation beyond 36 repeats [1],is highly and progressively invalidating. The severity of thesymptom progression is variab<strong>le</strong> among patients, with juveni<strong>le</strong>(onset


S108Loss-of-units<strong>per</strong> year at TFCLoss-of-units<strong>per</strong> year at DSExpanded CAG repeat numberFig. 1a-d. Linear regressionanalysis between expandedCAG repeat number (xaxis)and loss of units <strong>per</strong>year (y-axis) at the TFC andDS. a,b Analysis inclu<strong>di</strong>ngthe two patients with infanti<strong>le</strong>form. c,d Exclu<strong>di</strong>ng the2 patients with infanti<strong>le</strong>Huntington’s <strong>di</strong>sease. Theinfanti<strong>le</strong> patients are highlightedby a circ<strong>le</strong> in a and bmovement <strong>di</strong>sorder [9]. In that study, we found meanexpanded repeats longer in the juveni<strong>le</strong> rigid patients than inthe juveni<strong>le</strong>s initiating with chorea, pre<strong>di</strong>cting therefore theoccurrence of an influence of <strong>la</strong>rge expansions on HD phenotype.We confirm such hypothesis as both patients affectedwith infanti<strong>le</strong> form here described presented at onsetmovement <strong>di</strong>sorder other than chorea, characterized by rigi<strong>di</strong>tyin one and dystonia and limb ataxia in the other. In thesecases, as in the rigid juveni<strong>le</strong> patients, the particu<strong>la</strong>rlyexpanded and toxic mutation <strong>le</strong>ads to a highly invalidatingphenotype. Conversely, in most patients the CAG repeatcontributes to the age at onset for about 50%–60%, theremaining <strong>per</strong>centage is influenced by other genetic factorsprobably of familial origin [3], but not to the rate of <strong>di</strong>seaseprogression. Other factors affecting the severity of the phenotypehave to be stu<strong>di</strong>ed on cohorts of patients well characterizedclinically and genetically. The <strong>di</strong>scovery of factorsinfluencing the symptom progression and, possibly pre<strong>di</strong>ctingthe HD prognosis, will offer new opportunities in thetherapeutic strategies for this devastating <strong>di</strong>sease.References1. Kremer B, Goldgerg P, Andrew SE et al (1994) A worldwidestudy of the Huntington’s <strong>di</strong>sease mutation. N Engl J Med330:1401–14062. Nance MA (1997) The US HD Genetic Testing Group: genet-ic testing of children at risk for Huntington’s <strong>di</strong>sease.Neurology 49:1050–10533. Squitieri F, Sabba<strong>di</strong>ni G, Man<strong>di</strong>ch P et al (2000) Family andmo<strong>le</strong>cu<strong>la</strong>r data for a fine analysis of age at onset inHuntington <strong>di</strong>sease. Am J Med Genet 95:366–3734. Rubinsztein DC, Leggo J, Co<strong>le</strong>s R et al (1996) Phenotypiccharacterization of in<strong>di</strong>viduals with 30–40 CAG repeats in theHuntington <strong>di</strong>sease (HD) gene reveals HD cases with 36repeats and apparently normal elderly in<strong>di</strong>viduals with 36–39repeats. Am J Hum Genet 59:16–225. Squitieri F, Cannel<strong>la</strong> M, Giallonardo P et al (2001) Onset andpre-onset stu<strong>di</strong>es to define the Huntington’s <strong>di</strong>sease naturalhistory. Brain Res Bull 56:233–2386. Shoulson I, Kur<strong>la</strong>n R, Rubin AJ et al (1989) Assessment offunctional capacity in neurodegenerative movement <strong>di</strong>sorders:Huntington’s <strong>di</strong>sease as a prototype. In: Munsat TL (ed)Quantification of neurological deficit. Butterworths, Boston,pp 271–2837. Myers RH, Sax DS, Koroshetz WJ et al (1991) Factors associatedwith slow progression in Huntington’s <strong>di</strong>sease. ArchNeurol 48:800–8048. Warner JP, Barron LH, Brock DJH (1993) A new polymerasechain reaction (PCR) assay for the trinuc<strong>le</strong>otide repeat that isunstab<strong>le</strong> and expanded on Huntington’s <strong>di</strong>sease chromosome.Mol Cell Probes 7:235–2399. Squitieri F, Berardelli A, Nargi E et al (2000) Atypical movement<strong>di</strong>sorders in the early stages of Huntington’s <strong>di</strong>sease:clinical and genetic analysis. Clin Genet 58:50–5610. Il<strong>la</strong>rioshkin NS, Igarashi S, Onodera O et al (1994)Trinuc<strong>le</strong>otide repeat <strong>le</strong>ngth and rate of progression ofHuntington’s <strong>di</strong>sease. Ann Neurol 36:630–635


Neurol Sci (2002) 23:S109–S110 © Springer-Ver<strong>la</strong>g 2002Anatomo-clinical corre<strong>la</strong>tionof intrao<strong>per</strong>ative stimu<strong>la</strong>tion-inducedside-effects during HF-DBSof the subtha<strong>la</strong>mic nuc<strong>le</strong>usF. Tamma 1 () • E. Caputo 1 • V. Chiesa 1 • M. Egi<strong>di</strong> 2M. Locatelli 2 • P. Rampini 2 • C. Cinnante 3A. Pesenti 3 • A. Priori 31 Department of Neurology, Ospeda<strong>le</strong> San Paolo, Via <strong>di</strong> Ru<strong>di</strong>nì 8, I-20142, Mi<strong>la</strong>n, Italy2 Department of Neurosurgery, IRCCS Ospeda<strong>le</strong> MaggiorePoliclinico, Mi<strong>la</strong>n, Italy3 Department of Neurology, IRCCS Ospeda<strong>le</strong> MaggiorePoliclinico, Mi<strong>la</strong>n, ItalyAbstract The efficacy of deep brain stimu<strong>la</strong>tion of the subtha<strong>la</strong>micnuc<strong>le</strong>us (STN) is dependent on the accuracy of targeting.In order to reduce the number of passes and, consequently,the duration of surgery and risk of b<strong>le</strong>e<strong>di</strong>ng, we haveset up a new method based on <strong>di</strong>rect magnetic resonanceimaging (MRI) localisation of the STN. This procedureallows a short duration of the neurophysiological session(one or two initial tracks). Whenever a supp<strong>le</strong>mentary trackis needed, the stimu<strong>la</strong>tion-induced side effects are analysedto choose from one of the remaining ho<strong>le</strong>s in Ben’s gun. Agood know<strong>le</strong>dge of anatomical structures surroun<strong>di</strong>ng theSTN is mandatory to re<strong>la</strong>te side effects to the actual positionof the track. In our series of 11 patients (22 sides, 37 tracks),the most common and reproducib<strong>le</strong> side effects were thosecharacterised by motor, sensorial, oculomotor and vegetativesigns and symptoms. Moreover, the therapeutic window (<strong>di</strong>stancebetween the current intensity needed to obtain the bestclinical effect and the intensity capab<strong>le</strong> to induce sideeffects) pre<strong>di</strong>cted clinical efficacy in the long-term, and contributedto the choice of which among the examined trackshad to be imp<strong>la</strong>nted with the chronic macroe<strong>le</strong>ctrode.The efficacy of high frequency deep brain stimu<strong>la</strong>tion (HF-DBS) of the subtha<strong>la</strong>mic nuc<strong>le</strong>us (STN) for the treatment ofadvanced Parkinson’s <strong>di</strong>sease (PD) is strictly dependent on theprecision of reaching the target. This concept was well c<strong>le</strong>ar tothe first authors when they proposed a multitrack approach todetermine target location in DBS [1]. Afterwards, severa<strong>la</strong>uthors have replicated their good clinical results. Various <strong>di</strong>fferentstrategies have been developed to reach the target, generallywith a multip<strong>le</strong> simultaneous or independent trajectoriesapproach to cover with both recor<strong>di</strong>ng and stimu<strong>la</strong>tion a widerarea. Recently a review on STN-imp<strong>la</strong>nted patients suggestedthat the higher the number of microe<strong>le</strong>ctrode passes, the higherthe risk of intracranial b<strong>le</strong>e<strong>di</strong>ng during DBS maneuvers [2].In order to improve the risk-benefit ratio, we proposed anovel technique guided by magnetic resonance imaging(MRI) and based on multip<strong>le</strong> sequential image fusion(MuSIF), which allows a <strong>di</strong>rect localisation of the subtha<strong>la</strong>micnuc<strong>le</strong>us [3]. In our ex<strong>per</strong>ience, the high targeting precisionreached by this technique made it possib<strong>le</strong> to reduce thenumber of intrao<strong>per</strong>ative tracks. We present our DBS methodand <strong>di</strong>scuss how the analysis of stimu<strong>la</strong>tion-induced sideeffects evoked during the initial neurophysiological session(one or two tracks) can help address a supp<strong>le</strong>mentary track,if needed, and limit the total number of passes.Twelve consecutive PD patients were se<strong>le</strong>cted for bi<strong>la</strong>teralSTN imp<strong>la</strong>ntation of e<strong>le</strong>ctrodes. The o<strong>per</strong>ative session wasstarted after a navigation session in which the stereotactic coor<strong>di</strong>nateswere obtained by means of the fusion of stereotacticcomputed tomography (CT) and frame<strong>le</strong>ss MRI data. Througha burr ho<strong>le</strong> located 2 cm anteriorly to the coronal suture, onemicroe<strong>le</strong>ctrode inserted into the central ho<strong>le</strong> of the Ben’s gunwas advanced with a micrometric drive towards the target(patients 1–7). For patients 8–12, two paral<strong>le</strong>l microe<strong>le</strong>ctrodeswere simultaneously inserted into the brain. The Ben’s gun has5 ho<strong>le</strong>s 2.0 mm apart from each other and arranged in a crossfashion.Recor<strong>di</strong>ng and then semi-microstimu<strong>la</strong>tion were <strong>per</strong>formedat several <strong>le</strong>vels with a coaxial e<strong>le</strong>ctrode starting 6 mmbefore and en<strong>di</strong>ng 6 mm after the theoretical target. The resultswere col<strong>le</strong>cted and then <strong>di</strong>scussed during a brain storm session.If the required criteria were satisfied, i.e. recor<strong>di</strong>ng and stimu<strong>la</strong>tiondata met our standard, the chronic macroe<strong>le</strong>ctrode wasimp<strong>la</strong>nted; otherwise a new neurophysiological session wasstarted with a supp<strong>le</strong>mentary track.Position of chronic e<strong>le</strong>ctrodes was regu<strong>la</strong>rly checked a fewdays after stimu<strong>la</strong>tors imp<strong>la</strong>ntation, by means of a posto<strong>per</strong>ativeMR image, which was eventually compared with the preo<strong>per</strong>ativeMR image. The e<strong>le</strong>ctrode was visualised as a 1.5-mm <strong>di</strong>ameter artefact which is an acceptab<strong>le</strong> size to reliablyin<strong>di</strong>cate the e<strong>le</strong>ctrode’s actual position. We have arbitrarilysub<strong>di</strong>vided the STN into nine regions: 1 central and 8 <strong>per</strong>ipheral(Fig. 1). All the e<strong>le</strong>ctrodes were c<strong>la</strong>ssified as central (C),anterome<strong>di</strong>al (AM), anterior (A), antero<strong>la</strong>teral (AL), <strong>la</strong>teral(L), postero<strong>la</strong>teral (PL), posterior (P), posterome<strong>di</strong>al (PM), orme<strong>di</strong>al (M), accor<strong>di</strong>ng to the position of the artefact.We analysed the side effects evoked by intrao<strong>per</strong>ativesemi-microstimu<strong>la</strong>tion at 5 <strong>le</strong>vels (-2, -1, 0, +1, +2; <strong>le</strong>vel 0was the target), thereby covering a 4-mm area along the trajectoryof each track.The therapeutic window for each side effect was considered,i.e. the ratio between the current intensity needed foreliciting the side effect and the current intensity required atthe same <strong>le</strong>vel to obtain the maximum clinical effect.At the 3-month follow-up, all patients benefited fromchronic stimu<strong>la</strong>tion as shown by improvement of the UPDRSIII score in med-off con<strong>di</strong>tions and reduction in off-time anddyskinesia scores and <strong>le</strong>vodopa equiva<strong>le</strong>nt daily dose (LEDD).The first patient <strong>di</strong>d not undergo posto<strong>per</strong>ative MRI and wasnot enrol<strong>le</strong>d in this study. In the remaining consecutive 11patients (22 sides), we used 37 passes with a mean of 1.7 <strong>per</strong>side. No intracranial b<strong>le</strong>e<strong>di</strong>ng occurred during the procedures.In the 12 patients (24 sides), the central track was acceptedfor definitive imp<strong>la</strong>ntation in 20 sides (83.3%). In 6 sides,


S110Fig. 1 The position of the definitive e<strong>le</strong>ctrocatheter asvisualised on MRI is then reported onto theSchaltenbrandt At<strong>la</strong>s axial view of the subtha<strong>la</strong>mic area.Each circ<strong>le</strong> corresponds to a <strong>di</strong>screte part of this areain which clinical and neurophysiological results were mildlyconsistent with our criteria, a supp<strong>le</strong>mentary trajectory wasexplored and eventually chosen for definitive imp<strong>la</strong>ntation in2 sides. The side effects evoked during stimu<strong>la</strong>tion addressedthe choice of which of the avai<strong>la</strong>b<strong>le</strong> ho<strong>le</strong>s in the gun had tobe used and consequently the orientation of the ad<strong>di</strong>tionaltrack. We never needed a fourth pass.The most common side effects elicited during the 37 tracksstimu<strong>la</strong>tion were pyramidal, sensorial, oculomotor and vegetative.Unspecific side effects, like chest constriction, ma<strong>la</strong>iseand <strong>di</strong>zziness, were hardly re<strong>la</strong>ted to specific structures, sothey were not useful to locate the position of the track.Motor contractions in the contra<strong>la</strong>teral hemibody were dueto stimu<strong>la</strong>tion of the corticobulbar and corticospinal tracts.They were frequent (30 tracks, 81%), particu<strong>la</strong>rly in C and ALregions, but not in M and PM regions. They were time-lockedto stimu<strong>la</strong>tion and of paramount importance for the definitionof the therapeutic window. We presumed that horizontal gazedeviation can be considered to be a motor side effect derivingfrom stimu<strong>la</strong>tion of the corticofugal pathways which traversethe anterior limb of the internal capsu<strong>le</strong>. Moreover we consideredboth reduced ipsi<strong>la</strong>teral gaze and contra<strong>la</strong>teral gaze deviationsto be part of the same phenomenon, since they wereevoked at the same <strong>le</strong>vel and since the <strong>la</strong>tter comes out as thecurrent intensity is increased.Sensorial side effects were <strong>per</strong>ceived by the patient asparaesthesic sensations and were due to stimu<strong>la</strong>tion of theme<strong>di</strong>al <strong>le</strong>mniscus fibres. They were fairly common (12tracks, 32.5%; <strong>per</strong>sistent in 6 of 12) and more frequent in Mand PM regions and at lower <strong>le</strong>vels.Oculomotor side effects were quite uncommon (9 tracks,24%), exclusive of C, M and AM regions. They were due tostimu<strong>la</strong>tion of the third nerve (adduction or reduced abduction,e<strong>le</strong>vation of su<strong>per</strong>ior eyelid in ipsi<strong>la</strong>teral eye) or of therostral interstitial nuc<strong>le</strong>us (oblique, i.e. <strong>la</strong>teral and upward,gaze deviation).Vegetative side effects (nausea, heat sensation, sweating,bradycar<strong>di</strong>a, observed in 15 tracks, 40.5%) were frequent inthe anterome<strong>di</strong>al area and <strong>le</strong>ss common in C region. Theywere not reproducib<strong>le</strong> in the long-term stimu<strong>la</strong>tion.DiscussionIn order to reduce the risk of b<strong>le</strong>e<strong>di</strong>ng, we used only one or twotrajectories during the initial neurophysiological session ofSTN surgery. Stimu<strong>la</strong>tion-induced side effects are a reliab<strong>le</strong>and reproducib<strong>le</strong> tool for determining the actual localisation ofthe trajectories. Therefore, if a supp<strong>le</strong>mentary trajectory isneeded, the analysis of side effects can help in deci<strong>di</strong>ng whichone among the avai<strong>la</strong>b<strong>le</strong> ho<strong>le</strong>s in the Ben’s gun has to be used.Moreover, the ratio of side effects to clinical effects (therapeuticwindow) is important in deci<strong>di</strong>ng where to imp<strong>la</strong>nt thedefinitive e<strong>le</strong>ctrode. It is a marker of the <strong>di</strong>stance between thetip of the e<strong>le</strong>ctrode and the structure which is responsib<strong>le</strong> forthat side effect. The lower is the value of the therapeutic window,the nearer is the tip of the e<strong>le</strong>ctrode to structures otherthan STN. The amplitude of the therapeutic window pre<strong>di</strong>ctsthe safety of increasing current intensity in the long term.References1. Limousin P, Pol<strong>la</strong>k P, Benazzouz A et al (1995) Effect on<strong>parkinson</strong>ian signs and symptoms of bi<strong>la</strong>teral subtha<strong>la</strong>micnuc<strong>le</strong>us stimu<strong>la</strong>tion. Lancet 345:91-952. The Deep-Brain Stimu<strong>la</strong>tion for Parkinson’s Disease StudyGroup (2001) Deep brain stimu<strong>la</strong>tion of subtha<strong>la</strong>mic nuc<strong>le</strong>usor pars interna of the globus pallidus in Parkinson’s <strong>di</strong>sease. NEngl J Med 345:956–9633. Egi<strong>di</strong> M, Rampini P, Locatelli M et al (2001) Direct MRI localisationof the subtha<strong>la</strong>mic nuc<strong>le</strong>us: a multip<strong>le</strong>, sequential imagefusiontechnique. J Neurol 248 [Suppl 2]:79


Neurol Sci (2002) 23:S111–S112 © Springer-Ver<strong>la</strong>g 2002Deep brain stimu<strong>la</strong>tion of thesubtha<strong>la</strong>mic nuc<strong>le</strong>us in Parkinson’s<strong>di</strong>sease: long-term follow-upA. Tavel<strong>la</strong> 1 () • B. Bergamasco 1,2 • E. Bosticco 1M. Lanotte 3 • P. Perozzo 1 • M. Rizzone 1 • E. Torre 1L. Lopiano 11 Department of Neuroscience, University of Turin, Via Cherasco15, I-10126 Turin, Italy2 Salvatore Maugeri Foundation, IRCCS, Pavia, Italy3 Department of Neurosurgery, CTO Hospital, Turin, ItalyAbstract Deep brain stimu<strong>la</strong>tion (DBS) of the subtha<strong>la</strong>micnuc<strong>le</strong>us (STN) has been shown to be an effective therapy forthe treatment of advanced Parkinson’s <strong>di</strong>sease (PD). Fortysevenpatients were bi<strong>la</strong>terally imp<strong>la</strong>nted for STN DBS andclinically evaluated accor<strong>di</strong>ng to the Core AssessmentProgram for Intracerebral Transp<strong>la</strong>ntations before surgery and3, 12 and 24 months after surgery. E<strong>le</strong>ctrical stimu<strong>la</strong>tion <strong>le</strong>d toa significant improvement in motor symptoms and in the qualityof life, allowing a significant reduction of dopaminergicdrugs with a consequent improvement of drug-induced dyskinesias.Statistical <strong>di</strong>fferences were observed between UPDRSparts II, III and IV values and daily <strong>le</strong>vodopa dosage in thepre- and posto<strong>per</strong>ative <strong>per</strong>iods, whi<strong>le</strong> no <strong>di</strong>fferences were evidentbetween the 3 posto<strong>per</strong>ative con<strong>di</strong>tions.Deep brain stimu<strong>la</strong>tion (DBS) of the subtha<strong>la</strong>mic nuc<strong>le</strong>us(STN) is effective for the <strong>contro</strong>l of the main motor symptomsand of drug-re<strong>la</strong>ted dyskinesias in advancedParkinson’s <strong>di</strong>sease (PD) [1–4]. The aim of this study was toevaluate the effectiveness of high frequency e<strong>le</strong>ctrical stimu<strong>la</strong>tionin 47 patients (18 women and 29 men) affected byi<strong>di</strong>opathic PD, who underwent surgery for the imp<strong>la</strong>ntationof a bi<strong>la</strong>teral stimu<strong>la</strong>ting system of STN.The mean age of the patients was 62.8 years, the meanduration of the <strong>di</strong>sease was 15.6 years, the average durationof <strong>le</strong>vodopa therapy was 14.5 years, and the mean score onthe Hoehn and Yahr sca<strong>le</strong> in off phase was 4. A clinical evaluationof the stimu<strong>la</strong>tion effectiveness was <strong>per</strong>formed in 39patients at the 3-month follow-up, in 21 patients at 1 year an<strong>di</strong>n 7 patients at 2 years after surgery accor<strong>di</strong>ng to the CoreAssessment Program for Intracerebral Transp<strong>la</strong>ntations(CAPIT) [5, 6]. The patients were evaluated in the preo<strong>per</strong>ative<strong>per</strong>iod in the off phase (med off) after a 12-hour withdrawalof all anti<strong>parkinson</strong>ian drugs and after the administrationof a supramaximal dose of <strong>le</strong>vodopa (150–400 mg) (medon). After surgery the clinical assessment was <strong>per</strong>formed infour <strong>di</strong>fferent con<strong>di</strong>tions: stim on-med off, stim off-med off,stim off-med on (with the same dose of <strong>le</strong>vodopa) and stimon-med on. The statistical analysis was <strong>per</strong>formed using theANOVA test, with a <strong>le</strong>vel of significance of p=0.05.The analysis of the stim on-med off con<strong>di</strong>tion showedthat STN DBS was responsib<strong>le</strong> for a 58% improvement inthe UPDRS part II score (activities of daily life) at 3-monthfollow-up, 57% at 1 year and 55% at 2 years with respect tothe preo<strong>per</strong>ative med off con<strong>di</strong>tion. The <strong>di</strong>fferences betweenthe preo<strong>per</strong>ative UPDRS part II scores and the correspon<strong>di</strong>ngposto<strong>per</strong>ative values were significant (p


S112The mean daily <strong>le</strong>vodopa dosage taken by patients olderthan 60 years was 30% greater at the 3-month follow-up and40% greater at 1 year. However, these <strong>di</strong>fferences were notsignificant. In the same way no <strong>di</strong>fferences in the voltagevalues utilized in the two groups were observed in the imme<strong>di</strong>ateposto<strong>per</strong>ative <strong>per</strong>iod or during the follow-up.Another analysis concerned the mo<strong>di</strong>fication of theUPDRS III score in off phase after surgery (21 patients in thepreo<strong>per</strong>ative <strong>per</strong>iod and 3 and 12 months after the surgicaltreatment). There was a slight increment in the mean value ofthe motor score from 59/108 in preo<strong>per</strong>ative med off con<strong>di</strong>tionto 62.2/108 at 3 months and 64.1/108 at 1 year of follow-up,without any significant <strong>di</strong>fferences.DiscussionOur fin<strong>di</strong>ngs confirm that STN DBS represents an effectivetherapy for advanced PD. The clinical improvement of the<strong>parkinson</strong>ian symptoms was re<strong>le</strong>vant, with a remarkab<strong>le</strong>reduction of the daily <strong>le</strong>vodopa dosage and a progressive <strong>di</strong>sappearanceof drug-induced dyskinesias and motor fluctuations.In ad<strong>di</strong>tion, no <strong>di</strong>fferences between the two groups ofPD patients younger and older than 60 years were observed,suggesting that an age of more than 60 years cannot be, probably,an exclusion criterion for the se<strong>le</strong>ction of patients ascan<strong>di</strong>dates for STN surgery.The analysis of the mo<strong>di</strong>fications of the off motor scoresafter surgery seems to suggest a substantial stability of theclinical picture of the patients. However, the <strong>per</strong>iod analyzedwas undoubtedly very short but the preliminary data at 2 and3 years of follow-up seem to confirm this observation.In our pool of patients the surgical complications werenot re<strong>le</strong>vant, confirming that in se<strong>le</strong>cted PD patients theo<strong>per</strong>atory risk of STN DBS can be acceptab<strong>le</strong> in considerationof the significant motor improvement.References1. Limousin P, Krack P, Pol<strong>la</strong>k P, Benazzouz A, Ardouin C,Hoffmann D, Benabid AL (1998) E<strong>le</strong>ctrical stimu<strong>la</strong>tion of thesubtha<strong>la</strong>mic nuc<strong>le</strong>us in advanced Parkinson’s <strong>di</strong>sease. N EnglJ Med 339(16):1105–11112. Benabid AL, Koudsié A, Benazzouz A, Vercueil L, Fraix V,Chabardes S, LeBas JF, Pol<strong>la</strong>k P (2001) Deep brain stimu<strong>la</strong>tionof the corpus luysi (subtha<strong>la</strong>mic nuc<strong>le</strong>us) and other targetsin Parkinson’s <strong>di</strong>sease. Extension to new in<strong>di</strong>cations suchas dystonia and epi<strong>le</strong>psy. J Neurol 248[Suppl 3]:III/37–III/473. The Deep-Brain Stimu<strong>la</strong>tion for Parkinson’s Disease StudyGroup (2001) Deep-brain stimu<strong>la</strong>tion of the subtha<strong>la</strong>micnuc<strong>le</strong>us or the pars interna of the globus pallidus inParkinson’s <strong>di</strong>sease. N Engl J Med 345(13):956–9634. Lopiano L, Rizzone M, Bergamasco B, Tavel<strong>la</strong> A, Torre E,Perozzo P, Va<strong>le</strong>ntini MC, Lanotte M (2001) Deep brain stimu<strong>la</strong>tionof the subtha<strong>la</strong>mic nuc<strong>le</strong>us: clinical effectiveness andsafety. Neurology 56:552–5545. Langston JW, Widner H, Goetz CG et al (1992) CoreAssessment Program for Intracerebral Transp<strong>la</strong>ntations(CAPIT). Mov Disord 7:2–136. Fahn S, Elton RL, Members of the UPDRS DevelopmentCommittee (1987) The Unified Parkinson’s Disease RatingSca<strong>le</strong>. In: Fahn S, Marsden CD, Calne DB, Goldstein M (eds)Recent developments in Parkinson’s <strong>di</strong>sease, vol 2.Macmil<strong>la</strong>n Healthcare Information, Florham Park, pp153–163, 293–304


Neurol Sci (2002) 23:S113–S114 © Springer-Ver<strong>la</strong>g 2002The ro<strong>le</strong> of somatosensory feedbackin dystonia: a psycophysica<strong>le</strong>valuationM. Tinazzi 1 () • T. Rosso 1 • A. Fiaschi 1G. Gambina 1 • S. Farina 1 • S.M. Fiorio 2 • S.M. Aglioti 21 Department of Neurological and Vision Sciences, Section ofNeurological Rehabilitation, University of Verona, Verona, Italy2 Department of Neurological and Vision Sciences, Section ofHuman Physiology, University of Verona, Verona, Italy andDepartment of Psychology, University of Rome La Sapienza, andIRCCS, Fondazione S. Lucia, Rome, ItalyAbstract Ten patients with i<strong>di</strong>opathic dystonia and twelvehealthy <strong>contro</strong>ls were tested with pairs of non-noxious e<strong>le</strong>ctricalstimuli separated by <strong>di</strong>fferent time intervals. Stimuli weredelivered (i) to the pad of the index finger (same-point con<strong>di</strong>tion),(ii) to the pad and to the base of the index finger (samefingercon<strong>di</strong>tion) and (iii) to the pad of the index and ring fingers(<strong>di</strong>fferent-fingers con<strong>di</strong>tion). Subjects were asked toreport if they <strong>per</strong>ceived sing<strong>le</strong> or doub<strong>le</strong> stimuli in the firstcon<strong>di</strong>tion and synchronous or asynchronous stimuli in the secondand third con<strong>di</strong>tions. STDTs were significantly higher indystonic than <strong>contro</strong>l subjects in all three con<strong>di</strong>tions. Resultsextend current know<strong>le</strong>dge on deficits of somesthetic temporal<strong>di</strong>scrimination in dystonia by showing that temporal deficitsare not influenced by spatial variab<strong>le</strong>s.I<strong>di</strong>opathic dystonia is a poorly understood neurological syndromein which the most dramatic symptoms appear to bemotor in nature. It is <strong>la</strong>rgely believed that a dysfunction ofcortical-striato-tha<strong>la</strong>mo-cortical motor circuits p<strong>la</strong>ys a majorro<strong>le</strong> in the pathophysiology of primary dystonia [1]. Severallines of evidence, however, hint at the involvement of thesomatosensory system in dystonia [2–6]. Using a psychophysica<strong>la</strong>pproach, we recently found that the capabilityto <strong>per</strong>ceive as temporally separate two sequential somestheticstimuli was highly impaired in dystonic patients [2, 3].This result has been confirmed in subsequent stu<strong>di</strong>es onpatients with focal hand dystonia, who <strong>per</strong>formed like normal<strong>contro</strong>ls in a sing<strong>le</strong>-touch, gross localization task thatpurportedly tapped spatial <strong>di</strong>scrimination abilities [4, 5]. It isnot known, however, if there is any interaction between spatia<strong>la</strong>nd temporal deficits observed in dystonia.We tested 10 patients with i<strong>di</strong>opathic dystonia (5women and 5 men; mean age, 39.2 years) and 12 healthysubjects matched for age and sex (6 women and 6 men;mean age, 38 years). In all patients, dystonia involved at<strong>le</strong>ast one up<strong>per</strong> limb (8 patients had generalized dystonia, 1patient had a right-sided writer’s cramp and 1 patient hadsegmental dystonia involving the right arm and the trunk).Motor impairment of the arm was graded by evaluating thearm subscore of the Burke-Fahn-Marsden sca<strong>le</strong>.Each stimulus consisted in a pair of square wave e<strong>le</strong>ctricalpulses, of 0.2 ms duration and intensity three-times the sensorythreshold (non-painful). Surface skin e<strong>le</strong>ctrodes were used.For each hand, three ex<strong>per</strong>imental con<strong>di</strong>tions were stu<strong>di</strong>ed.In the first, a sing<strong>le</strong> e<strong>le</strong>ctrode was positioned on thevo<strong>la</strong>r surface of the pad of the index finger (same-pointcon<strong>di</strong>tion). In the second (same-finger con<strong>di</strong>tion) two e<strong>le</strong>ctrodes,one on the base and one on the pad of the index finger,were used. In the third con<strong>di</strong>tion (<strong>di</strong>fferent-fingers)two e<strong>le</strong>ctrodes, positioned on the pad’s vo<strong>la</strong>r surface of theindex and of the ring finger, were used.In same-point con<strong>di</strong>tion, the shortest temporal interva<strong>la</strong>t which subjects reported two separate stimuli (instead ofa sing<strong>le</strong> stimulus) was taken as threshold value. In thesame-finger and <strong>di</strong>fferent-fingers con<strong>di</strong>tions, the first interva<strong>la</strong>t which subjects judged the two stimuli to be asynchronouswas taken as thresholds value. For each ex<strong>per</strong>imentalcon<strong>di</strong>tion, ascen<strong>di</strong>ng and descen<strong>di</strong>ng temporal <strong>di</strong>scriminationthreshold (ATDT, DTDT) were determined forboth <strong>le</strong>ft and right hands.For each subject, mean threshold values were entered ina 2 (hand: <strong>le</strong>ft and right) x 3 (ex<strong>per</strong>imental con<strong>di</strong>tion: samepoint,same-finger, <strong>di</strong>fferent fingers) ANOVA for mixeddesign, where the group (<strong>contro</strong>ls or dystonic patients) wasthe between-subjects factor. A corre<strong>la</strong>tional analysisbetween STDT values (average of ATDT and DTDT) andmotor impairment of the dystonic hand was carried out bylinear regression analysis.As shown in Figure 1 the factor group was significant,threshold values being much lower in <strong>contro</strong>ls (35.7 ms)than in dystonic patients (107.3 ms). The factor ex<strong>per</strong>imentalcon<strong>di</strong>tion was also significant. Post-hoc comparisons,carried out by means of the Student-Newman-Kuels test,showed that STDT in the <strong>di</strong>fferent-finger con<strong>di</strong>tions (82.6ms) was significantly higher than in the same-finger (66.1ms) and same-point (65.9 ms) con<strong>di</strong>tions, which in turnwere not <strong>di</strong>fferent from each other.There was no significant corre<strong>la</strong>tion between the severityof motor impairment of the dystonic hand and theincrease of temporal <strong>di</strong>scrimination thresholds.DiscussionI<strong>di</strong>opathic dystonia is currently thought of as a model of dysfunctionof neostriatal parts of the basal ganglia and of theirre<strong>la</strong>tionships with tha<strong>la</strong>mic and cortical motor structures [1].Functional connectivity of basal ganglia, however, does notinvolve only cortical motor structures, but also somatosensoryareas. For examp<strong>le</strong>, heavy connections between the puta-


S114Fig. 1. Mean values (over fiveblocks) of ascen<strong>di</strong>ng and descen<strong>di</strong>ngtemporal <strong>di</strong>scrimination thresholds indystonic and <strong>contro</strong>l groups in thethree ex<strong>per</strong>imental con<strong>di</strong>tions. Errorbars in<strong>di</strong>cate standard errors. LH, <strong>le</strong>fthand; RH, right hand; ATDT, ascen<strong>di</strong>ngtemporal <strong>di</strong>scrimination threshold;DTDT, descen<strong>di</strong>ng temporal <strong>di</strong>scriminationthresholdminal portions of the neostriatum and S1 and posterior parietalcortices have been traced [1]. A possib<strong>le</strong> ro<strong>le</strong> for sensoryareas in dystonia is also supported by PET stu<strong>di</strong>es documentingthat, in ad<strong>di</strong>tion to a hy<strong>per</strong>metabolism in the striatum,dystonic patients show a reduced activation of the sensorimotorcortices contra<strong>la</strong>teral to the hand where vibrotacti<strong>le</strong>stimu<strong>la</strong>tion was delivered [6]. Psychophysical stu<strong>di</strong>es inbrain-damaged patients have shown that focal <strong>le</strong>sions notonly of S1 but also of basal ganglia induced significantimpairments in the ex<strong>per</strong>imental task of judging as simultaneousor sequential pairs of tacti<strong>le</strong> stimuli [1]. Deficits ofboth spatial and temporal aspects of somatosensory processinghave been reported recently in focal-hand dystonicpatients. This study not only confirms previous stu<strong>di</strong>esreporting temporal <strong>di</strong>scrimination deficits in i<strong>di</strong>opathic dystoniaand in focal hand-dystonia patients [2-5], but also addssignificantly to previous know<strong>le</strong>dge by showing that temporal<strong>di</strong>scrimination in dystonic patients is <strong>la</strong>rgely independentfrom spatial variab<strong>le</strong>s. Indeed, although <strong>per</strong>formance wassignificantly worse in the <strong>di</strong>fferent-fingers con<strong>di</strong>tions than inthe same-point and same-finger con<strong>di</strong>tions, this figure wascomparab<strong>le</strong> in dystonic patients and <strong>contro</strong>l subjects.Previous stu<strong>di</strong>es in normal subjects reported that reactiontimes for identifying pairs of spatial patterns presented totwo fingers were longer than for patterns presented to onefinger. This effect has been attributed to subject’s limitationsof shifting attention effectively across two spatial locations.Since mechanisms responsib<strong>le</strong> for the advantage of havingtemporal stimuli delivered close in space appear simi<strong>la</strong>rlyspared in patients and <strong>contro</strong>ls, the <strong>di</strong>scrimination deficitobserved in dystonic patients does not appear to interactwith the spatial <strong>di</strong>stance between the loci on the hand, wheretacti<strong>le</strong> stimuli were delivered. The increase of temporal <strong>di</strong>scriminationthresholds observed in dystonic patients was notsignificantly re<strong>la</strong>ted to the severity of motor impairment an<strong>di</strong>t was also observed at the side of the clinically normal hand.This means that dystonic symptoms are not a <strong>di</strong>rect consequenceof these somatosensory abnormalities, but the deficitreported here may contribute at <strong>le</strong>ast partially to the motorimpairment present in dystonia.References1. Berardelli A, Rothwell JC, Hal<strong>le</strong>tt M, Thompson PD,Manfre<strong>di</strong> M, Marsden CD (1998) The pathophysiology of primarydystonia. Brain 121:1195–12122. Tinazzi M, Priori A, Berto<strong>la</strong>si L, Frasson E, Mauguiere F,Fiaschi A (2000) Abnormal central integration of a dualsomatosensory input in dystonia. Evidence for sensory overflow.Brain 123:42–503. Tinazzi M, Frasson E, Berto<strong>la</strong>si L, Fiaschi A, Aglioti S (1999)Temporal <strong>di</strong>scrimination of somesthetic stimuli is impaired indystonic patients. Neuroreport 10:1547–15504. Bara-Jimenez W, Shelton P, Hal<strong>le</strong>tt M (2000) Spatial <strong>di</strong>scriminationin abnormal in focal hand dystonia. Neurology55:1869–18735. Sanger TD, Tarsy D, Pascual-Leone A (2001) Abnormalitiesof spatial and temporal sensory <strong>di</strong>scrimination in writer’scramp. Mov Disord 16:94–996. Tempel LW, Perlmutter JS (1990) Abnormal vibrationinducedcerebral blood flow responses in i<strong>di</strong>opathic dystonia.Brain 113:691–707


Neurol Sci (2002) 23:S115–S116 © Springer-Ver<strong>la</strong>g 2002Combination of two <strong>di</strong>fferentdopamine agonists in themanagement of Parkinson’s <strong>di</strong>seaseF. Stocchi 1,2 () • A. Berardelli 1,2 • L. Vacca 1A. Thomas 3 • M.F. De Pan<strong>di</strong>s 4 • N. Modugno 2M. Va<strong>le</strong>nte 1 • S. Ruggieri 1,21 Department of Neurosciences, La Sapienza University, Via<strong>le</strong>dell’Università 30, I-00185 Rome, Italy2 IRCCS Neuromed, Pozzilli (IS), Italy3 Department of Neuroncology, G. d’Annunzio University, Chieti,Italy4 Vil<strong>la</strong> Margherita Rehabilitation Centre, Benevento, ItalyAbstract An alternative approach to the symptomatic treatmentof <strong>parkinson</strong>ian patients with and without motor fluctuationsis to use dual dopamine agonists. The aim of thisstudy was to investigate the symptomatic effect of administratinga second dopamine agonist to <strong>parkinson</strong>ian patientsalready assuming pramipexo<strong>le</strong> or ropiniro<strong>le</strong>. As the seconddopamine agonist we chose cabergoline, a drug with a longhalf life, whose pharmacological profi<strong>le</strong> <strong>di</strong>ffers from that ofthe newer non-ergot-derived dopamine-receptor agonists. Inthis pilot study we enrol<strong>le</strong>d 27 patients: 21 patients hadmotor fluctuations and were receiving <strong>le</strong>vodopa plus adopamine agonist, and 6 patients without motor fluctuationswere receiving a dopamine agonist without <strong>le</strong>vodopa. Thisopen study shows that dual dopamine agonist therapy (cabergolineplus pramipexo<strong>le</strong> or ropiniro<strong>le</strong>) may be used in thesymptomatic treatment of patients with Parkinson’s <strong>di</strong>seasereceiving therapy with or without <strong>le</strong>vodopa.Different stu<strong>di</strong>es have demonstrated that dopamine agonistsinduce <strong>le</strong>ss motor complications than <strong>le</strong>vodopa in patientswith Parkinson’s <strong>di</strong>sease (PD) [1–3]. Moreover, motor complicationsappear or worsen when <strong>le</strong>vodopa is added to theagonists. Other stu<strong>di</strong>es showed that dyskinesia is moresevere in patients taking higher doses of <strong>le</strong>vodopa.Unfortunately, dopamine agonist monotherapy has a limitedspan of efficacy that varies from 3 to 6 years, and then <strong>le</strong>vodopamust be added. In patients taking <strong>le</strong>vodopa plus adopamine agonist, <strong>le</strong>vodopa dosage must be increased duringthe course of the <strong>di</strong>sease to maintain good <strong>contro</strong>l ofsymptoms. This strategy is often necessary despite the agonistbeing used at fairly high dose. Since dopamine agonistshave <strong>di</strong>fferent receptor bin<strong>di</strong>ng proprieties and <strong>di</strong>fferentpharmacokinetic characteristics, it may be possib<strong>le</strong> that twodopamine agonists induce a better clinical effect than onewith the advantage of a prolonged dopamine agonistmonotherapy or an increased <strong>le</strong>vodopa-sparing effect.Epi<strong>le</strong>psy therapy is an examp<strong>le</strong> of how the combination oftwo drugs with simi<strong>la</strong>r modes of action may be more effectivethan one for the patient. We report the preliminaryresults of a pilot study to determine the usefulness of thecombination of two agonists. Cabergoline was chosen as theagonist to add to ropiniro<strong>le</strong> or pramipexo<strong>le</strong> because of itsunique half life and <strong>di</strong>fferent dopamine receptor bin<strong>di</strong>ngpro<strong>per</strong>ties [4–6].The study was conducted as a three-month prospective,open <strong>la</strong>bel pilot trial in patients with PD. After signing anInstitutional Review Board (IRB)-approved informed consentfrom, 27 patients (16 men) with i<strong>di</strong>opathic PD wereincluded in the study. All the patients reported unsatisfactory<strong>contro</strong>l of their symptoms. The 27 patients were subgroupedaccor<strong>di</strong>ng to their previous therapy. The first groupcomprised 21 patients (12 men) of mean age 60.8±6.8 years,mean <strong>di</strong>sease duration 9.1±4.1; they were treated with adopamine agonist (11 pramipexo<strong>le</strong>, 10 ropiniro<strong>le</strong>) plus <strong>le</strong>vodopa.The mean daily dosage of <strong>le</strong>vodopa was 471.4mg/day (SD, 239.0), and the mean daily dosages ofdopamine agonists were 3.7 mg/day (SD, 0.9) for pramipexo<strong>le</strong>and 17.3 mg/day (SD, 6.1) for ropiniro<strong>le</strong>. These 21patients suffered from motor fluctuations (wearing OFF),and 10 of 21 showed also mild dyskinesia.The second group comprised 6 patients (3 men) treatedwith dopamine agonist monotherapy (3 pramipexo<strong>le</strong>, 3ropiniro<strong>le</strong>); their mean age was 51.7±9.9 years and the mean<strong>di</strong>sease duration was 4.5±1.6. The mean daily dosages ofdopamine agonists were 3.5 mg/day (SD, 0.7) for pramipexo<strong>le</strong>and 22.6 mg/day (SD, 6.8) for ropiniro<strong>le</strong>. These patientshad no motor fluctuations or dyskinesia, and their clinicalcon<strong>di</strong>tions were stab<strong>le</strong> though not optimal.In both subgroups, cabergoline was started at 0.5 mg in asing<strong>le</strong> daily dose and then titrated up to the optimal dose. Theoptimal dose was defined as that ab<strong>le</strong> to induce the bestimprovement in the clinical state without inducing adverseeffects. Levodopa dosage was kept unchanged during thestudy <strong>per</strong>iod. Patients were assessed every two weeks duringthe titration phase and every four weeks once the optimaldose was reached. During the week before entering the study,patients in the subgroup treated with a dopamine agonist plus<strong>le</strong>vodopa were asked to maintain a home-<strong>di</strong>ary, recor<strong>di</strong>ng ina flow-chart the number of hours spent OFF. The motor andfunctional assessment included the Unified Parkinson’s <strong>di</strong>seaserating sca<strong>le</strong> (UPDRS motor score), assessed during theON and OFF states, and the abnormal involuntary movements(AIMS) rating sca<strong>le</strong>. Patients in the subgroup treatedwith a dopamine agonist without <strong>le</strong>vodopa were assessed forthe best motor state using the UPDRS sca<strong>le</strong>. Clinical assessmentswere done by an independent blind observer at baseline,every two weeks thereafter, and four weeks after the


S116optimal treatment was reached. During the <strong>la</strong>st assessment,patients and observers were asked to rate changes in eachsubject’s con<strong>di</strong>tion after cabergoline had been added to therapy;they used the global clinical impression (GCI) sca<strong>le</strong> tose<strong>le</strong>ct among: worse, no change, slightly improved, ormarkedly improved.All data are expressed as means and standard deviations.Student’s paired two-tai<strong>le</strong>d test was used to assess the significanceof changes in continuous or normalized data.In the subgroup of 21 patients receiving a dopamine agonistplus <strong>le</strong>vodopa, when cabergoline was added to therapy(4.04±1.62 mg, administered in 12 patients once a day, and inthe other 9 patients twice a day), the UPDRS motor scoremeasured in the OFF <strong>per</strong>iod decreased from 38.9±12.6 to35.4±10.22 (p


Neurol Sci (2002) 23:S117–S118 © Springer-Ver<strong>la</strong>g 2002PARK6 is a common cause of familial<strong>parkinson</strong>ismE.M. Va<strong>le</strong>nte 1,2 () • F. Brancati 1 • V. Caputo 1E.A. Graham 2 • M.B. Davis 2 • A. Ferraris 1 • M.M.B.Brete<strong>le</strong>r 3 • T. Gasser 4 • V. Bonifati 5 • A.R. Bentivoglio 6G. De Miche<strong>le</strong> 7 • A. Dürr 8 • P. Cortelli 9 • A. Fil<strong>la</strong> 7G. Meco 5 • B.A. Oostra 3 • A. Brice 8 • A. Albanese 10B. Dal<strong>la</strong>picco<strong>la</strong> 1 • N.W. Wood 2 and the EuropeanConsortium on Genetic Susceptibility inParkinson’s Disease1C.S.S. Mendel Institute, Via<strong>le</strong> Regina Margherita 261, I-00198Rome, Italy; 2 Institute of Neurology, London, UK; 3 ErasmusUniversity, Rotterdam, The Nether<strong>la</strong>nds; 4 Ludwig MaximiliansUniversität, Münich, Germany; 5 University “La Sapienza”, Rome,Italy; 6 Catholic University, Rome, Italy; 7 University “Federico II”,Nap<strong>le</strong>s, Italy; 8 INSERM U289, Hôpital de <strong>la</strong> Salpêtrière, Paris,France; 9 University of Modena, Modena, Italy 10 NationalNeurological Institute “Carlo Besta”, Mi<strong>la</strong>n, ItalyAbstract The Parkin gene is responsib<strong>le</strong> for about 50% ofautosomal recessive juveni<strong>le</strong> <strong>parkinson</strong>ism (ARJP) and <strong>le</strong>ssthan 20% of spora<strong>di</strong>c early onset cases. We recently mapped anovel ARJP locus (PARK6) on chromosome 1p. Linkage toPARK6 was confirmed in 8 families from 4 <strong>di</strong>fferent Europeancountries. These families share some clinical features with theEuropean Parkin-positive cases, with a wide range of ages atonset and slow progression. However, features typical of ARJP,such as dystonia and s<strong>le</strong>ep benefit, were not observed, makingthe clinical presentation of <strong>la</strong>te-onset cases in<strong>di</strong>stinguishab<strong>le</strong>from that of i<strong>di</strong>opathic PD. The determination of the smal<strong>le</strong>stregion of homozygosity in one consanguineous family allowedreducing the can<strong>di</strong>date interval to 9 cM. PARK6 appears to bean important locus for ARJP in Europe.Autosomal recessive juveni<strong>le</strong> <strong>parkinson</strong>ism (ARJP) sharesmany clinical features with Parkinson’s <strong>di</strong>sease (PD), but ischaracterized by early onset of symptoms, slow progression,good response to <strong>le</strong>vodopa and early <strong>le</strong>vodopa-induceddyskinesias. Hy<strong>per</strong>ref<strong>le</strong>xia, dystonia at onset and s<strong>le</strong>ep benefitmay be present. Pathological stu<strong>di</strong>es have shown a se<strong>le</strong>ctivedegeneration of dopaminergic neurons in the substantianigra and, usually, the absence of Lewy bo<strong>di</strong>es [1, 2]. A genefor ARJP (Parkin) has been identified on human chromosome6q [3]. The Parkin gene codes for a protein with ubiquitin-ligaseactivity [4]. In a recent study on the preva<strong>le</strong>nceof Parkin in Europe, mutations were detected in about 50%of ARJP families and only 18% of spora<strong>di</strong>c cases with earlyonset <strong>parkinson</strong>ism. Some patients with Parkin mutationshad a <strong>la</strong>ter age of onset (up to 65 years) and were clinicallyin<strong>di</strong>stinguishab<strong>le</strong> from patients with i<strong>di</strong>opathic PD [5].We have recently mapped a novel ARJP locus (PARK6) toa 12.5 cM interval on chromosome 1p35-p36 [6]. The clinicalpresentation was simi<strong>la</strong>r to that of i<strong>di</strong>opathic PD, the only <strong>di</strong>stinctivefeatures being an earlier age at onset (range, 32–48years) and a slower progression of the <strong>di</strong>sease. A third ARJPlocus (PARK7) has also been mapped on chromosome 1p36,25 cM telomeric to PARK6 [7]. To assess the ro<strong>le</strong> of PARK6 inEurope, we stu<strong>di</strong>ed 28 ARJP Parkin-negative families [8]. Of28 families, 8 were British, 6 were Dutch, 6 were Italian, 5were German and 3 were French. All affected in<strong>di</strong>viduals fromthe 28 families were genotyped for 11 microsatellite markersspanning the PARK6 region. The 11 loci are ordered as follows:tel-D1S483-0 cM-D1S199-3.2 cM-D1S2732-0 cM-D1S2828-0 cM-D1S478-0.6 cM-D1S2702-3.6 cM-D1S2734-0 cM-D1S2698-1.6 cM-D1S2674-0.8 cM-D1S2885-2.7 cM-D1S247-cen. Microsatellite markers were PCR-amplified fromgenomic DNA and e<strong>le</strong>ctrophoresed on a capil<strong>la</strong>ry using a 3100DNA Sequencer (ABI Prism). Pairwise LOD scores wereobtained using the MLINK program, assuming equal ma<strong>le</strong>fema<strong>le</strong>recombination rate, autosomal recessive inheritance,reduced penetrance (0.90) and a gene frequency of 0.001.Cumu<strong>la</strong>tive multipoint LOD scores were generated by use ofthe SIMWALK2 program. Marker order and genetic <strong>di</strong>stanceswere based on the Marshfield chromosome 1 genetic map.Eight families generated positive LOD scores at allrecombination fractions (thetas) and the affected members ofeach family shared identical haplotypes. The other familiesgenerated negative LOD scores at all thetas and affecte<strong>di</strong>n<strong>di</strong>viduals had <strong>di</strong>fferent haplotypes across the entire region;these were therefore excluded from further analysis.In the 8 families showing evidence of linkage, the 11markers were genotyped in all avai<strong>la</strong>b<strong>le</strong> unaffected familymembers and haplotypes were phased based on the minimumnumber of recombinants (Fig. 1). Linkage analysis inclu<strong>di</strong>ngunaffected family members gave a cumu<strong>la</strong>tive maximum pairwiseLOD score of 5.39 for marker D1S478. The maximumcumu<strong>la</strong>tive multipoint LOD score obtained on the 8 familiesusing 6 markers (D1S483, D1S478, D1S2734, D1S2674,D1S2885 and D1S247) was 6.29 at locus D1S478.No recombination events were detected at the telomericend of the region. The determination of the smal<strong>le</strong>st regionof homozygosity in consanguineous family IT-GR identifieda recombination event between markers D1S2698 andD1S2674, allowing the refinement of the can<strong>di</strong>date intervalto 9 cM between f<strong>la</strong>nking markers D1S483 and D1S2674.Each family showed a <strong>di</strong>fferent haplotype.Of the 8 PARK6-linked families, 3 were Dutch, 2 wereGerman, 2 were Italian and 1 was British. Mean age at onsetwas 42.1±9.4 years, and at <strong>le</strong>ast 1 of the 2 affected siblingshad onset before the age of 45 years. However, 4 affecte<strong>di</strong>n<strong>di</strong>viduals had an age at onset above 45 years, with the <strong>la</strong>testonset at 68 years. Clinical features were in<strong>di</strong>stinguishab<strong>le</strong>from Parkin-positive European ARJP cases, being characterizedby a mild to moderate <strong>parkinson</strong>ian syndrome withgood response to <strong>le</strong>vodopa and slow progression (mean <strong>di</strong>seaseduration, 12.0±6.9 years). Levodopa-induced dyskinesiasoccurred in 8 (50%) of 16 patients. Dystonia at onset ands<strong>le</strong>ep benefit were not detected. Hy<strong>per</strong>ref<strong>le</strong>xia was observedonly in 2 patients [8].


S118Fig. 1 Pe<strong>di</strong>grees of the 8 linked families with haplotypes spanning the PARK6 regionDiscussionThree loci for ARJP have been identified so far, in<strong>di</strong>catinggenetic heterogeneity [8]. The Parkin gene is responsib<strong>le</strong> forabout 50% of ARJP European cases [5]. We have analyzed 28Parkin-negative ARJP families and detected linkage toPARK6 in 8 families from 4 <strong>di</strong>fferent European countries. Nocommon haplotype could be detected, suggesting independentmutational events. These results in<strong>di</strong>cate that PARK6 is notrestricted to Italy, thus confirming the ro<strong>le</strong> of this locus indetermining ARJP in European Parkin-negative cases. ThePARK6-associated phenotype does not resemb<strong>le</strong> the typicalARJP presentation, as is often seen in Japanese patients [1, 2].The phenotype is more simi<strong>la</strong>r to that described in Parkin-positiveEuropean ARJP patients, with a broader clinical spectrumand range of ages at onset [5]. In the <strong>la</strong>te-onset cases, clinicalfeatures are in<strong>di</strong>stinguishab<strong>le</strong> from i<strong>di</strong>opathic PD. However,no pathological data on PARK6-linked patients are avai<strong>la</strong>b<strong>le</strong>at present. It is still unc<strong>le</strong>ar whether PARK6 shares the sameneuropathological features as Parkin or i<strong>di</strong>opathic PD, or representsa <strong>di</strong>stinct <strong>di</strong>sorder within familial <strong>parkinson</strong>isms.Parental consanguinity was detected only in 1 (IT-GR) of8 families. The 2 affected siblings were homozygous acrossthe entire linked region except for the 3 centromeric markers.This fin<strong>di</strong>ng, which strongly suggests a recombinationbetween markers D1S2698 and D1S2674, allows the refinementof the can<strong>di</strong>date interval from 12.5 to 9 cM.A <strong>la</strong>rge number of genes map within the can<strong>di</strong>date intervalbut none of them is a striking can<strong>di</strong>date for <strong>parkinson</strong>ism.The linked region is still too <strong>la</strong>rge to allow searching forcan<strong>di</strong>date genes. Ad<strong>di</strong>tional work is needed to test a <strong>la</strong>rgergroup of ARJP families for linkage to PARK6, in order tofurther refine the linked interval.Acknow<strong>le</strong>dgements Part of this work is copyrighted by Annals ofNeurology (Wi<strong>le</strong>y-Liss, 2002).References1. Ishikawa A, Tsuji S (1996) Clinical analysis of 17 patients in 12Japanese families with autosomal recessive type juveni<strong>le</strong> <strong>parkinson</strong>ism.Neurology 47:160–1662. Takahashi H, Ohama E, Suzuki S et al (1994) Familial juveni<strong>le</strong><strong>parkinson</strong>ism: clinical and pathological study in a family.Neurology 44:437–4413. Kitada T, Asakawa S, Hattori N et al (1998) Mutations in theparkin gene cause autosomal recessive juveni<strong>le</strong> <strong>parkinson</strong>ism.Nature 392:605–6084. Shimura H, Hattori N Kubo S et al (2000) Familial Parkinson <strong>di</strong>seasegene product, parkin, is a ubiquitin-protein ligase. NatureGenet 25:302–3055. Lücking CB, Dürr A, Bonifati V et al (2000) Association betweenearly-onset Parkinson’s <strong>di</strong>sease and mutations in the parkin gene.New Engl J Med 342:1560–15676. Va<strong>le</strong>nte EM, Bentivoglio AR, Ialongo T et al (2001)Localization of a novel locus for autosomal recessive earlyonset <strong>parkinson</strong>ism (PARK6) on human chromosome 1p35-p36. Am J Hum Genet 68:895–9007. Van Dujin CM, Dekker MCJ, Bonifati V et al (2001) PARK7, anovel locus for autosomal recessive early-onset <strong>parkinson</strong>ism, onchromosome 1p36. Am J Hum Genet 69:629–6348. Va<strong>le</strong>nte EM, Brancati F, Ferraris A et al (2002) PARK6-linked<strong>parkinson</strong>ism occurs in several European families. Ann Neurol51:14–18


Neurol Sci (2002) 23:S119–S120 © Springer-Ver<strong>la</strong>g 2002Evaluation of risk of Parkinson’s<strong>di</strong>sease in a cohort of licensedpesticide usersN. Vanacore 1 () • A. Nappo 2 • M Genti<strong>le</strong> 2A. Brustolin 3 • S. Pa<strong>la</strong>nge 4 • A. Liberati 3S. Di Rezze 1 • G. Caldora 1 • M. Gasparini 1F. Benedetti 5 • V. Bonifati 1 • F. Forastiere 4A. Quercia 3 • G. Meco 11Department of Neurological Sciences, La Sapienza University,Via<strong>le</strong> dell’Università 30, I-00185 Rome, Italy; 2 NeurologicalDivision, Belcol<strong>le</strong> Hospital, Viterbo, Italy; 3 Service of Prevention,Hygiene and Safety in the Work Environment, ASL Viterbo, Italy; 4Department of Epidemiology, ASL RM-E, Rome, Italy; 5 Departmentof Ex<strong>per</strong>imental Me<strong>di</strong>cine, La Sapienza University, Rome, ItalyAbstract In the <strong>la</strong>st two years, the environmental theory on theaetiology of Parkinson <strong>di</strong>sease has acquired new data. From anex<strong>per</strong>imental point of view, a new model of <strong>parkinson</strong>isminduced by rotenone, a <strong>di</strong>ffuse insecticide, has been proposed,and in vitro stu<strong>di</strong>es have provided proof that several pesticidesstimu<strong>la</strong>te the formation of α-synuc<strong>le</strong>in fibrils (one of the principalconstituents of Lewy bo<strong>di</strong>es). Moreover, a meta-analysisof all case-<strong>contro</strong>l stu<strong>di</strong>es so far <strong>per</strong>formed showed a positive,statistically significant association between pesticide exposureand PD. In this context, we are <strong>per</strong>forming a cohort study on5575 licensed pesticide users in the province of Viterbo. After27 years of follow-up, 4788 subjects are still alive. The aim ofthis study is to measure the preva<strong>le</strong>nce of Parkinson’s <strong>di</strong>seasein a <strong>la</strong>rge group of workers with theoretically increased risk.In the early 1980s, the environmental theory on the aetiology ofParkinson’s <strong>di</strong>sease (PD) was <strong>le</strong>nt support by the <strong>di</strong>scovery ofMPTP (1-methyl-4-phenyl-1,2,3,6 tetrahydropyri<strong>di</strong>ne), a potentneurotoxin for the substantia nigra, which caused an extrapyramidalsyndrome in a group of young Californian drug-ad<strong>di</strong>ctsshortly after exposure. This mo<strong>le</strong>cu<strong>le</strong> is chemically simi<strong>la</strong>r toparaquat, a <strong>di</strong>ffuse herbicide, and its toxic metabolite, MPP +,has been sold as a herbicide under the name of cy<strong>per</strong>quat.More recently, an ex<strong>per</strong>imental model of <strong>parkinson</strong>isminduced by rotenone, a <strong>di</strong>ffuse insecticide, has been proposed[1]. Rotenone inhibits comp<strong>le</strong>x I of the mitochondrial respiratorychain. In rats, it induces a nigral degeneration with fibril<strong>la</strong>rcytop<strong>la</strong>smic inclusions simi<strong>la</strong>r to the Lewy bo<strong>di</strong>esobserved in the substantia nigra of PD patients [1].Some pesticides (e.g. <strong>di</strong>ethyl<strong>di</strong>thiocarbamate, rotenone,<strong>di</strong>eldrin and paraquat) stimu<strong>la</strong>te the formation of α-synuc<strong>le</strong>infibrils and induce a conformational change in α-synuc<strong>le</strong>in[2]. α-Synuc<strong>le</strong>in is a protein that accumu<strong>la</strong>tes in Lewy bo<strong>di</strong>es;a mutation in its gene (A<strong>la</strong>53Thr) is responsib<strong>le</strong> for amonogenic form of PD.In the <strong>la</strong>st twenty years, numerous case-<strong>contro</strong>l stu<strong>di</strong>eshave been published on the association between occupationalpesticide exposure and PD (Fig. 1) [3]. A meta-analysis of thesestu<strong>di</strong>es estimated the risk for PD to be approximately twice ashigh for subjects with previous pesticide exposure (OR=1.85;95% CI, 1.31–2.60) compared with the general popu<strong>la</strong>tion [4].However, an overall evaluation of other environmental factorsassociated with PD onset (e.g. rural living, well-water drinking,farming) in<strong>di</strong>cated that it is likely that all peop<strong>le</strong> living in rura<strong>la</strong>reas have an increased risk of PD [4]. Moreover, in a study ofa cohort of orchar<strong>di</strong>sts, a high preva<strong>le</strong>nce ratio (PR) for <strong>parkinson</strong>ismwas found in those exposed to pesticides for more than50 years (PR=2; 95% CI, 1.0–4.2) [5].The fact that epidemiological evidence for associationbetween pesticide exposure and PD is supported by previousex<strong>per</strong>imental data suggests that this theory is biologicallyp<strong>la</strong>usibi<strong>le</strong>. This is a crucial criterium in the evaluation of apossib<strong>le</strong> association between exposure and <strong>di</strong>sease in way ofa cause-effect re<strong>la</strong>tionship. Neverthe<strong>le</strong>ss, no specific pesticidehas yet been associated with <strong>di</strong>sease onset in <strong>parkinson</strong>ianpatients, even if it is possib<strong>le</strong> that several pesticidesinduce the typical dopaminergic degeneration of PD througha <strong>di</strong>fferent mechanism of action.The methodological aspects of case-<strong>contro</strong>l stu<strong>di</strong>es <strong>per</strong>formedon PD patients must, however, be evaluated carefully.The results of a case-<strong>contro</strong>l study may in fact be influencedby the clinical definition of cases, the ascertainment ofcases, the recall bias in elderly subjects, the definition ofexposure and the se<strong>le</strong>ction of appropriate <strong>contro</strong>ls. Thesepotential sources of variability should, therefore, be considered.In this context, we propose another epidemiologica<strong>la</strong>pproach which is simi<strong>la</strong>r to the cohort study but <strong>per</strong>mits abetter c<strong>la</strong>rification of the re<strong>la</strong>tionship between exposure and<strong>di</strong>sease. In epidemiological research on PD, this study designhas recently been recommended by the National Institute ofEnvironmental Health Sciences (NIEHS) [6].We identified a cohort of licensed pesticide usersbetween 1971 and 1973 in the province of Viterbo, a townnear Rome. This cohort included ma<strong>le</strong> farmers (n=2977) andtheir wives (n=2598), for a total of 5575 subjects. On 31December 2000, after 27 years of follow-up (1974–2000),4788 subjects were still alive, 757 were deceased (inclu<strong>di</strong>ng7 for whom the specific cause on death certificate was missing),and 28 were lost to follow-up. In accordance with theInternational C<strong>la</strong>ssification of Diseases (ICD, IX Revision),the specific causes of death (ICD code) were: 326 malignantneop<strong>la</strong>sms (140–208), 16 <strong>di</strong>seases of the nervous system(320–359), 215 <strong>di</strong>seases of the circu<strong>la</strong>tory system(390–459), 23 <strong>di</strong>sease of the respiratory system (460–519),48 <strong>di</strong>seases of the <strong>di</strong>gestive system (520–579), 11 <strong>di</strong>seases ofthe genitourinary system (580–629), 14 cases of symptoms,signs and ill-defined con<strong>di</strong>tions (780–799), 56 cases ofinjury and poisoning (800–999), and 42 infectious <strong>di</strong>seases(209–319, 629–780).The 16 specific <strong>di</strong>seases of the nervous system comprised:1 bacterial meningitis (320.9), 2 encephalitis-myelitis (323.9),


S120Fig. 1 Association between pesticide exposure andParkinson’s <strong>di</strong>sease. Case-<strong>contro</strong>l stu<strong>di</strong>es. i, exposure toinsecticides; h, exposure to herbicidesODDS ratio5 Alzheimer’s <strong>di</strong>sease (331.0), 2 unspecified cerebral degeneration(331.9), 2 Parkinson’s <strong>di</strong>sease (332.0), 1 motor neuron<strong>di</strong>sease (335.2), 1 epi<strong>le</strong>psy (345.9), 1 unspecified encephalopathy(348.3), and 1 progressive muscu<strong>la</strong>r dystrophy (359.1).To evaluate the risk of PD in this cohort, we are carryingout a study with a two-phase design: in the first, a questionnairefor the screening of <strong>parkinson</strong>isms, previously validated on theItalian popu<strong>la</strong>tion [7], is mai<strong>le</strong>d to all subjects in the cohort; inthe second phase, all subjects with at <strong>le</strong>ast one positive item inthe questionnaire are visited for a clinical <strong>di</strong>agnosis of PDaccor<strong>di</strong>ng to the United Kingdom PD Brain Bank criteria. Theratio between the numbers of observed and expected PD casesin this cohort is the standar<strong>di</strong>zed preva<strong>le</strong>nce ratio (SPR) andmay be interpreted as a new index of the association betweenpesticide exposure and PD.In conclusion, future epidemiological research on PDwill probably be conducted in the following three ways:1. Cohort stu<strong>di</strong>es on workers exposed to neurotoxins (e.g.solvents, metals, pesticides) that must consider as outcomenot the death certificate, which does not accuratelyestimate the frequency of neurodegenerative <strong>di</strong>seases,but rather the SPR or a simi<strong>la</strong>r index;2. Stu<strong>di</strong>es on the re<strong>la</strong>tionship between genetic and environmentalfactors that consider the data on <strong>di</strong>fferences in thegenotype profi<strong>le</strong> of detoxificant enzymes between PDpatients and the general popu<strong>la</strong>tion (NAT2, GSTT1,CYP2D6) [8, 9];3. Follow-up stu<strong>di</strong>es on subjects with previous, acute pesticideintoxication to evaluate the possib<strong>le</strong> presence ofneurological seque<strong>la</strong>e [10].Acknow<strong>le</strong>dgements Part of this study was supported by grants from theMinistry of Labour and MURST to G. Meco. We thank Lewis Baker forhaving reviewed the English and Capitta Clotilde, Castel<strong>la</strong>ni Giovanni,Mangiabene Maria Teresa, Notarangelo Cecilia, Scrocchia I<strong>la</strong>ria andSor<strong>di</strong>ni Annarita for the having administered the questionnaire.References1. Betarbet R, Sherer TB, MacKenzie G et al (2000) Chronicsystemic pesticide exposure reproduces features ofParkinson’s <strong>di</strong>sease. Nat Neurosci 3:1301–13062. Uversky VN, Li J, Fink AL (2001) Pesticides <strong>di</strong>rectly acce<strong>le</strong>ratethe rate of α-synuc<strong>le</strong>in fibril formation: a possib<strong>le</strong> factorin Parkinson’s <strong>di</strong>sease. FEBS Lett 500:105–1083. Vanacore N (2001) Epidemiologia dei <strong>parkinson</strong>ismi. In:Colosimo C (ed) La ma<strong>la</strong>ttia <strong>di</strong> Parkinson e i <strong>di</strong>sturbi delmovimento. CIC E<strong>di</strong>zioni Internazionali, Rome, pp 1–204. Priyadarshi A, Khuder SA, Schaub EA, Priyadarshi SS (2001)Environmental risk factors and Parkinson’s <strong>di</strong>sease: a metaanalysis.Environ Res A86:122–1275. Engel LS, Checkoway H, Keifer MC et al (2001)Parkinsonism and occupational exposure to pesticides. OccupEnviron Med 58:582–5896. Booker SM (2001) Optimism <strong>per</strong>vades Parkinson’s conference.Environ Health Persp 109:A580–A5817. Pramstal<strong>le</strong>r PP, Falk M, Schoenhuber R, Poewe W (1999)Validation of a mail questionnaire for <strong>parkinson</strong>ism in two<strong>la</strong>nguage (German and Italian). J Neurol 246:79–868. Tan EK, Khajavi M, Thornby JI et al (2000) Variability andvali<strong>di</strong>ty of polymorphism association stu<strong>di</strong>es in Parkinson’s<strong>di</strong>sease. Neurology 55:533–5389. McCann SJ, Pond SM, James KM, Le Couter DG (1997) Theassociation between polymophism in the cytochrome P450-2D6 gene and Parkinson’s <strong>di</strong>sease: a case-<strong>contro</strong>l study andmeta-analysis. J Neurol Sci 153:50–5310. Bhatt MH, Elias MA, Manko<strong>di</strong> AK (1999) Acute andreversib<strong>le</strong> <strong>parkinson</strong>ism due to organophosphate pesticideintoxication. Neurology 52:1467–1471 intoxication.Neurology 52:1467–1471


Neurol Sci (2002) 23:S121–S122 © Springer-Ver<strong>la</strong>g 2002Clinical assessment of dysphagia inearly phases of Parkinson’s <strong>di</strong>seaseM.A. Volonté () • M. Porta • G. ComiNeuroscience Department, University of Mi<strong>la</strong>n, IRCCS SanRaffae<strong>le</strong>, Mi<strong>la</strong>n, ItalyAbstract Dysphagia is a frequent symptom in <strong>parkinson</strong>ism,but it is <strong>le</strong>ss commonly reported by patients with i<strong>di</strong>opathicParkinson’s <strong>di</strong>sease (IPD), especially in the earlyphases. Sixty-five patients with IPD were questioned aboutsymptoms of dysphagia and an objective swallowing testwas administered. Reduced swallowing speed for food andcomp<strong>la</strong>ints of food sticking in the throat, wet voice andcough after liquid intake and nocturnal sialorrhea werereported, respectively, by 35%, 20% and 15% of patients.On objective examination, oral-phase (facial, tongue andpa<strong>la</strong>tal muscu<strong>la</strong>ture) abnormalities were found in 70% ofpatients. Lingual transfer movements, mainly propulsion,and pa<strong>la</strong>tal e<strong>le</strong>vation were severely hypokinetic. Wet voiceafter liquid intake and cough ref<strong>le</strong>x after solid/liquid intakewere detected in 40% of patients. On the other hand, severedysphagia with frequent food aspiration and chest infectionsrequiring antibiotics in the <strong>la</strong>st 12 months was notfound; cough ref<strong>le</strong>x was retained in all patients. On thebasis of these results, a regu<strong>la</strong>r assessment on swallowingabilities in patients with IPD is warranted in the clinical settingbecause with simp<strong>le</strong> <strong>di</strong>etary advice and a short rehabilitativetraining, the quality of life in these patients can beimproved.Hoehn-Yahr sca<strong>le</strong> ranged from 1.5 to 3.0.Patients were interviewed about symptoms of dysphagiaby means of a short questionnaire (food, liquid and tab<strong>le</strong>tswallowing <strong>di</strong>fficulties; food sticking in the throat; coughand wet voice after solid/liquid intake; nocturnal sialorrhea;recent history of weight loss or chest infections requiringantibiotics) [5].An objective swallowing test, based on Kennedy et al.’s[6] dysphagia rating sca<strong>le</strong>, was administered. In the 12-itemmo<strong>di</strong>fied dysphagic rating sca<strong>le</strong> (mDRS, Tab<strong>le</strong> 1) each itemis scored from 1 to 5, in which 1 represents the worst <strong>per</strong>formanceand 5 the normal con<strong>di</strong>tion.From the analysis of the questionnaire we obtained thefollowing results: 35% of patients, when questioned, comp<strong>la</strong>inedof occasional liquid and solid dysphagia; 30% ofpatients reported the sensation of food sticking in the throat;20% comp<strong>la</strong>ined of wet voice and cough after liquid intake;15% had nocturnal sialorrhea; 1% ex<strong>per</strong>ienced weight loss;but no chest infections requiring antibiotics in the <strong>la</strong>st 12months were reported.The results from the objective swallowing test were:mean score (SD) of mDRS was 55.3±3.1 (normal score forthe mDRS is 60). No corre<strong>la</strong>tion was found between mDRSscore and age of patients as well as between mDRS and illnessduration. mDRS score showed a trend towards a corre<strong>la</strong>tionwith UPDRS score (R=-0.7) and the corre<strong>la</strong>tion wassignificant (p=0.004). Mouth opening and pa<strong>la</strong>tal e<strong>le</strong>vationwere impaired in 60%, lingual protrusion in 70% of patients.Wet voice after liquid intake was observed in 40% of cases.Cough ref<strong>le</strong>x after liquid intake was found in 40%, whi<strong>le</strong>cough after solid intake was hardly ever observed (5%).Cough ref<strong>le</strong>x was retained in all patients.Dysphagia is a symptom reported by 20%–40% of patientswith i<strong>di</strong>opathic Parkinson’s <strong>di</strong>sease (IPD) when questioned[1], even though it is seldom self-reported by the patientsthemselves. How <strong>di</strong>sabling symptom it appears after 10–14years from the beginning of the <strong>di</strong>sease [2].Using videofluoroscopy, swallowing prob<strong>le</strong>ms havebeen demonstrated up to 100% of IPD patients [3].Dysphagia, especially when unrecognized, p<strong>la</strong>ys a ro<strong>le</strong> inweight loss in IPD [4] and in the recurrence of airway infections.The mean survival from the beginning of an overtdysphagia is about 2–3 years [2]. The aim of this study wasto determine the usefulness of a clinically objective test indetecting dysphagia in IPD patients, even in asymptomaticcases.Sixty-five patients with IPD (36 women, 29 men) werestu<strong>di</strong>ed. Mean age was 66.3±9.1 years (range, 33–81),mean illness duration was 6.6±4.1 years (range, 1–13),mean UPDRS score was 28.2±6.9 (range, 17–38); andTab<strong>le</strong> 1 Mo<strong>di</strong>fied dysphagia rating sca<strong>le</strong> (mDRS), based on thework of Kennedy et al. [6]1. Maintaining lip closure throughout the oral phase2. Mouth opening3. Lateral movements of the tongue during mastication4. Protrusion of the tongue5. Pa<strong>la</strong>tal e<strong>le</strong>vation6. Triggering of the swallow for liquid/solid intake7. Triggering of cough before, during or after liquid intake8. Triggering of cough before, during or after solid intake9. Solid bolus swallowing test (6 times)10. Liquid bolus swallowing test (6 times)11. Voice quality after liquid intake12. Reflux of material through cricopharyngeal sphincter <strong>le</strong>a<strong>di</strong>ngto oral or nasal emission


S122DiscussionOnly 35% of the IPD patients reported subjective dysphagia.However, a higher preva<strong>le</strong>nce of objective <strong>di</strong>sorder was found.The main fin<strong>di</strong>ngs were the abnormalities of the oral phase,either for preva<strong>le</strong>nce (around 70%) or severity. These changes,being corre<strong>la</strong>ted to the UPDRS motor scores, are likely toexpress bradykinesia of oropharyngeal musc<strong>le</strong>s. Lingual protrusionand pa<strong>la</strong>tal e<strong>le</strong>vation were severely hypokinetic,exp<strong>la</strong>ining <strong>di</strong>fficulties in bolus preparation, bolus <strong>di</strong>rectioning,posterior propulsion of the bolus by the tongue and correctopening of the pharynx.Videofluoroscopic data support the evidence thatderangements of the oral phase are greatly responsib<strong>le</strong> fordysphagia in IPD patients. Indeed, oral transit duration isusually longer than normal, whi<strong>le</strong> the duration of the pharyngealphase is only slightly impaired [7].mDRS can supply only in<strong>di</strong>rect information about pharyngea<strong>la</strong>nd esophageal phases. Wet voice and cough afterliquid intake, observed in 40% of patients, can be consideredthe clinical expression of deficient epiglottic positioning andrange of motion and <strong>la</strong>ryngeal <strong>di</strong>smobility. Comp<strong>la</strong>ints ofreduced swallowing speed for food and food sticking in thethroat can be referred to the abnormalities of pharyngoesophagealmotility. Prolonged transit time for solid foodand retained cough ref<strong>le</strong>x may exp<strong>la</strong>in <strong>la</strong>ck of true aspirationand chest infection in our patients. Therefore, dysphagia canbe seen as a frequent but not severe dysfunction in IDPpatients, at <strong>le</strong>ast in the early stage of the <strong>di</strong>sease. Howeverit may interfere with an adequate nutrition [4].In conclusion, monitoring swallowing abilities by a regu<strong>la</strong>rclinical assessment is useful in detecting manifest aswell as si<strong>le</strong>nt swallowing impairments. mDRS is an easyand reliab<strong>le</strong> tool to evaluate mainly the oral and up<strong>per</strong> pharyngealphases of swallowing, which are actually the mostimpaired phases in IPD, also in asymptomatic patients.Since the <strong>di</strong>sorders of the oral phase seem to be the mostresponsive to dopaminergic therapy [8] and amenab<strong>le</strong> toshort rehabilitative training as well as to simp<strong>le</strong> <strong>di</strong>etaryadvice, detecting them by means of an objective swallowingtest could provide a further opportunity to monitor thegeneral health and to improve the quality of life of thesepatients.References1. Bushmann M, Dobmeier SM, Leeker L et al (1989)Swallowing abnormalities and their response to treatment inParkinson’s <strong>di</strong>sease. Neurology 139:1309–13142. Mul<strong>le</strong>r J, Wenning GK, Verny M et al (2001) Progression ofdysarthria and dysphagia in postmortem-confirmed <strong>parkinson</strong>ian<strong>di</strong>sorders. Arch Neurol 58(2):259–2603. Bird MR, Woodward MC, Gibson EM et al (1994)Asymptomatic swallowing <strong>di</strong>sorder in elderly patients withParkinson’s <strong>di</strong>sease: a description of fin<strong>di</strong>ngs on clinica<strong>le</strong>xamination and videofluoroscopy in 16 patients. Age Ageing123:251–2544. Nozaki S, Saito T, Matsumura T et al (1999) Re<strong>la</strong>tionshipbetween weight loss and dysphagia in patients withParkinson’s <strong>di</strong>sease. Rinsho Shinkeigaku 39:1010-10145. C<strong>la</strong>rke CE, Gul<strong>la</strong>ksen E, Macdonald S, Lowe F (1998)Referral criteria for speech and <strong>la</strong>nguage therapy assessmentof dysphagia caused by i<strong>di</strong>opathic Parkinson’s <strong>di</strong>sease. ActaNeurol Scand 97:27–356. Kennedy G, Pring T, Fawcus R (1993) No p<strong>la</strong>ce for motorspeech acts in the assessment of dysphagia? Intelligibility andswallowing <strong>di</strong>fficulties in stroke and Parkinson’s <strong>di</strong>seasepatients. Eur J Disord Commun 28:213–2267. Stroudly J, Walsh M (1991) Ra<strong>di</strong>ological assessment of dysphagiain Parkinson’s <strong>di</strong>sease. Br J Ra<strong>di</strong>ol 64:890–8938. Tison F, Wiart L, Guatterie M et al (1996) Effects of centraldopaminergic stimu<strong>la</strong>tion by apomorphine on swallowing <strong>di</strong>sordersin Parkinson’s <strong>di</strong>sease. Mov Disord 11:729–732


Neurol Sci (2002) 23:S123–S124 © Springer-Ver<strong>la</strong>g 2002ALS-plus: 5 cases of concomitantamyotrophic <strong>la</strong>teral sc<strong>le</strong>rosisand <strong>parkinson</strong>ismS. Zocco<strong>le</strong>l<strong>la</strong> 1 () • G. Pa<strong>la</strong>gano 1 • A. Fraddosio 1I. Russo 1 • E. Ferrannini 1 • L. Ser<strong>le</strong>nga 3F. Maggio 2 • S. Lamberti 1 • G. Iliceto 11 Department of Neurological Sciences, Ospeda<strong>le</strong> Consorzia<strong>le</strong>Policlinico, University of Bari, Piazza Giulio Cesare 11, I-70124Bari, Italy2 Department of Neurology, Ospeda<strong>le</strong> “F. Iaia” Conversano (Bari),Italy3 Department of Neurology, Ospeda<strong>le</strong> “L. Bonomo”, Andria (Bari),ItalyAbstract Accor<strong>di</strong>ng to El Escorial criteria, amyotrophic <strong>la</strong>teralsc<strong>le</strong>rosis (ALS), combined with other neurologic <strong>di</strong>sorders,such as dementia and <strong>parkinson</strong>ism, is defined as ALSplus.These over<strong>la</strong>ping syndromes are extremely rare. Herewe report 5 cases (3 men, 2 women) of ALS-plus; mean ageat the onset of symptoms was 67 years (range, 65-72). In 3patients, motoneuronal signs preceded the onset of <strong>parkinson</strong>iansyndrome. In 4 cases, the clinical picture was characterizedby the preva<strong>le</strong>nce of motoneuronal signs.Parkinsonism was poorly responsive to L-dopa treatment inall patients. The clinical course <strong>di</strong>d not <strong>di</strong>ffer from thatexpected in patients with only ALS. Our clinical observationsand neuropathological reports of nigral neuronal loss inALS patients suggest a common pathogenic mechanismunderlying these <strong>di</strong>sorders.Accor<strong>di</strong>ng to the El-Escorial c<strong>la</strong>ssification [1], amyotrophic<strong>la</strong>teral sc<strong>le</strong>rosis (ALS) combined with other neurologic <strong>di</strong>sorders,such as <strong>parkinson</strong>ism and dementia, is defined as ALSplus.These over<strong>la</strong>ping syndromes are rare outside of the is<strong>le</strong>sof Guam and Kii peninsu<strong>la</strong>. The most common fin<strong>di</strong>ng is ALSassociated with dementia. Clinical and neuropathologicalstu<strong>di</strong>es of patients presenting ALS and <strong>parkinson</strong>ism supportedthe hypothesis of a common pathogenesis [2–6] Here wedescribe 5 patients, observed in Puglia (southern Italy), whodeveloped both ALS and <strong>parkinson</strong>ism.Case 1. A 67-year-old woman presented with progressivedysarthria and dysphagia. After 1 year, the patient becameanarthric and developed weakness and wasting of up<strong>per</strong> limbmusc<strong>le</strong>s; at that time ALS was <strong>di</strong>agnosed. In the followingmonths, dysphagia became extremely severe requiring theapplication of <strong>per</strong>cutaneous endoscopic gastrostomy (PEG).Two years after the onset of neurological symptoms, shebegan to comp<strong>la</strong>in of rest tremor of her right hand, which ina few months involved the remaining limbs. On neurologica<strong>le</strong>xamination, she was anarthric, dysphagic and dyspnoic,with hy<strong>per</strong>active jaw jerk. Up<strong>per</strong> limb examination revea<strong>le</strong>d<strong>di</strong>stal hypotrophy and brisk tendon ref<strong>le</strong>xes, associated withrest tremor and rigi<strong>di</strong>ty. On lower limb examination, therewere rest tremor and rigi<strong>di</strong>ty mainly on the right side, withsparing of strength and trophism. Coor<strong>di</strong>nation and all sensorymodalities were normal. E<strong>le</strong>ctromyography (EMG)revea<strong>le</strong>d a <strong>di</strong>ffuse pattern of chronic denervation with fascicu<strong>la</strong>tions.Magnetic resonance imaging (MRI) of the brainshowed mild cortical atrophy. Treatment with L-dopa,benserazide, and riluzo<strong>le</strong> was started. In the followingmonths, tremor and rigi<strong>di</strong>ty continued to increase despite L-dopa therapy. After five years, the patient is anarthric andseverely tetraparethic and needs subcontinuous positivepressure venti<strong>la</strong>tion.Case 2. A 66-year-old man presented with progressive weaknessof the lower limbs. After 2 years, he began to comp<strong>la</strong>inof rest tremor of his <strong>le</strong>ft up<strong>per</strong> limb, rapidly involving the<strong>le</strong>g. Neurological examination showed rest tremor and rigi<strong>di</strong>tyof <strong>le</strong>ft up<strong>per</strong> and lower limbs; lower limbs were markedlyparaparethic with increased deep tendon ref<strong>le</strong>xes andbi<strong>la</strong>teral Babinski’s sign. Coor<strong>di</strong>nation and all sensorymodalities were preserved. EMG showed <strong>di</strong>ffuse denervationwith fascicu<strong>la</strong>tions in the four limbs; MRI of the brainrevea<strong>le</strong>d some hy<strong>per</strong>intense areas of the deep white matteron T2-weighted images. Levodopa treatment <strong>di</strong>d notimprove the <strong>parkinson</strong>ian symptoms. The neurological con<strong>di</strong>tionprogressively worsened in the following months; thepatient <strong>di</strong>ed 4 years after the onset of motoneuron <strong>di</strong>sease.Case 3. A 65-year-old woman presented musc<strong>le</strong> weaknessand hypotrophy of her <strong>le</strong>ft up<strong>per</strong> limb, progressively involvingthe remaining limbs. Two years <strong>la</strong>ter, rest tremor of the<strong>le</strong>ft hand became evident, associated with mild rigi<strong>di</strong>ty of the<strong>le</strong>ft limbs. In the following few months, tremor involved theright side of the body. In ad<strong>di</strong>tion, neurological examinationshowed musc<strong>le</strong> weakness, hypotrophy and fascicu<strong>la</strong>tions inthe four limbs, associated with hy<strong>per</strong>active deep tendonref<strong>le</strong>xes. Bi<strong>la</strong>teral Hoffmann’s and Babinski’s signs werepresent. Coor<strong>di</strong>nation and all sensory modalities were preserved.EMG revea<strong>le</strong>d denervation with fibril<strong>la</strong>tions in thefour limbs; MRI of the brain showed mild cortical atrophy.Levodopa treatment was ineffective on tremor and rigi<strong>di</strong>ty.The clinical course was rapidly progressive; the patient <strong>di</strong>ed3 years after the onset of symptoms.Case 4. A 65-year-old man presented with start hesitationand freezing on turning on the <strong>le</strong>ft side. Neurological examinationrevea<strong>le</strong>d mild rigi<strong>di</strong>ty and bradykinesia of <strong>le</strong>ft up<strong>per</strong>and lower limbs. L-Dopa treatment was poorly effective on<strong>parkinson</strong>ism. In the following 6 months, he becamedysarthric and dysphagic and developed progressive weak-


S124ness of both up<strong>per</strong> and lower limbs. On examination,dysarthria and dysphagia, with hypotrophy and fascicu<strong>la</strong>tionsof the tongue, were present. He presented marked musc<strong>le</strong>weakness and hypotrophy in the four limbs, associatedwith fascicu<strong>la</strong>tions and brisk deep tendon ref<strong>le</strong>xes.Babinski’s sign was present bi<strong>la</strong>terally. EMG revea<strong>le</strong>d activedenervation with <strong>di</strong>ffuse fascicu<strong>la</strong>tions in all four limbs;MRI of the brain and spinal cord were normal. After oneyear, the patient became wheelchair-bound. The clinicalcourse was rapidly progressive; the patient <strong>di</strong>ed 4 years afterthe onset of symptoms, because of respiratory failure.Case 5. A 73-year-old man presented with a 2-year history of<strong>di</strong>ffuse bradykinesia. Neurological examination revea<strong>le</strong>dbradykinesia and rigi<strong>di</strong>ty in the four limbs, associated withbrisk deep tendon ref<strong>le</strong>xes. In ad<strong>di</strong>tion, fascicu<strong>la</strong>tions andhypotrophy of the <strong>le</strong>ft up<strong>per</strong> limb were evident. EMGrevea<strong>le</strong>d <strong>di</strong>ffuse denervation and fascicu<strong>la</strong>tions. MRI of thebrain showed mild cortical atrophy. L-Dopa and riluzo<strong>le</strong> therapieswere initiated. The clinical course of the patient wasslowly progressive; after one year he developed impairmentof short-term memory. Nearly two years after the <strong>di</strong>agnosisof motor neuron <strong>di</strong>sease and <strong>parkinson</strong>ism, neurologica<strong>le</strong>xamination reveals mild weakness of up<strong>per</strong> limbs, associatedwith marked and <strong>di</strong>ffuse rigi<strong>di</strong>ty and bradykinesia.DiscussionIn the 5 cases here reported, the mean age at the onset ofsymptoms was slighly higher (67 years; range, 65–72) thanthat commonly reported for ALS patients. In 3 of these cases,motoneuronal signs preceded the onset of <strong>parkinson</strong>ism. In 3cases, the <strong>parkinson</strong>ian picture was characterized mainly byrest tremor and rigi<strong>di</strong>ty, whi<strong>le</strong> the other 2 cases presentedwith bradykinesia. Parkinsonism was poorly responsive to L-dopa treatment in all patients. In 4 cases the clinical picturewas characterized by the preva<strong>le</strong>nce of motoneuronal signs;in these patients the clinical course <strong>di</strong>d not <strong>di</strong>ffer from thatusually expected in ALS.The association between ALS and <strong>parkinson</strong>ism, althoughrare, occurred more frequently than expected by chance [2, 3].Epidemiological stu<strong>di</strong>es showed a higher preva<strong>le</strong>nce of <strong>parkinson</strong>ismin family members of ALS patients. [5]. Some authorsreported that ALS is associated with <strong>parkinson</strong>ism in 5% ofcases, during the course of the <strong>di</strong>sease [2]. The mean age at theonset of this over<strong>la</strong>ping syndrome is almost 10 years greaterthan in ALS without <strong>parkinson</strong>ism [2, 4].The most common extrapyramidal signs are bradykinesiaand rigi<strong>di</strong>ty; usually these signs respond poorly to <strong>le</strong>vodopatreatment. The clinical course does not <strong>di</strong>ffer from thatexpected in ALS without <strong>parkinson</strong>ism [4].Neuropathological evidence of neuronal loss, with neurofi<strong>la</strong>mentousand Lewy body inclusions in the basal ganglia,has been demonstrated in some ALS patients [3, 6]. Inad<strong>di</strong>tion, in recent years, positron emission tomography(PET) and sing<strong>le</strong> photon emission computed tomography(SPECT) stu<strong>di</strong>es revea<strong>le</strong>d a progressive midbrain dopaminergicdeficit in ALS patients, even in the absence ofextrapyramidal signs [6, 7].These fin<strong>di</strong>ngs suggest that the <strong>di</strong>sorders share a commonpathogenic mechanism. Mitochondrial dysfunction, oxidativestress and neurotoxic effects of free ra<strong>di</strong>cals have beensupposed to be involved in neuronal death in Parkinson’s<strong>di</strong>sease and ALS [7]. In ad<strong>di</strong>tion, other pathogenic mechanismshave been proposed in these <strong>di</strong>sorders, inclu<strong>di</strong>ng glutamatergicneuroexcitotoxicity, as well as apoptotic neuronaldeath [8]. However, the real pathogenesis still remains to beproved and the occurrence of <strong>parkinson</strong>ism in elderly ALSpatients needs to be determined.References1. Brooks BR (1994) El Escorial World Federation of Neurologycriteria for the <strong>di</strong>agnosis of amyotrophic <strong>la</strong>teral sc<strong>le</strong>rosis.Subcommittee on Motor Neuron Diseases/AmyotrophicLateral Sc<strong>le</strong>rosis Federation of Neurology Research Group onNeuromuscu<strong>la</strong>r Diseases and the El Escorial “Clinical limitsof amyotrophic <strong>la</strong>teral sc<strong>le</strong>rosis” Workshop contributors. JNeurol Sci 124[Suppl]:96–1072. Eisen A, Calne D (1992) Amyotrophic <strong>la</strong>teral sc<strong>le</strong>rosis,Parkinson’s <strong>di</strong>sease and Alzheimer’s <strong>di</strong>sease: phylogenetic<strong>di</strong>sorders of the human neocortex sharing many characteristics.Can J Neurol Sci 19:117–1203. Williams TL, Shaw PJ, Lowe J et al (1995) Parkinsonism inmotor neuron <strong>di</strong>sease; case report and literature review. ActaNeuropathol 89:275–2834. Qureshi AI, Wilmot G, Dthenia B et al (1996) Motor neuron<strong>di</strong>sease with <strong>parkinson</strong>ism. Arch Neurol 53:987–9915. Takahashi H, Snow BJ, Bhatt MH et al (1993) Evidence for adopaminergic deficit in spora<strong>di</strong>c amyotrophic <strong>la</strong>teral sc<strong>le</strong>rosison positron emission scanning. Lancet 342:1016–10186. Desai J, Swash M (1999) Extrapyramidal involvement inamyotrophic <strong>la</strong>teral sc<strong>le</strong>rosis: backward falls and retropulsion.J Neurol Neurosurg Psychiatry 67:214–2167. Beal MF (1998) Mitochondrial dysfunction in neurodegenerative<strong>di</strong>sorders. Biochim Biophys Acta 1366:211–2238. Tatton WG, Chalmers-Redman RM, Ju WY et al (1997)Apoptosis in neurodegenerative <strong>di</strong>sorders: potential for therapy bymo<strong>di</strong>fying gene transcription. J Neural Transm Suppl 49:245–268

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!