22.04.2013 Views

Prof.Dr. Fathy Ahmed Tantawy

Prof.Dr. Fathy Ahmed Tantawy

Prof.Dr. Fathy Ahmed Tantawy

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Zagazig University<br />

Faculty of Medicine<br />

Radiology Department<br />

Imaging of Cerebral Stroke with Different MRI<br />

Techniques<br />

By<br />

Mona Abdel Ghaffar Mohammad Mohammad El-Hariri<br />

Assistant Lecturer of Radiodiagnosis<br />

Faculty of Medicine<br />

Zagazig University<br />

Thesis<br />

Submitted for Partial Fulfillment of M.D. Degree in Radiodiagnosis<br />

Supervisors<br />

<strong>Prof</strong>.<strong>Dr</strong>. <strong>Fathy</strong> <strong>Ahmed</strong> <strong>Tantawy</strong><br />

<strong>Prof</strong>essor of Radiodiagnosis<br />

Faculty of Medicine<br />

Zagazig University<br />

<strong>Prof</strong>.<strong>Dr</strong>. Dalia Nabil Khalifa<br />

<strong>Prof</strong>essor of Radiodiagnosis<br />

Faculty of Medicine<br />

Zagazig University<br />

<strong>Prof</strong>.<strong>Dr</strong>. Magdy Abdel Hamead Aidaros<br />

Assistant <strong>Prof</strong>essor of Neurology<br />

Faculty of Medicine<br />

Zagazig University<br />

Faculty of Medicine<br />

Zagazig University<br />

2009


Acknowledgment<br />

First of all, I would like to thank ALLAH who gives me the<br />

ability to finish this work.<br />

I would like to express my profound and greatest gratitude<br />

and cordial appreciation to <strong>Prof</strong>. <strong>Dr</strong>. <strong>Fathy</strong> <strong>Ahmed</strong> <strong>Tantawy</strong>,<br />

professor of radiodiagnosis, faculty of medicine, Zagazig university<br />

for his kind encouragement , kind supervision, continuous support<br />

and beneficial remarks that helped me too much to accomplish this<br />

work.<br />

I would like to express my deepest respect to <strong>Prof</strong>. <strong>Dr</strong>. Dalia<br />

Nabil Khalifa, professor of radiodiagnosis, faculty of medicine,<br />

Zagazig university for her advices and contact observations<br />

throughout the work. Her encouragement was impetus for<br />

achievement of the current study.<br />

I would like to express my sincere thanking and gratitude to<br />

<strong>Prof</strong>.<strong>Dr</strong>. Magdy Abdel Hamead Aidaros, assistant professor of<br />

Neurology, faculty of medicine, Zagazig university for his help,<br />

outstanding encouragement that made the accomplishment of this<br />

work possible.<br />

I am really grateful to ALL STAFF of Radiodiagnosis<br />

department , Zagazig university for their continuous encouragement<br />

and moral support.<br />

Last, but not least, I would like to record my appreciation and<br />

gratitude to all my patients, without their kind for their kind help and<br />

cooperation.<br />

Mona A. Ghaffar


List of Abbreviations<br />

2D : two-dimensional.<br />

3D TOF : three-dimensional time of- flight.<br />

ACA : Anterior cerebral artery.<br />

ADC: apparent diffusion coefficient<br />

AICA : Anterior inferior cerebellar artery.<br />

AIF : Anterior internal frontal.<br />

AIS : acute ischemic stroke.<br />

ASL : arterial spin labelling technique.<br />

ATP :adenosine triphosphate.<br />

AVM: Arteriovenous malformation.<br />

BBB : blood–brain barrier.<br />

CBF : Cerebral blood flow .<br />

CBV : cerebral blood volume<br />

cc: cubic centimeter.<br />

CE MRA :Contrast-enhanced MRA.<br />

Ch :choline<br />

cm: centimeter.<br />

CNS: central nervous system.<br />

Cr : creatine.<br />

CSF: cerebrospinal fluid.<br />

CSI : Chemical Shift Imaging.<br />

CT : Computed tomography.<br />

DWI : Diffusion-Weighted Image.<br />

EPI : echo planar imaging.<br />

FLAIR : fluid attenuated inversion recovery.<br />

FMD : Fibromuscular dysplasia.<br />

FPol : Frontopolar.<br />

FSE : fast spin echo.<br />

g: gram<br />

GRE : Gradient recalled echo "T2*-weighted images".<br />

h:hour<br />

ICA : Internal Carotid Artery.<br />

ICH : intracranial hemorrhage.<br />

IIP : Inferior internal parietal .<br />

IV : Intravenous.<br />

IVHs : intraventricular hemorrhages.<br />

Kg: kilogram.<br />

III<br />

Abbreviations


III<br />

Abbreviations<br />

MCA : middle cerebral artery .<br />

MI :Myocardial infarction.<br />

MIF: Middle internal frontal.<br />

min: minute<br />

Ml: Mililitre<br />

mm: millimeter.<br />

mmol: milimole.<br />

MRA : magnetic resonance angiography.<br />

MRI: magnetic resonance Imaging.<br />

MRS: Magnetic Resonance spectroscopy<br />

MTT : mean transit time.<br />

NAA:N-acetylaspartate.<br />

Na+/ K+ ATPase: Sodium/Potasiam adenosine triphosphatase.<br />

NCCT: Non Contrast Computed Tomography.<br />

PC : phase-contrast.<br />

PCA : Posterior cerebral artery.<br />

PCT1: post contrast T1.<br />

PD : proton density .<br />

PICA : Posterior inferior cerebellar artery.<br />

PIF : Posterior internal frontal.<br />

ppm : parts per millions.<br />

PWI : perfusion weighted images.<br />

RBCs : Red Blood Cells.<br />

rCBF : relative Cerebral Blood Flow .<br />

rCBV : relative Cerebral Blood volume.<br />

RF pulse: radiofrequency pulse.<br />

ROI region of interest.<br />

S: second.<br />

SAH : Subarachnoid hemorrhage.<br />

SIP : Superior internal parietal.<br />

S/N : signal-noise ratio.<br />

SVS : Single Voxel Spectroscopy.<br />

T: Tesla.<br />

TE : echo time<br />

TGA :Transient global amnesia.<br />

TIA :transient ischemic attack.<br />

TR : repetition time.<br />

TTP : time to peak.<br />

VOI : volume of interest.


Table of contents<br />

I<br />

Table of contents<br />

Introduction----------------------------------------------------------------------1<br />

Aim of the work------------------------------------------------------------------3<br />

Review of literature-------------------------------------------------------------4<br />

Blood Supply of the Brain----------------------------------------------4<br />

MRI Anatomy of the brain--------------------------------------------15<br />

Pathophysiology of stroke---------------------------------------------20<br />

Conventional MRI in Stroke------------------------------------------26<br />

Clinical Application of Diffusion Weighted Imaging in Stroke-38<br />

Clinical Application of Perfusion Weighted Imaging in Stroke-60<br />

Clinical Application of MRA in Stroke-----------------------------68<br />

Clinical Application of MRS in Stroke------------------------------75<br />

Patients and Methods----------------------------------------------------------83<br />

Results----------------------------------------------------------------------------88<br />

Illustrative cases---------------------------------------------------------------118<br />

Discussion-----------------------------------------------------------------------186<br />

Summary and Conclusion---------------------------------------------------211<br />

References----------------------------------------------------------------------215<br />

Arabic summary--------------------------------------------------------------4 -1


Introduction<br />

- 1 -<br />

Introduction<br />

Stroke can be defined as an acute central nervous system injury with<br />

an abrupt onset (Srinivasan et al.,2006).<br />

Stroke is a leading cause of mortality and morbidity in the developed<br />

world. The goals of an imaging evaluation for acute stroke are to establish a<br />

diagnosis as early as possible and to obtain accurate information about the<br />

intracranial vasculature and brain perfusion for guidance in selecting the<br />

appropriate therapy (Srinivasan et al.,2006).<br />

Acute ischemia constitutes approximately 80% of all strokes and is an<br />

important cause of morbidity and mortality (Srinivasan et al.,2006).<br />

Conventional spin-echo MR imaging is more sensitive and more<br />

specific than CT for the detection of acute cerebral ischemia within the first<br />

few hours after the onset of stroke (Srinivasan et al.,2006).<br />

DW imaging is much more sensitive than T2-weighted imaging or<br />

fluid attenuated inversion recovery (FLAIR) imaging for the detection of<br />

stroke during the first 6 hours after symptom onset (Kim et al.,2005).<br />

Diffusion-weighted imaging (DWI) and the apparent diffusion<br />

coefficient (ADC) are of particular interest, because these parameters show<br />

changes in ischemic brain tissue within hours after symptom onset, when no<br />

abnormalities are typically seen on conventional MR images. Acute<br />

ischemic lesions are characterized by a high signal on DWI and a low ADC.<br />

The ADC, a measure of the freedom of water diffusion, is believed to be<br />

low because of a shift of water, within hypoxic brain parenchyma, from the<br />

extracellular to the intracellular compartment, where water diffusion is<br />

relatively restricted (Lansberg et al., 2001).


- 2 -<br />

Introduction<br />

While diffusion-weighted MR imaging is most useful for detecting<br />

irreversibly infarcted tissue, perfusion-weighted imaging may be used to<br />

identify areas of reversible ischemia as well (Srinivasan et al.,2006).<br />

DW MR imaging depicts infarcted tissue within 5 minutes after the<br />

occlusion of the feeding vessel. PW imaging is able to depict hypoperfused<br />

brain tissue around the infarcted core (Karonen et al., 2000). In many<br />

patients with acute ischemic stroke, the volume of hypoperfused tissue on<br />

perfusion-weighted maps is larger than the volume of tissue with decreased<br />

diffusion on diffusion-weighted images. This mismatch between the<br />

volumes of abnormal tissue on perfusion- and diffusion-weighted images<br />

(perfusion-diffusion mismatch) in the same imaging session can be<br />

considered as an estimate of the ischemic penumbra and thus may be a<br />

predictor of potential infarct growth (Liu et al., 2004).<br />

The perfusion-diffusion mismatch can be determined by using<br />

different perfusion parameters, such as relative cerebral blood volume<br />

(rCBV), relative cerebral blood flow (rCBF), and mean transit time (MTT)<br />

(Karonen et al., 2000).<br />

Hydrogen 1 (1H) magnetic resonance (MR) spectroscopy enables<br />

noninvasive quantification of in vivo metabolite concentrations in the brain<br />

(Jansen et al, 2006).<br />

It has become generally accepted that early intracranial hematomas<br />

typically change over time, mainly, but not exclusively, in two important<br />

ways that alter MR signal intensity patterns: 1) the oxygenation state of<br />

hemoglobin changes, and 2) initially intact RBC membranes eventually lyse<br />

(Ebisu et al., 1997)..


Aim of the work<br />

-3-<br />

Aim of the work<br />

The aim of this work is to assess how we can use different MRI<br />

techniques in the evaluation of patients having cerebrovascular stroke.


Normal vascular anatomy<br />

Review of literature<br />

Fig .18(Weir and Abrahams, 2005) Circle of Willis, arteries of the<br />

brain with MRA, (a) axial and (b) lateral projection.<br />

1- anterior cerebral artery.<br />

2- Anterior communicating artery.<br />

3- Basilar artery.<br />

4- Branches( in insula) of the middle cerebral artery.<br />

5- Cavernous portion of the internal carotid artery.<br />

6- Cervical portion of the internal carotid artery.<br />

7- Genu of the middle cerebral artery.<br />

8- Intracranial (supraclinoid) internal carotid artery.<br />

9- Middle cerebral artery.<br />

10- Ophthalmic artery.<br />

11- Petrous portion of the internal carotid artery.<br />

12- Posterior cerebral artery.<br />

13- Posterior cerebral artery in ambient cistern.<br />

14- Posterior cerebral artery in interpeduncular cistern.<br />

15- Posterior communicating artery.<br />

16- Posterior inferior cerebellar artery.<br />

17- Guadrigeminal portion of posterior cerebral artery.<br />

18- Superior cerebellar artery.


Fig.1: Arterial supply of the brain-mid sagittal view (Gazzaniga et al., 2002).<br />

Fig.2: Arterial supply of the brain-lateral view (Gazzaniga et al., 2002).<br />

Fig.3: Arterial supply of the brain-inferior view (Gazzaniga et al., 2002).<br />

Review of literature


Review of literature<br />

Fig.4: Blood vessels and cranial nerves at the base of the brain (Tank, 1999).<br />

Fig.5: Venous drainage of<br />

the brain (Tank, 1999).


Arterial supply<br />

- 4 -<br />

Review of literature<br />

The arterial supply of the CNS is derived from the internal carotid and<br />

vertebral arteries. All arteries enter the surface of the brain are end arteries, that is,<br />

they have no precapillary anastmosis with other arteries and obstruction of these<br />

arteries causes infarct of the supplied territory (Ryan et al, 2004).<br />

Internal carotid artery<br />

Once the carotid artery enters the carotid canal it has a very tortuous course-<br />

six bends in all before its terminal division.<br />

Branches<br />

The cervical portion has no branches. Two small branches each arise from<br />

the petrous and cavernous parts of the internal carotid artery. These are seldom<br />

visible on angiography. These are:<br />

The carotictympanic artery to the ear drum.<br />

The pterygoid artery to the pterygoid canal and plate.<br />

The cavernous artery to the walls of the cavernous sinus.<br />

The meningohypophyseal artery, which supplies the dura of the anterior<br />

cranial fossa and sends branches to the pituitary (Ryan et al, 2004).<br />

The ophthalmic artery is the first supraclinoid branch of the internal carotid<br />

artery (Butler et al, 1999).<br />

The posterior communicating artery runs posteriorly to anastmose with the<br />

posterior cerebral artery (Ryan et al, 2004).<br />

The anterior choroidal artery orginates from the posterior aspect of the internal<br />

carotid artery(Uflacker, 2007).<br />

Blood Supply of the Brain<br />

Striate arteries are perforating arteries to the lentiform and the caudate nuclei and<br />

the internal capsule (Ryan et al, 2004).<br />

The terminal division of the internal carotid artery is into anterior and<br />

middle cerebral arteries (Ryan et al, 2004).


The anterior cerebral artery<br />

- 5 -<br />

Review of literature<br />

The horizontal portion of the anterior cerebral artery arises anteriorly as the<br />

smaller of the two terminal branches of the internal carotid artery. It courses<br />

anteriorly and medially to the interhemispheric fissure. The anterior cerebral artery<br />

gives origin to two groups of small arteries: an inferior and superior group. The<br />

small inferior group of branches supplies the superior surface of the optic nerve<br />

and chiasm and the superior group is formed by the medial striate arteries<br />

including the artery of Heubner.<br />

These are 5 to 10 small arteries that supply anterior hypothalamus, the<br />

septum pellucidum, the medial portion of the anterior commissure, the pillars of<br />

the fornix and the anterior inferior part of the striatum. (Uflacker, 2007).<br />

Anterior communicating artery<br />

The Anterior communicating artery is a very short and communicates both<br />

anterior cerebral arteries in the interhemispheric fissure. It completes the anterior<br />

portion of the circle of Willis (Uflacker, 2007).<br />

Pericallosal artery<br />

It ascends in front of the lamina terminals, between the two hemispheres<br />

along the longitudinal fissure, making a curve around the genu of the corpus<br />

callosum in the pericallosal cistern.<br />

Segments<br />

Infracallosal segment.<br />

Precallosal segment: curved portion of the artery around the genu of the<br />

corpus callosum.<br />

Supracallosal segment follows the upper surface of the corpus callosum.<br />

Branches of the pericallosal artery<br />

*Orbital artery (Frontobasilar) artery supplies the medial basal region of the<br />

frontal lobe, including the gyrus rectus, the medial part of the medial gyrus and the<br />

olfactory bulb and tract.<br />

*Frontopolar artery arises from the infracallosal segment and supplies anterior<br />

portion of the medial and lateral surfaces of the superior frontal gyrus.<br />

*Callosomarginal artery runs over the cingulate gyrus at the cingulated sulcusand


supplys premotor, motor and sensory areas.<br />

Branches<br />

Anterior Internal Frontal Artery.<br />

Middle Internal Frontal Artery.<br />

Posterior Internal Frontal Artery.<br />

Paracentral Artery (Uflacker, 2007).<br />

Cortical Branches of the anterior cerebral artery<br />

Orbitofrontal (OF).<br />

Frontopolar (FPol).<br />

Anterior internal frontal (AIF).<br />

Middle internal frontal (MIF).<br />

Posterior internal frontal (PIF).<br />

Paracentral (PC).<br />

Superior internal parietal (SIP).<br />

Inferior internal parietal (IIP) (Uflacker, 2007).<br />

The middle cerebral artery<br />

- 6 -<br />

Review of literature<br />

This is the largest and most direct of the branches of the internal carotid<br />

artery and is, therefore, the most prone to emblism ((Ryan et al, 2004).<br />

The middle cerebral artery has been divided into three segments: horizontal,<br />

sylvian and cortical. The horizontal segment supplies the basal ganglia, the orbital<br />

surface of the frontal lobe and the temporal lobe. The sylvian segment supplies the<br />

insula. The cortical branches supply the lateral convexities of the cerebrum<br />

(Uflacker, 2007).<br />

Horizontal segment of the middle cerebral artery<br />

The horizontal segment or M1 of the middle cerebral artery begins at the<br />

internal carotid bifurcation, runs laterally in the lateral cerebral fissure, and<br />

terminates entering the sylvian fissure. The junction between the end of the<br />

horizontal segment and the beginning of the insular segment is known as the<br />

"knee" of the middle cerebral artery. The horizontal segment of the middle cerebral<br />

artery bifurcates or trifurcates near the island of Reil in most patients.


The segment M1 presents three main groups of branches:<br />

Lenticulostriate branches.<br />

Orbitofrontal branch.<br />

Anterior temporal arteries.<br />

Sylvian segment of the middle cerebral artery<br />

- 7 -<br />

Review of literature<br />

When passing through the sylvian fissure the several branches of the middle<br />

cerebral artery feed the insula cortex (Uflacker, 2007).<br />

Cortical branches of the middle cerebral artery<br />

The middle cerebral artery supplies the entire superficial lateral surface of<br />

the cerebral hemisphere in the frontal, temporal, parietal and occipital lobes.<br />

Branches:<br />

Orbitofrontal artery.<br />

Prefrontal artery.<br />

Precental artery.<br />

Central arteries.<br />

Anterior parietal artery.<br />

Posterior parietal artery.<br />

Angular artery (terminal artery).<br />

Tempro-occipital artery.<br />

Posterior temporal artery.<br />

Middle temporal artery.<br />

Anterior temporal artery.<br />

Temporal polar artery (Uflacker, 2007).<br />

Vertebrobasilar system<br />

Vertebral artery<br />

The vertebral artery arises from the posterosuperior aspect of the first part of<br />

the subclavian artery.The artery enters the foramen tranversarium of the sixth<br />

cervical vertebra (occasionally the seventh, fifth or fourth) and ascends through the<br />

foramina of the upper six (or seven, five or four) cervical vertebrae. It passes<br />

posterior to the lateral mass of the atlas and enters the skull through the foramen


- 8 -<br />

Review of literature<br />

magnum. It pierces the dura and arachnoid mater and passes anterior to the<br />

medulla until it unites with its fellow at the lower border of the pons.<br />

*In the neck the branches are:<br />

Spinal branches to the vertebrae and the spinal cord.<br />

Muscular branches to the deep muscles of the neck.<br />

*Within the skull the branches are:<br />

Meningeal branch to the cerebellar fossa and falx.<br />

Posterior spinal artery descends as two branches anterior and posterior to<br />

the dorsal roots.<br />

Anterior spinal artery unites with its fellow anterior to the medulla and<br />

descends as a single anterior spinal artery.<br />

Small branches to the medulla oblongata (Ryan et al, 2004).<br />

Posterior inferior cerebellar artery (PICA): The posterior inferior cerebellar<br />

artery (PICA) arises from the vertebral artery as its largest and most distal branch.<br />

The PICA then proceeds to supply the undersurface of the cerebellar hemispheres<br />

(Butler et al, 1999).<br />

PICA terminates as:<br />

The medial branch (inferior vermian artery) to the inferior vermis and<br />

adjacent inferior surface of the cerebellar hemispheres.<br />

The lateral branch to the lateral aspect of the inferior surface of the<br />

cerebellum (Ryan et al, 2004).<br />

Basilar artery<br />

Formed by the union of the two vertebral arteries at the lower border of the<br />

pons, the basilar artery lies close to (but seldom exactly in) the midline, anterior to<br />

pons in the pontine cistern.<br />

The branches of the basilar artery are as follow:<br />

Pontine artery.<br />

Labyrinthine ( internal auditory artery).<br />

Anterior inferior cerebellar artery (AICA). It supplies the anterior and<br />

lateral aspect of the undersurface of the cerebellum.<br />

Superior cerebellar artery, which arises very close to the terminal division of


- 9 -<br />

Review of literature<br />

the basilar artery and runs laterally around the cerebellar peduncle of the<br />

midbrain to the superior surface of the cerebellum which it supplies. This<br />

artery also sends branches to the pons and the pineal gland.<br />

Right and left posterior cerebral arteries-the basilar artery teminates by<br />

dividing into these (Ryan et al, 2004).<br />

The posterior cerebral arteries<br />

The posterior cerebral artery receives the posterior communicating artery of<br />

the internal carotid artery to complete the circle of Willis (Ryan et al, 2004).<br />

Each posterior cerebral artery can be divided into a number of segments.<br />

The P1 or the precommunicating segment, extends from the basilar bifurcation to<br />

the origin of the posterior communicating artery. It lies within the interpeduncular<br />

fossa and thalamic perforating arteries arise from both this P1 segment and from<br />

the posterior communicating artery. The P2 or ambient segment, runs around the<br />

brainstem in the ambient cistern. The P3 segment extends from the quadrigeminal<br />

plate cistern to the calacrine fissure (Butler et al, 1999).<br />

The branches of the posterior cerebral artery are as follow:<br />

Small branches to cerebral peduncle, the posterior thalamus, the medial<br />

geniculate body and the quadrigeminal plate.<br />

Thalamostriate arteries: supply the thalamus and the lentiform nucleus.<br />

Medial and lateral posterior choroidal arteries: supply (with the anterior<br />

choroidal artery) the choroids plexus of this ventricle.<br />

Cortical branches are as follow:<br />

The posterior temporal branch, which supplies the inferior aspect of the<br />

posterior part of the temporal lobe.<br />

The internal occipital branch which passes to the medial aspect of the<br />

occipital lobe and divides terminally into calcarine and parieto-occipital<br />

arteries (Ryan et al, 2004).<br />

Circle of Willis (Circulus Arteriosus of Willis)<br />

Most of the brain is supplied by two internal carotid arteries and the central<br />

anastomosis between them, called the circle of Willis, which connects the internal


- 10 -<br />

Review of literature<br />

carotid arteries to the vertebrobasilar system that supplies the remainder brain. The<br />

circle of Willis is more polygonal than circular. It is located in the cisterna<br />

interpeduncularis surrounding the optic chiasm, the neural infundibular stem of the<br />

hypophysis gland, and other neural structures in the interpeduncular fossa.<br />

Anteriorly the anterior cerebral arteries are joined by the anterior communicating<br />

artery. Posteriorly the basilar artery divides and orginates the two cerebral arteries,<br />

and each artery is joined by the ipsilateral internal carotid by a posterior<br />

communicating artery (Uflacker, 2007).<br />

Venous <strong>Dr</strong>ainage of the brain<br />

The veins draining the central nervous system do not follow the same<br />

course as the arteries that supply it. Generally, venous blood drains the nearest<br />

venous sinus, except in the case of that draining from the deepest structures, which<br />

drain to deep veins. These drain, in turn, to the venous sinuses (Ryan et al, 2004).<br />

Venous sinuses<br />

The venous sinuses of the cranial cavity are blood filled spaces situated<br />

between the layers of the dura matter, they are lined by endothelium. Their walls<br />

are thick and composed of fibrous tissue, they have no muscular tissue. The sinuses<br />

have no valves. They receive tributaries from the brain , the diploe of the skull, the<br />

orbit and the internal ear (Snell, 2008).<br />

The superior sagittal sinus starts anteriorly and runs posteriorly in the midline to<br />

the internal occipital protuberence . Veins enter the sinus obliquely against the<br />

flow of blood (Ryan et al, 2004) Posteriorly the sinus turns to one side, usually the<br />

right, to become the transverse sinus (Ryan et al, 2004).<br />

The inferiorsagittal sinus lies in the free lower margin of the falx cerebri. It joins<br />

the great cerebral vein to form the straight sinus (Snell, 2008).<br />

The straight sinus lies at the junction of the falex cerebri with the tentorium<br />

cerebelli, formed by the union of the inferior sagittal sinus with the great cerebral<br />

vein. It drains into the transverse sinus (Snell, 2008).<br />

The transverse and sigmoid sinuses-the transverse sinuses run right and left from<br />

the confluence of the sinuses to the mastoid bones where they turn inferiorly and


- 11 -<br />

Review of literature<br />

become the sigmoid sinus, which continues at the jugular foramen as the internal<br />

jugular vein (Ryan et al, 2004).<br />

The cavernous sinuses- this sinus is on either side of the pituitary gland connected<br />

across the midline by the intercavernous sinuses. The cavernous sinus receives the<br />

ophthalmic vein, the sphenoid sinus and the superficial middle cerebral vein and<br />

drains via the petrosal sinuses to the sigmoid sinus and the beginning of the<br />

internal jugular vein (Ryan et al, 2004).<br />

The superior and inferior petrosal sinuses which run along the upper and lower<br />

borders of the prtrous part of the temporal bone (Snell, 2008).<br />

Veins of the brain<br />

The veins of the brain have no muscular tissue in their thin walls, and they<br />

possess no valves. They emerge from the brain and drain into the cranial venous<br />

sinuses (Snell, 2007).<br />

Superficial cerebral veins: They drain the nearest dural sinus.<br />

The superior anastomotic vein (of Troland).<br />

The inferior anastomotic vein (of Labbe).<br />

The superficial middle cerebral vein (Ryan et al, 2004).<br />

Deep cerebral veins<br />

Three veins unit just behind the interventricular foramen (of Monoro) to<br />

form the internal cerebral vein. These are:<br />

The choroids vein.<br />

The septal vein.<br />

The thalamostriate vein.<br />

The point of union of these veins is called the venous angle.<br />

The internal cerebral veins of each side unit to from the great cerebral vein.<br />

The great cerebral vein (of Galen) receives the basal veins and posterior fossa<br />

veins and drains to the anterior end of the straight sinus.<br />

The basal vein (of Rosenthal) begins to the anterior perforated substance by the<br />

union of three veins:<br />

The anterior cerebral vein.<br />

The deep middle cerebral vein.


The striate veins from the basal ganglia.<br />

- 12 -<br />

Review of literature<br />

The basal vein of each side passes around the midbrain to join the great cerebral<br />

vein (Ryan et al, 2004).<br />

Veins of the posterior fossa<br />

The anterior pontomesencephalic vein.<br />

The posterior mesencephalic vein.<br />

The precentral cerebellar vein.<br />

The superior vermian vein drains also to the great vein while the inferior vermian<br />

veins (paired) drain to the sagittal sinus (Ryan et al, 2004).


- 13 -<br />

Review of literature


- 14 -<br />

Review of literature


- 15 -<br />

Review of literature<br />

MR sectional anatomy of the brain (Brant and Helms,<br />

2007)<br />

MRI ANATOMY OF THE BRAIN<br />

Normal axial MRI anatomy [figure 6 to 17].


- 16 -<br />

Review of literature


- 17 -<br />

Review of literature


- 18 -<br />

Review of literature


- 19 -<br />

Review of literature


- 20 -<br />

Review of literature<br />

Stroke is and has been the third leading cause of death in most countries<br />

around the world for a very long time (Caplan, 2006).<br />

Although strokes are much more common in people over 65, and many<br />

people believe that strokes only happen to old folks, strokes can occur at any age,<br />

including infancy, childhood, adolescence and early adulthood (Caplan, 2006).<br />

The brain, and every other organ in the body depends on a constant supply<br />

of energy to function normally. Fuel for the brain is carried in the blood. The brain<br />

requires more fuel than any other organ in the body. The two main energy sources<br />

that the brain uses are sugar and oxygen. Oxygen is carried mainly in the<br />

hemoglobin of red blood cells; sugar is carried in the serum of the blood. When a<br />

part of the brain does not receive an adequate supply of the blood, or when the<br />

blood does not carry enough oxygen or sugar, that portion of the brain becomes<br />

unable to perform its normal functions. Stroke is a term that is used to describe<br />

brain injury caused by abnormality of the blood supply to apart of the brain<br />

(Caplan, 2006).<br />

Strokes can be divided into two very broad groups: hemorrhage and<br />

ischemia. Hemorrhage refers to bleeding inside the skull, either into the brain or<br />

into the fluid surrounding the brain. The second major types of strokes are called<br />

ischemia, a term that refers to lack of blood. (Caplan, 2006).Cerebral infarction<br />

accounts for approximately 85% of all strokes (Haaga et al, 2003).<br />

Causes of Stroke:<br />

a) Ischemic Stroke according to (Torbey and selim, 2007)<br />

1)Cardiac embolism<br />

Conditions have been associated with an increased risk of ischemic stroke from<br />

cardioembolism.<br />

Acute myocardial infarction (AMI).<br />

CHF.<br />

PATHOPHYSIOLOGY OF STROKE<br />

Atrial Fibrillation.


Patent foramen ovale and paradoxical emboli.<br />

Valvular disease.<br />

2) Artery to artery embolism<br />

- 21 -<br />

Review of literature<br />

Embolic material is derived from sources distal to the aortic valve in artery<br />

to artery embolism. This material subsequently mobilizes distal to its site of origin.<br />

1) Atheroemboli in large and medium artery atherosclerotic disease.<br />

2) Aortic arch atheromatous disease.<br />

3) Arterial dissection.<br />

4) Arterial thrombus formation in hypercoagulable states.<br />

5) Large vessel arteriopathy associated with sickle cell disease.<br />

6) Cardiothoracic surgery<br />

3) Small artery occlusive disease, lacunar infarction<br />

Lacunar infarcts are small, deep cerebral infarcts, most often encountered<br />

in the setting of hypertension. These infarcts result from occlusion of very small<br />

penetrating branches (Brown et al., 1988).<br />

4) Hypoperfusion and hypoxemia<br />

Some causes of decreased systemic perfusion and hypoxic states include<br />

sepsis, intraoperative hypotension,cardiopulmonary arrest,cardiac arrhythmia,<br />

volume depletion and antihypertensive medication.<br />

5) Thrombotic stroke and hypercoagulable states.<br />

Other Causes<br />

1- Vasospasm as subarchnoid hemorrhage related contribute to ischemic event,<br />

especially with the presence of a distal arterial stenosis (Torbey and selim, 2007).<br />

2- Venous sinus thrombosis (Gonzalez et al., 2006).<br />

3- Migraine (Gonzalez et al., 2006).<br />

b) Hemorrhagic Stroke<br />

1) Intracerebral hemorrhage (Torbey and selim, 2007).<br />

a) Traumatic ( cerebral contusion).<br />

b) Spontaneous (non traumatic).<br />

1- Hypertension.


2- Amyloid angiopathy.<br />

3- Vascular malformations .<br />

4- Intracranial aneurysms.<br />

5- Arterial thrombosis( hemorrhagic transformation).<br />

6- Venous thrombosis (dural sinuses or deep venous system).<br />

7- Coagulopathy<br />

8- Neoplastic as leukemia.<br />

9- Vasculitis (Torbey and selim, 2007).<br />

2) Subarachnoid hemorrhages<br />

a) Arterial aneurysm.<br />

b) Vascular malformations near the brain surface.<br />

c) Bleeding tendancies.<br />

d) Head injury.<br />

3) Subdural hemorrhages<br />

4) Epidural hemorrhages (Caplan, 2006).<br />

Mechanism of ischemia<br />

- 22 -<br />

Review of literature<br />

Focal cerebral ischemia differs from global ischemia. In global ischemia<br />

irreversible neuronal damage occurs after 4–8 min at normal body temperature. In<br />

focal cerebral ischemia collateral vessels almost always provide some degree of<br />

residual blood flow, which may be insufficient to preserve neuronal survival.<br />

Location of arterial occlusion affects the impairment of cerebral function:<br />

Obstruction below the circle of Willis often permits collateral flow through the<br />

anterior or the posterior communicating arteries. Vertebral artery obstruction can<br />

be bypassed through small deep cervical arteries. In obstructions of the cervical<br />

internal carotid artery, limited collateral flow can be provided through external<br />

carotid artery branches such as the periorbital or the ethmoidal arteries. Collateral<br />

flow mainly derives from the arteries of the circle of Willis.<br />

Basically, the loss of oxygen and glucose supply results in the collapse of<br />

cellular energy production with subsequent changes in cellular metabolism,<br />

degradation of cell membranes, and finally necrosis.<br />

The margins of the infarction are usually hyperemic due to activated


- 23 -<br />

Review of literature<br />

meningeal collaterals. The ischemic tissue swells rapidly because of increased<br />

intracellular water content. During ischemia, the arteries first dilate to increase<br />

blood supply to the oligemic tissue, but will subsequently constrict due to ischemic<br />

damage. Reperfusion may then lead to hyperemia due to impaired autoregulative<br />

capacity of the damaged arteries. In prolonged ischemia sludging and endothelial<br />

damage will prevent reperfusion (Kummer and Back, 2006).<br />

Unlike muscle, brain tissue is exquisitely sensitive to ischemia, because of<br />

the absence of neuronal energy stores. In the complete absence of blood flow, the<br />

available energy can maintain neuronal viability for approximately 2–3 minutes<br />

(Srinivasan et al, 2006).<br />

Two main factors dictate whether brain tissue subjected to hypoperfusion<br />

becomes necrotic: the magnitude and the duration of cerebral hypoperfusion. The<br />

former is the most important. Normal cerebral blood flow is 55ml/100g per minute<br />

(Wityk and Llinas, 2006).<br />

It appears that the first result of ischemia is an alteration of the cellular<br />

metabolism from an aerobic to an anaerobic state. This produces increased<br />

intracellular lactate with a concomitant decrease in intracellular ATP. This<br />

decrease in the energy stores of the cell leads to a number of physiological<br />

changes, including decreased pH, increased extracellular potassium, and/ or<br />

increased extracellular neurotransmitters (glutamate) (Sorensen and Reimer,<br />

2000).<br />

With the loss of ATP production, Na + / K + ATPase, an enzyme that is<br />

important to cell homeostasis, fails. This events permits the unbalanced influx of<br />

extracellular calcium and sodium and secondarily, the influx of extracellular water<br />

into cells. This increased intracellular water is termed cytotoxic oedema (Haaga et<br />

al, 2003).<br />

Ischemic penumbra<br />

Brain tissue is exquisitely vulnerable to ischemia. In the complete absence<br />

of blood flow, available energy stores can maintain neuronal activity for only a<br />

couple of minutes. However, in the event of an acute ischemic stroke, the ischemia<br />

is often incomplete, with the injured areas of the brain continuing to receive blood


- 24 -<br />

Review of literature<br />

from uninjured arterial and leptomeningeal collaterals. The result is a central zone<br />

of completely infracted tissue referred to as the "core" surrounded by a peripheral<br />

zone of ischemic but salvageable tissue referred to as the "penumbra' (Sanghvi et<br />

al., 2007).<br />

The ischemic penumbra is defined as functionally impaired but salvageable<br />

ischemic brain tissue surrounding an irreversibly damaged core1 and is the target<br />

of most recen ttreatments for acute stroke (Oppenheim et al, 2001).<br />

Evoked potentials in the peripheral region are abnormal, and the cells have<br />

ceased to function, but this region is potentially salvageable with early<br />

recanalization. The transition from ischemia to irreversible infarction depends on<br />

both the severity and the duration of the diminution of blood flow. (Srinivasan et<br />

al, 2006).<br />

b) Hemorrhagic Stroke<br />

Intracerebral hemorrhage<br />

Bleeding into the brain results from rupture of small blood vessels- the<br />

arterioles and capillaries within the brain substance. Intracerebral hemorrhage is<br />

most often caused by uncontrolled hypertension. The blood usually oozes into the<br />

brain under pressure and forms a localized, blood collection called a hematoma.<br />

Hematoma exert pressure on brain regions adjacent to the collection of blood and<br />

can injure these tissues. Large hemorrhages are often fatal because they increase<br />

pressure within the skull squeezing vital regions within the brain stem (Caplan,<br />

2006).<br />

Subarachnoid hemorrhage<br />

Bleeding into the fluid around the brain is called subarachnoid hemorrhage,<br />

because blood collects and stays under the arachnoid membrane that lies over the<br />

pia mater. Subarachnoid hemorrhages are usually caused by rupture of of an<br />

aneurysm. The artery breaks, spilling blood instantly into the spinal fluid that<br />

circulates around the brain and spinal cord. The sudden release of blood under high<br />

pressure increases the pressure inside the skull and causes sever, sudden onset<br />

headache, often with vomiting. The sudden increase in pressure causes a lapse in<br />

brain function (Caplan, 2006).


- 25 -<br />

Review of literature


1) Ischemic Stroke<br />

- 26 -<br />

Review of literature<br />

Conventional spin-echo MR imaging is more sensitive and more specific<br />

than CT for the detection of acute cerebral ischemia within the first few hours after<br />

the onset of stroke (Srinivasan et al, 2006).<br />

Conventional spin-echo MRI is more sensitive to the accumulation of tissue<br />

water than CT and this increased tissue water content due to edema provides the<br />

signal contrast in ischemic parenchyma on MR imaging. However, the MR image<br />

is often normal in the first 8 hours and gradually becomes more hyperintense<br />

during the first 24 hours (Rima et al., 2003).<br />

Strokes have a characteristic appearance on conventional MRI that varies<br />

with infarct age. Temporal evolution of strokes is typically categorized into<br />

hyperacute (0–6 h), acute (6–24 h), subacute (24 h to approximately 2 weeks), and<br />

chronic stroke (>2 weeks old) (Gonzalez et al., 2006).<br />

Hyperacute Infarction<br />

In the hyperacute period (first 6 h), there is shift of water from the<br />

extracellular to the intracellular space but there may be little increase in overall<br />

tissue water (Gonzalez et al., 2006).<br />

Conventional MRI in Stroke<br />

If imaging is initiated within the first 6 hours, during which time cytotoxic<br />

edema predominates, standard T2- and T1-weighted images are completely normal<br />

in the majority of cases. In a minority of cases careful scrutiny of the T1-weighted<br />

images may show subtle evidence of mass effect (Davis et al, 2003).<br />

In the hyperacute stage of infarct, there is occlusion or slow flow in the<br />

vessels supplying the area of infracted tissue. Within minutes of the infarct, the<br />

signal flow void on T2-weighted images is lost. Increased signal intensity in the<br />

lumen of large and small vessels may be observed on FLAIR images as the only<br />

indication of infarction, a finding that has been called the “hyperintense vessel<br />

sign” or arterial hyperintensity. The physiopathologic basis of this phenomenon


- 27 -<br />

Review of literature<br />

remains unclear. Several hypotheses have been proposed, such as slowly moving<br />

or stationary blood, intraluminal thrombus or embolus, or even retrograde<br />

collateral circulation (Makkat et al., 2002).<br />

Absence of the normal flow void signal is seen in either or both T1- and T2-<br />

weighted scans. This sign is more easily identified in the larger vessels of the<br />

Circle of Willis and the M1 and M2 segments of the middle cerebral artery. When<br />

present in the middle cerebral artery, this sign can be regarded as the MRI<br />

equivalent of the dense middle cerebral artery sign, as seen with CT (Davis et al,<br />

2003).<br />

Gradient recalled echo (GRE) T2*-weighted images can detect an<br />

intraluminal thrombus (deoxyhemoglobin) in hyperacute infarcts as a linear low<br />

signal region of magnetic susceptibility (Gonzalez et al., 2006).<br />

Contrast-enhanced T1-weighted images show arterial enhancement in 50%<br />

of hyperacute strokes. This arterial enhancement is thought to be secondary to slow<br />

flow, collateral flow or hyperperfusion following early recanalization. It may be<br />

detected as early as 2 h after stroke onset and can persist for up to 7 days. During<br />

this period, there is usually no parenchymal enhancement because inadequate<br />

collateral circulation prevents contrast from reaching the infarcted tissue. Rarely,<br />

early parenchymal enhancement may occur when there is early reperfusion or good<br />

collateralization (Gonzalez et al., 2006).<br />

Acute infarction:<br />

By 24 h, as the overall tissue water content increases due to vasogenic<br />

edema following blood–brain barrier disruption, conventional MRI becomes more<br />

sensitive for the detection of parenchymal infarcts (Gonzalez et al., 2006).<br />

Water is also associated with longer T1 and T2 relaxation times than those<br />

for normal brain tissue. Therefore, one effect of cytotoxic oedema on MRI scans is<br />

to increase the T1 and T2 relaxation times of the involved tissues. Although<br />

regions of the brain that are involved with ischemic stroke may be depicted as<br />

areas of relative T1 and T2 prolongation, the changes on a T1-weighted image are<br />

not usually seen as early as the changes on T2-weighted images. In the acute stroke<br />

patient, the earliest findings on conventional MRI scans are regions of increased


signal intensity on long repetition time (TR) images (Haaga et al, 2003).<br />

- 28 -<br />

Review of literature<br />

Signal changes during the first 24 h are best appreciated in the cortical and<br />

deep gray matter. Infarcts in the acute stage usually demonstrate focal or confluent<br />

areas of T2 and FLAIR hyperintensity with sulcal effacement. During this time, the<br />

white matter may be hyperintense, but also may show no abnormality or<br />

demonstrate hypointensity. Proposed etiologies for the subcortical white matter<br />

hypointensity are free radicals, sludging of deoxygenated red blood cells, and iron<br />

deposition. Because cerebrospinal fluid (CSF) is hypointense, FLAIR has<br />

improved detection of small infarctions in brain parenchyma, such as cortex and<br />

periventricular white matter, adjacent to CSF. By 24 h, T2-weighted and FLAIR<br />

weighted images detect 90% of infarcts. An increase in tissue water also leads to<br />

hypointensity on T1-weighted images. However, in the acute period, T1-weighted<br />

images are relatively insensitive at detecting parenchymal changes compared with<br />

T2- weighted images. At 24 h, sensitivity is still only approximately 50%<br />

(Gonzalez et al., 2006).<br />

Cytotoxic oedema can also influence the appearance of brain tissues on<br />

imaging studies by creating regional mass effect. In acute stroke cases involving<br />

the cortex of the brain, stroke-related edema is commonly associated with swelling<br />

of the cortical gyri. Gyral swelling is typically identified on imaging studies by the<br />

observation that the sulci between the gyri are effaced (i.e., narrowed). Regional<br />

sulcal effacement is, therefore, a sign that can be used to detect the changes of<br />

acute stroke (Haaga et al, 2003).<br />

Subacute infarction:<br />

In the subacute phase of infarct (1 day to 2 weeks), the increase in<br />

vasogenic edema results in increased T2 and FLAIR hyperintensity, increased T1<br />

hypointensity, and better definition of the infarction and swelling. The brain<br />

swelling is manifest as gyral thickening, effacement of sulci and cisterns,<br />

effacement of adjacent ventricles, midline shift, and brain herniation. The swelling<br />

reaches a maximum at about 3 days and resolves by 7–10 days. There is increased<br />

T2 and FLAIR signal within the first week that usually persists but there may be<br />

“MR fogging”. MR fogging occurs when the infarcted tissue becomes difficult to


- 29 -<br />

Review of literature<br />

see because it has developed a signal intensity similar to that of normal tissue<br />

(Gonzalez et al., 2006).<br />

The causes of fogging are not clear, but it has been proposed that a decrease<br />

in oedema, accompanied by increase macrophage activity, may be responsible. In<br />

such cases, parenchymal enhancement on T1-weighted images is typically seen<br />

after injection of gadolinium contrast material (Haaga et al, 2003).<br />

In the subacute phase, arterial enhancement peaks at 1–3 days. Large<br />

infarcts will also demonstrate meningeal enhancement that may represent reactive<br />

hyperemia, which peaks at 2–6 days. Arterial and meningeal enhancement both<br />

typically resolve by 1 week. In addition, parenchymal enhancement occurs during<br />

this phase. Gray matter enhancement can appear band-like or gyriform. This is<br />

secondary to disruption of the blood–brain barrier and restored tissue perfusion<br />

from a recanalized occlusion or collateral flow. This parenchymal enhancement<br />

may be visible at 2–3 days but is consistently present at 6 days and persists for 6–8<br />

weeks. Some infarcts, such as watershed and non cortical infarcts may enhance<br />

earlier(Gonzalez et al., 2006).<br />

Chronic Infarcts<br />

After 2 weeks, the mass effect and edema within infarcts decrease and the<br />

parenchyma develops tissue loss and gliosis. During this time, parenchymal<br />

enhancement peaks at 1–4 weeks and then gradually fades. The chronic stage of<br />

infarction is well established by 6 weeks. At this point, necrotic tissue and edema<br />

are resorbed, the gliotic reaction is complete, the blood–brain barrier is intact, and<br />

reperfusion is established . There is no longer parenchymal, meningeal or vascular<br />

enhancement, and the vessels are no longer hyperintense on FLAIR images. There<br />

is tissue loss with ventricular, sulcal, and cisternal enlargement. There is increased<br />

T2 hyperintensity and T1 hypointensity due to increased water content associated<br />

with cystic cavitation .With large middle cerebral artery (MCA) territory<br />

infarctions, there is Wallerian degeneration, characterized by T2 hyperintensity and<br />

tissue loss, of the ipsilateral cortical spinal tract (Gonzalez et al., 2006).<br />

In some cases, areas of gyral increased density and increased signal on T1-<br />

weighted images may appear to reflect mineralization (i.e. calcification) within


areas of cortical necrosis (Haaga et al, 2003).<br />

Finally:<br />

- 30 -<br />

Review of literature<br />

Standard MRI sequences are extremely sensitive to the detection of acute,<br />

subacute and chronic infarcts. However, standard sequences are negative in the<br />

majority of infarcts (within the first 6–8 hours). Sensitivity is increased by the use<br />

of contrast enhancement and FLAIR, but accurate diagnosis of hyperacute<br />

infarction depends on the use of diffusion-weighted imaging (Davis et al, 2003).<br />

2) Hemorrhagic Stroke<br />

Non-traumatic intracranial hemorrhage (ICH) accounts for 10–15% of all<br />

strokes, but up to 25% of more severe strokes. In the hyperacute emergency<br />

assessment (< 6–12 h) computed tomography (CT) is the diagnostic standard and<br />

modality of choice to differentiate between hyperacute ICH and ischemic stroke.<br />

In general, MRI at this stage is considered to be of little value for the diagnosis of<br />

intracerebral or subarachnoidal hemorrhage and many authors claim that the<br />

sensitivity of MRI for detecting hyperacute ICH is poor. While hyperacute ICH is<br />

hyperdense on acute CT scans with progressing time and hematoma degradation,<br />

there is a loss of density and the ICH may appear isodense or hypodense. MRI is<br />

far superior to CT in the subacute and chronic stages especially with regard to<br />

concomitant or underlying pathology.<br />

The ability to detect acute hemorrhage by MRI is related to the oxygen<br />

saturation of hemoglobin and its degradation. As a hematoma ages, the hemoglobin<br />

passes through several forms (oxyhemoglobin, deoxyhemoglobin, and<br />

methemoglobin) prior to red cell lysis and breakdown into ferritin and hemosiderin<br />

(Davis et al, 2003).<br />

The Evolution of MR Signals in ICH<br />

Based on the most mature form of the hemoglobin present in the clot, five<br />

stages of evolving ICH have been described. The hyperacute stage lasts for 24 h,<br />

acute stage for 2–3 days, early subacute stage for 3–7 days, late subacute stage up<br />

to 2 weeks and chronic stage more than 2 weeks. The evolution of an ICH is a<br />

complex and dynamic process and the estimation of the time of onset based on<br />

MRI findings may be inaccurate (Kummer and Back, 2006).


Hyperacute ICH<br />

- 31 -<br />

Review of literature<br />

Hyperacute stroke does not demand a complete diagnostic workup, with<br />

which all the important pathophysiologic aspects of hyperacute ischemic stroke<br />

can be investigated but also differentiation between ischemic stroke and intra-<br />

cranial hemorrhage by the radiologist, which is impossible by clinical means only<br />

(Fiebach and Schellinger, 2003).<br />

It was initially believed that on MRI an ICH of less than 24 h duration could<br />

not be distinguished reliably either from a mass lesion or from an acute infarct,<br />

because of an unspecific pattern of isointensity in T1 and slight hyperintensity on<br />

T2 imparted by the predominance of oxyhemoglobin. However, due to a reduced<br />

oxygen tension at the periphery of the clot, some deoxygenation of hemoglobin<br />

occurs immediately after the bleeding, and several studies have demonstrated that<br />

the resulting susceptibility effects can be visualized very early on T2- and<br />

particularly on T2*-weighted images as a hypointense rim . This finding may be<br />

less apparent or even not apparent at all with low-field systems, or if FSE T2<br />

sequences are used. It was reported that the ability of high field MRI to reliably<br />

detect a spontaneous ICH as early as 20 min after onset and schematized the<br />

appearance of the hyperacute hemorrhage as consisting of three distinct concentric<br />

regions. The center of the hemorrhage appears isointense or heterogeneously<br />

hyperintense on T2- and T2*-weighted images, probably reflecting the presence of<br />

intact oxyhemoglobin. The periphery is hypointense on T2- and T2*-weighted images<br />

reflecting the susceptibility effects due to deoxyhemoglobin, and finally there is an outer<br />

rim that appears hypointense on T1-weighted images and hyperintense on T2- and T2*weighted<br />

images, suggestive of edema (Kummer and Back, 2006).<br />

Acute ICH<br />

In the absence of rebleeding, deoxygenation of the clot is completed in most<br />

cases by 24 h. At that time deoxyhemoglobin becomes the predominant blood<br />

breakdown product in the lesion. Due to its paramagnetic properties and its<br />

compartmentalization within still intact RBCs, deoxyhemoglobin exerts a strong<br />

susceptibility effect, which makes acute hematomas appear distinctly and<br />

characteristically dark on T2 weighted SE images, and more so on T2* weighted


- 32 -<br />

Review of literature<br />

GRE images . On T1-weighted images, acute ICHs still appear isointense or mildly<br />

hypointense, as in the hyperacute stage. The parenchyma surrounding the clot<br />

exhibits a halo due to vasogenic edema and initial inflammatory reaction. The<br />

region of edema has long T1 and T2 relaxation times and appears hypointense on<br />

T1-weighted images and hyperintense on T2-weighted images. Edema is better<br />

shown by T2-weighted SE sequences than by T2* sequences. By the 5th to 12th<br />

day the hyperintense halo from edema begins to regress. These features of acute<br />

ICH last for about 3 days, after which formation of methemoglobin from<br />

deoxyhemoglobin begins and the clot evolves into the subacute stage(Kummer and<br />

Back, 2006).<br />

Early and Late Subacute ICH<br />

The appearance of methemoglobin in the clot marks the transition from the<br />

acute to the subacute phase. Too high or too low oxygen tension is likely to delay<br />

methemoglobin formation and the expected evolutionary changes of ICH. Unlike<br />

deoxyhemoglobin, methemoglobin allows water protons to approach the<br />

paramagnetic ion. This is possible due to a conformational change occurring in the<br />

structure of methemoglobin, which brings the ferric ion to the surface of the<br />

molecule. With this change PEDD interaction becomes possible and leads to the<br />

shortening of T1 that is characteristic of subacute hematomas. Since<br />

methemoglobin is formed initially at the interface of the hematoma with the<br />

parenchyma of the brain, the bright signals on T1-weighted images are observed<br />

initially at the periphery of the clot. Later, the bright signal extends centripetally<br />

until the whole clot becomes hyperintense in T1. This paramagnetic T1 shortening<br />

effect is observed at all field strengths. In the early subacute phase, methemoglobin<br />

is still compartmentalized within red blood cells and hence generates local field<br />

gradients that maintain a T2 shortening effect similar to that induced by<br />

deoxyhemoglobin. Therefore, in the early subacute phase relaxivity effects and<br />

susceptibility effects coexist, i.e. the clot appears bright in T1, and dark in T2 and<br />

especially in T2*. This stage is expected to last for approximately 1 week. As the<br />

clot ages, progressive fragmentation and osmotic phenomena damage RBC<br />

membranes until they eventually lyse. Hemolysis releases methemoglobin into the


- 33 -<br />

Review of literature<br />

extracellular fluid compartment of the hematoma. The resulting dilution of<br />

methemoglobin in the extracellular fluids eliminates the biological field gradients,<br />

and with them the susceptibility phenomena, an effect that is observed initially at<br />

the periphery of the clot. When all methemoglobin is extracellular, the T2-<br />

shortening induced signal loss is no longer observed, so that the late subacute ICH<br />

is predominantly hyperintense in both T1- and T2-weighted images. Focal brain<br />

lesions containing fat, calcium, or high concentrations of proteins can also appear<br />

hyperintense in T1 and T2 and therefore enter in the differential diagnosis of ICH<br />

at this stage (Kummer and Back, 2006).<br />

Chronic ICH<br />

The beginning of the chronic phase is characterized by the appearance of<br />

modified macrophages (microglia), which phagocytose the ferritin and the<br />

hemosiderin formed from the breakdown of extracellular methemoglobin. These<br />

macrophages accumulate at the periphery of the hematoma and form a ring around<br />

the remnants of the clot. Hemosiderin is highly paramagnetic as it has five<br />

unpaired electrons. Its location inside the lysosomes of macrophages is responsible<br />

for the creation of field gradients and of marked T2 shortening. Hemosiderin is<br />

also insoluble in water, which prevents PEDD interaction. In the absence of T1<br />

shortening no bright signals are observed from hemosiderin on T1-weighted<br />

images. However, at the beginning of the chronic stage there may still be free<br />

methemoglobin in the center of the clot giving bright signals on T1-weighted<br />

images. As a result, a chronic ICH initially shows a hypointense peripheral<br />

hemosiderin rim completely surrounding a central area hyperintense in all<br />

sequences due to the persistence of extracellular methemoglobin. The hemosiderin<br />

rim is less conspicuous on T1-weighted sequences and more conspicuous on T2-<br />

and T2*-weighted sequences. With time, the macromolecules in the hematoma<br />

cavity are slowly reabsorbed. This results in a gradual disappearance of<br />

paramagnetic T1 shortening effects. With the elimination of all macromolecular<br />

components, the lesion becomes more fluid and the signal intensity of the inner<br />

part of the clot becomes isointense to CSF in all sequences. Finally, the cyst-like<br />

cavity collapses leaving a residual hemosiderin-containing scar (Kummer and


What<br />

happens<br />

Back, 2006).<br />

- 34 -<br />

Review of literature<br />

Table 1: (Reimer et al., 2006:) Sequential signal intensity (SI) changes of intracranial<br />

haemorrhage on MR imaging (1.5 T).<br />

Hyperacute Acute haemorrhage Early subacute Late subacute Chronic haemorrhage<br />

haemorrhage<br />

haemorrhage haemorrhage<br />

Blood leaves the<br />

vascular system<br />

(extravasation)<br />

Deoxygenation with<br />

formation of deoxy-<br />

Hb<br />

Time frame


- 35 -<br />

Review of literature<br />

CT is also the (imaging) standard of care for the diagnosis of acute<br />

subarachnoid hemorrhage with a sensitivity of 85–100%. The CT-based diagnosis<br />

of SAH may be difficult in the posterior fossa due to bone artefacts as well as with<br />

small amounts of blood. In the subacute stage the sensitivity of CT for SAH<br />

diminishes substantially with sensitivities of 50% after 1 week, 30% after 2 weeks<br />

and ≈0% at 3 weeks necessitating the diagnostic gold standard in SAH, i.e. lumbar<br />

puncture. There currently is only scarce information about the sensitivity of stroke<br />

MRI for SAH. Initial assumptions that MRI may also be suitable in the diagnosis<br />

of acute SAH were derived from in vitro studies (Fiebach and Schellinger, 2003).<br />

CT shows the acute SAH as high attenuation areas filling the cisterns and<br />

sulci (Kummer and Back, 2006).<br />

Conventional T1–WI and T2–WI were shown to be ineffective in the<br />

diagnosis of acute SAH with sensitivities of 35–50%. This may, in part, be due to<br />

the fact that the rules, which apply for intracerebral degradation of ICH, may not<br />

be valid for SAH due to a higher oxygen partial pressure in cerebrospinal fluid<br />

(CSF) and that the formation of blood clots which also causes T2-shortening is<br />

impaired. The paramagnetic susceptibility effect, which may be utilized to detect<br />

intracerebral hemorrhage, is not suitable for the detection of SAH as the latter<br />

frequently is localized in the vicinity of the cranial vault or skull base, thus areas<br />

which are prone to artifacts. The detection of SAH may be improved by using PD–<br />

WI with shorter repetition times (TR) to visualize the T1-effect. FLAIR sequences<br />

have been reported to be suitable for the diagnosis of not only subacute but also<br />

acute SAH (Davis et al, 2003)<br />

In subacute SAH (5days to 2 weeks) MRI with fluid attenuated inversion<br />

recovery (FLAIR) sequences and proton density (PD) weighted images is clearly<br />

superior to CT (sensitivity 100% vs. 45%) (Davis et al, 2003).<br />

In FLAIR an inversion pulse is utilized to null the bright CSF signals in T2-<br />

weighted images with long TE. Due to its high protein concentration and T1<br />

shortening effect, the acute subarachnoid bleeding will appear bright on FLAIR.<br />

While FLAIR may be very sensitive in the detection of acute SAH, its specificity<br />

for this disease is suboptimal. All conditions causing an elevation of CSF proteins,


- 36 -<br />

Review of literature<br />

such as pyogenic meningitis, granulomatous meningitis, meningeal carcinomatosis<br />

etc., characteristically elevate the CSF protein levels and give rise to bright signals<br />

on FLAIR. Fat from ruptured dermoid or previous introduction of oily contrast<br />

media may also mimic SAH on FLAIR due to T1 shortening effects. Therefore, in<br />

order to confidently make a diagnosis of SAH on FLAIR, a careful appraisal of the<br />

clinical history is mandatory. Contrast may be helpful to exclude meningitis, while<br />

fat may be identified using chemical shift artifacts or fat saturation sequences.<br />

Another pitfall with FLAIR sequences may be represented by the CSF pulsation<br />

artifacts from inflow and misregistration effects. These artifacts create bright<br />

signals in the cisterns especially with fast FLAIR sequences. Hence, FLAIR may<br />

not be suitable to assess the posterior fossa SAH. Proton density-weighted images<br />

seem to be as sensitive as FLAIR in detecting SAHs of less than 24 h. CSF flow<br />

artifacts in cisterns and ventricles are less likely to be observed with proton density<br />

than with FLAIR (Kummer and Back, 2006).<br />

Intraventricular Hemorrhage<br />

Most intraventricular hemorrhages (IVHs) are secondary to ICH or SAH.<br />

Isolated IVH is rare and usually due to the rupture of underlying vascular<br />

malformations. Based on MR imaging, intraventricular bleedings have been<br />

classified into clotted IVH, usually unilateral, and layered IVH, usually bilateral.<br />

Since the CSF in ventricles has a higher content in oxygen and glucose, the layered<br />

form evolves more slowly compared to the clotted form. The latter evolves at a rate<br />

similar to ICH. However, in the chronic stage, unlike ICH, IVHs do not show<br />

hemosiderin formation. Among the conventional sequences, proton density depicts<br />

IVH better than T1- and T2-weighted images. In the acute stage FLAIR may have<br />

better sensitivity than CT scan to demonstrate IVH. On FLAIR, in the initial 48 h<br />

IVH appears hyperintense, but one should be aware of the CSF flow artifacts,<br />

which can mimic the presence of blood. Also, conditions like pyocephalus or<br />

intraventricular tumors may exhibit signals similar to IVH. After the acute phase,<br />

IVHs may appear iso- or hypointense on FLAIR. At this stage, a bright signal on<br />

T1-weighted images and a hypointense signal on T2*-weighted image will mark<br />

the presence of an IVH (Kummer and Back, 2006).


- 37 -<br />

Review of literature


Clinical Application of Diffusion Weighted<br />

Imaging in Stroke<br />

- 38 -<br />

Review of literature<br />

Diffusion weighted imaging is an MR imaging technique in which contrast<br />

within the image is based on microscopic motion of water (Rajeshkannan et<br />

al.,2006).<br />

Diffusion-weighted (DW) magnetic resonance (MR) imaging provides<br />

potentially unique information on the viability of brain tissue. It provides image<br />

contrast that is dependent on the molecular motion of water, which may be<br />

substantially altered by disease. Stejskal and Tanner provided an early description<br />

of a DW sequence in 1965. They used a spin-echo T2-weighted pulse sequence<br />

with two extra gradient pulses that were equal in magnitude and opposite in<br />

direction. This sequence enabled the measurement of net water movement in one<br />

direction at a time. To measure the rate of movement along one direction, for<br />

example the x direction, these two extra gradients are equal in magnitude but<br />

opposite in direction for all points at the same x location. However, the strength of<br />

these two balanced gradients increases along the x direction. Therefore, if a voxel<br />

of tissue contains water that has no net movement in the x direction, the two<br />

balanced gradients cancel each other out. The resultant signal intensity of that<br />

voxel is equal to its signal intensity on an image obtained with the same sequence<br />

without the DW gradients. However, if water molecules have a net movement in<br />

the x direction (eg, due to diffusion), they are subjected to the first gradient pulse at<br />

one x location and the second pulse at a different x location. The two gradients are<br />

no longer equal in magnitude and no longer cancel. The difference in gradient<br />

pulse magnitude is proportional to the net displacement in the x direction that<br />

occurs between the two gradient pulses, and faster-moving water protons undergo<br />

a larger net dephasing. The resultant signal intensity of a voxel of tissue containing<br />

moving protons is equal to its signal intensity on a T2-weighted image decreased


- 39 -<br />

Review of literature<br />

by an amount related to the rate of diffusion. The signal intensity (SI) of a voxel<br />

oftissue is calculated as follows:<br />

SI= SI0 x exp (-bx D).<br />

where SI0 is the signal intensity on theT2-weighted (or b = 0 sec/mm2) image,the<br />

diffusion sensitivity factor b is equal<br />

toγ 2 G 2 δ 2 (∆-δ /3), and D is the diffusion coefficient. γ is the gyromagnetic ratio; G<br />

is the magnitude of δ, the width of, and ∆ is the time between the two balanced<br />

DW gradient pulses. When measuring molecular motion with DW imaging, only<br />

the apparent diffusion coefficient (ADC) can be calculated. The signal intensity of<br />

a DW image is best expressed as:<br />

SI = SI0 x exp (-bx3 ADC).<br />

In the brain, apparent diffusion is not isotropic (the same in all directions); it is<br />

anisotropic (varies in different directions),particularly in white matter. The cause<br />

of the anisotropic nature of white matter is not completely understood, but<br />

increasing anisotropy has also been noted in the developing brain before T1-and<br />

T2-weighted imaging or histologic evidence of myelination becomes evident. It is<br />

likely that in addition to axonal direction and myelination, other physiologic<br />

processes, such as axolemmelic flow, extracellular bulk flow, capillary blood flow,<br />

and intracellular streaming, may contribute to white matter anisotropy. The<br />

anisotropic nature of diffusion in the brain can be appreciated by comparing<br />

images obtained with DW gradients applied in three orthogonal directions. In each<br />

of the images, the signal intensity is equal to the signal intensity on echo-planar<br />

T2-weighted images decreased by an amount related to the rate of diffusion in the<br />

direction of the applied gradients. Images obtained with gradient pulses applied in<br />

one direction at a time are combined to create DW images or ADC maps. The<br />

ADC is actually a tensor quantity or a matrix:<br />

ADCxx ADCxy ADCxz<br />

ADC= [ ADCyx ADCyy ADCyz]<br />

ADCzx ADCzy ADCzz<br />

The diagonal elements of this matrix can be combined to give information about<br />

the magnitude of the apparent diffusion:


(ADCxx 1 ADCyy 1 ADCzz)/3 (Schaefer et al, 2000).<br />

- 40 -<br />

Review of literature<br />

b value:<br />

The signal sensitivity to motion for the technique is determined by the b<br />

value. Larger b values can be obtained through larger gradient amplitudes, longer<br />

duration gradient pulses, or longer times between the gradient pulses.<br />

Increasing the b value will increase the amplitude of the diffusion gradients<br />

making the acquisition more sensitive to the diffusion of water in normal brain. As<br />

this occurs, the signal from areas with normal diffusion decreases, increasing the<br />

contrast between areas of normal and restricted diffusion (Westbrook , 1999).<br />

ADC map<br />

A parameter map in which the pixel intensity is equal to the value of ADC.<br />

The map may be obtained from images acquired at several different values of b-<br />

factor. Care must be taken in selecting a minimum b value as flow effects dominate<br />

at very low b. Alternatively, a two-point method may be used typically acquiring a<br />

b = 0 image and a second image at a high b value. ADC maps have proved useful<br />

in diagnosing stroke but do not provide any directional information (Liney, 2005).<br />

Motion artifact in DWI:<br />

All motion artifacts can produce false elevations in apparent diffusion<br />

coefficient values relative to the image in which b = 0. Patient motion, including<br />

gross head motion, cardiac related pulsations, and respiratory motions, can also<br />

produce large phase shifts, which create ghosting artifacts (Provenzale and<br />

Sorensen, 1999).<br />

With the development of fast imaging techniques such as single shot EPI<br />

and half Fourier RARE, it has been possible to suppress motion artifacts even with<br />

large diffusion gradients applied (Davis et al, 2003).<br />

MR Technique used for DWI<br />

With the development of high-performance gradients, DW imaging can be<br />

performed with an echo-planar spin echo T2-weighted sequence. With the original<br />

spin-echo T2-weighted sequence, even minor bulk patient motion was enough to<br />

obscure the much smaller molecular motion of diffusion. The substitution of an<br />

echo-planar spin-echo T2-weighted sequence markedly decreased imaging time


- 41 -<br />

Review of literature<br />

and motion artifacts and increased sensitivity to signal changes due to molecular<br />

motion. As a result, the DW sequence became clinically feasible to perform<br />

(Schaefer et al, 2000).<br />

1) Ischemic Stroke<br />

The early diagnosis of ischemic stroke is critical to the success of<br />

therapeutic interventions such as thrombolysis and anticoagulation (Mullins,<br />

2006).<br />

Magnetic resonance (MR) diffusion imaging allows detection of cerebral<br />

ischemia within minutes of onset, and the temporal evolution of diffusion<br />

characteristics enables differentiation of acute from chronic stroke (Beaucbamp et<br />

al., 1998).<br />

Because diffusion-weighted MR imaging is especially sensitive to shifts in<br />

water that occur between extracellular and intracellular compartments, this<br />

technique can demonstrate regions of brain that are undergoing ischemic injury<br />

within 1 hour or less of symptom onset. In patients with acute ischemic stroke,<br />

intracellular swelling, which occurs as a result of water shifting from the<br />

extracellular compartment, causes the initial increase in observed signal intensity.<br />

Other factors that contribute to the increase in signal intensity are the<br />

increased tortuousity of the extracellular and intracellular spaces and increased<br />

intracellular viscosity (Chen et al, 2006).<br />

Conventional CT and MR imaging cannot reliably detect infarction at early<br />

time points (less than 6 h) (Gonzalez et al., 2006). In the hyperacute stroke period<br />

(0–6 hours after onset of symptoms), CT and MR imaging yield a sensitivity of<br />

less than 50% (Mullins et al, 2002), because detection of hypoattenuation on CT<br />

and hyperintensity on T2-weighted and fluid attenuated inversion recovery<br />

(FLAIR) MR images requires substantial increases in tissue water. For infarctions<br />

imaged within 6 h after stroke onset (Gonzalez et al., 2006).<br />

In contrast to CT and conventional MR imaging without diffusion<br />

weighting, DW imaging provides detection of lesions in the first hours after the<br />

onset of clinical symptoms. Furthermore, DW imaging is superior in detecting very<br />

small ischemic lesions due to the high signal intensity- to-noise ratio (Wessels et


al, 2005).<br />

- 42 -<br />

Review of literature<br />

On ADC maps, tissues have an appearance opposite to that seen on the DWI<br />

trace. Thus, CSF is hyperintense on ADC maps, whereas on a DWI trace, CSF is<br />

hypointense. On diffusion-weighted images, lesions with restricted water diffusion<br />

are hyperintense while they are hypointense on ADC maps (Davis et al, 2003).<br />

In addition to Standard DW images and ADC maps, a third type of<br />

diffusion image can be created that combines some of the advantages of each. If<br />

one divides the DW image by the "b zero" reference image, or exponential image.<br />

Such an image is similar to standard DW scans, in that areas of acute ischemia<br />

appear hyperintense. Unlike standard DW images, however, so-called exponential<br />

images do not have any T2 weighting and thus do not show the effects of T2 shine-<br />

through (Haaga et al, 2003).<br />

Time course of Diffusion lesion evolution on acute stroke<br />

Information regarding the age of potentially salvageable tissue may be<br />

useful to establish rational time windows for stroke treatment and to identify<br />

patients who are most likely to benefit from acute stroke therapies. Second,<br />

knowledge of the natural evolution of MR characteristics may help estimate the<br />

age of a lesion when the time of symptom onset is unclear or when multiple lesions<br />

are present that could have different times of onset (Lansberg et al, 2001).<br />

In humans, decreased diffusion in ischemic brain tissue is observed as early<br />

as 30 min after vascular occlusion. The ADC continues to decrease with peak<br />

signal reduction at 1–4 days. This decreased diffusion is markedly hyperintense on<br />

DWI (a combination of T2 and diffusion weighting), less hyperintense on<br />

exponential images, and hypointense on ADC images. The ADC returns to<br />

baseline at 1–2 weeks. This is consistent with the persistence of cytotoxic edema<br />

(associated with decreased diffusion) as well as cell membrane disruption, and the<br />

development of vasogenic edema (associated with increased diffusion). At this<br />

point, a stroke is usually mildly hyperintense on the DWI images due to the T2<br />

component and isointense on the ADC and exponential images. Thereafter, the<br />

ADC is elevated secondary to increased extracellular water, tissue cavitation, and<br />

gliosis. There is slight hypointensity, isointensity or hyperintensity on the DWI


- 43 -<br />

Review of literature<br />

images (depending on the strength of the T2 and diffusion components), increased<br />

signal intensity on ADC maps, and decreased signal on exponential images<br />

(Gonzalez et al., 2006).<br />

The time course is influenced by a number of factors including infarct type<br />

and patient age. Minimum ADC is reached more slowly and transition from<br />

decreasing to increasing ADC is later in lacunes versus other stroke types<br />

(nonlacunes). In nonlacunes, the subsequent rate of ADC increase is more rapid in<br />

younger versus older patients (Gonzalez et al., 2006).<br />

Although evolutions of infarction may differ according to the cerebral<br />

territories involved (Huang et al, 2001). Early reperfusion may also alter the time<br />

course .Early reperfusion causes pseudonormalization as early as 1–2 days in<br />

humans who receive intravenous recombinant tissue plasminogen activator (rtPA)<br />

within 3 hours after stroke onset (Gonzalez et al., 2006).


- 44 -<br />

Review of literature<br />

Table 2: (Gonzalez et al., 2006) Diffusion MRI findings in stroke. (ADC Apparent<br />

diffusion coefficient, DWI diffusion-weighted image, EXP exponential image).<br />

Pulse<br />

sequance<br />

Reason for<br />

ADC<br />

changes<br />

DWI<br />

ADC<br />

EXP<br />

Hyperacute<br />

(0-6 h)<br />

Cytotoxic<br />

edema<br />

Hyperintense<br />

Hypointense<br />

Hyperintense<br />

Acute<br />

(6-24 h)<br />

Cytotoxic<br />

edema<br />

Hyperintense<br />

Hypointense<br />

Hyperintense<br />

Early<br />

subacute<br />

(1-7 days)<br />

Cytotoxic<br />

edema with<br />

small amount<br />

of vasogenic<br />

edema<br />

Hyperintense,<br />

gyral<br />

hypointensity<br />

from petechial<br />

hemorrhage<br />

Hypointense<br />

Hyperintense<br />

Late subacute Chronic<br />

Cytotoxic and<br />

vasogenic<br />

edema<br />

Hyperintense<br />

(due to T2<br />

component)<br />

Isointense<br />

Isointense<br />

vasogenic<br />

edema then<br />

gliosis and<br />

neuronal loss<br />

Isointense to<br />

hypointense<br />

Hyperintense<br />

Hypointense<br />

Diffusion-weighted (DW) imaging assesses the mobility of water. Severe<br />

cerebral hypoperfusion causes a restriction in the diffusion capacity of water due to<br />

cytotoxic edema and leads to an increased signal intensity on DW images.<br />

Increased signal intensity is also observed when the blood-brain barrier is<br />

disrupted. These two situations can be differentiated by calculating the apparent<br />

diffusion coefficient (ADC), a quantitative assessment of the diffusion capacity of<br />

water. When predominant cytotoxic edema is present, ADC levels are lower; by<br />

contrast, when the blood-brain barrier is disrupted, vasogenic edema develops,<br />

raising the ADC (Somford et al, 2004).<br />

Reversibility of DWI Stroke Lesions<br />

In the absence of thrombolysis, reversibility (abnormal on initial DWI but<br />

normal on follow-up images) of DWI hyperintense lesions is very rare (Gonzalez


et al., 2006).<br />

- 45 -<br />

Review of literature<br />

Only few convincing cases had been reported in the literature with<br />

resolution of DWI lesions (mostly small lesions, often located in white matter)<br />

(Kummer and Back, 2006)The etiologies are acute stroke or transient ischemic<br />

attack (TIA), transient global amnesia, status epilepticus, hemiplegic migraine, and<br />

venous sinus thrombosis. ADC ratios (ipsilateral abnormal over contralateral<br />

normal-appearing brain) were similar to those in patients with acute stroke. In the<br />

setting of intravenous and/or intra-arterial Thrombolysis, DWI reversibility is more<br />

common. A number of studies have demonstrated that ADCs are significantly<br />

higher in DWI-reversible tissue compared with DWI-abnormal tissue that infarcts<br />

(Gonzalez et al., 2006).<br />

Reliability<br />

In contrast to unenhanced CT or conventional MR imaging, which have low<br />

sensitivities (50%) for acute ischemia detection within the first 6 hours after onset,<br />

diffusion-weighted imaging was reported to have had high sensitivity and<br />

specificity, of 88%–100% and 86%–100%, respectively, in various studies<br />

(Srinivasan et al, 2006).<br />

DWI can show small lesions adjacent to the cerebrospinal fluid. The<br />

diagnosis of a small cortical or brainstem infarct may be difficult on T2–WI<br />

images while DWI easily depicts such lesions. Fluid attenuated inversion recovery<br />

(FLAIR) demonstrates similar sensitivity to ischemic lesions than DWI, but does<br />

not allow demonstration of early lesions and differentiation between new and old<br />

lesions (Davis et al, 2003).<br />

Although after 24 h, CT, FLAIR and T2-weighted images are reliable at<br />

detecting acute infarctions, diffusion imaging continues to improve stroke<br />

diagnosis in the subacute setting. Older patients commonly have FLAIR and T2<br />

hyperintense white matter abnormalities that are indistinguishable from acute<br />

infarctions. However, the acute infarctions are hyperintense on DWI and<br />

hypointense on ADC maps while the chronic foci are usually isointense on DWI<br />

and hyperintense on ADC maps due to elevated diffusion (Gonzalez et al., 2006).<br />

DWI has proved to be more sensitive than CT in the assessment of the


- 46 -<br />

Review of literature<br />

involved extent of the MCA territory. DWI and CT sensitivity ranges between 57%<br />

and 86% and 14% and 43%, respectively. Specificity has been reported excellent<br />

for both imaging techniques and interobserver agreement has shown only<br />

moderately good in both imaging techniques (kappa: 0.6 for DWI and 0.5 for CT)<br />

Furthermore, DWI has shown a more efficient early predictor of the volume of<br />

tissue ultimately infarcted than CT (Davis et al, 2003).<br />

Pitfalls of DWI in stroke:<br />

False negative DWI<br />

In the vertebro-basilar territory, there has been some concern about false-<br />

negative DWI studies,which have been reported in 19% to 31% of patients with<br />

stroke in the posterior circulation. The occurrence of false-negative DWI findings<br />

was significantly higher for small lesions and during the first 24 hours after stroke<br />

onset. In the anterior circulation, false negative initial DWI studies are much rarer<br />

(2%) (Davis et al, 2003).<br />

Most false-negative DWI images occur with punctuate lacunar brainstem or<br />

deep gray nuclei infarctions. Some lesions were presumed on the basis of an<br />

abnormal neurologic exam. While others were are seen on follow-up DWI. False<br />

negative DWI images also occur in patients with regions of decreased perfusion<br />

(increased mean transit time and decreased relative cerebral blood flow) that are<br />

hyperintense on follow-up DWI; that is, they initially had brain regions with<br />

ischemic but viable tissue that eventually infarcted. These findings stress the<br />

importance of obtaining early follow-up imaging in patients with normal DWI<br />

images and persistent stroke-like deficits so that appropriate treatment is initiated<br />

as early as possible (Gonzalez et al., 2006).<br />

False positive DWI<br />

False-positive DWI images occur in patients with a subacute or chronic<br />

infarction with “T2 shine through”. In other words, a lesion appears hyperintense<br />

on the DWI images due to an increase in the T2 signal rather than a decrease in<br />

diffusion. If the DW images are interpreted in combination with ADC maps or<br />

exponential images, this pitfall can be avoided. False-positive DWI images have


- 47 -<br />

Review of literature<br />

also been reported with cerebral abscess (restricted diffusion due to increased<br />

viscosity), and tumor (restricted diffusion due to dense cell packing). Other entities<br />

with decreased diffusion that may be confused with acute infarction are venous<br />

infarctions, demyelinative lesions (decreased diffusion due to myelin<br />

vacuolization), hemorrhage and herpes encephalitis (decreased diffusion due to cell<br />

necrosis) .When these lesions are reviewed in combination with routine T1,<br />

FLAIR, T2, and gadolinium-enhanced T1-weighted images, they are usually<br />

readily differentiated from acute infarctions (Gonzalez et al., 2006).<br />

Artifacts on Diffusion –Weighted Images<br />

Acute stroke is characterized by high signal intensity on DW images;<br />

however, there are other causes of high signal intensity on these images. Knowing<br />

these other causes can be helpful in assessing the likelihood that a given patient has<br />

evidence of cerebral ischemia.<br />

One commonly seen artifact that can mimic the appearance of acute stroke<br />

is known as T2 shine-through .Because DW scans also typically have substantial<br />

spin density and T2 weighting, areas of T2 prolongation may result in carryover of<br />

hyperintense signal to the DW image. Careful comparison with a reference image<br />

(i.e., the "b zero" image) or another type of T2-weighted image can be helpful in<br />

recognizing this artifact.<br />

Another artifact that can mimic the appearance of acute stroke on DW<br />

images is hyperintensity due to white matter diffusion anisotropy. If one reviews<br />

DW images that have been sensitized to diffusion in only one direction (e.g., the<br />

phase-encoding direction), the white matter tracts that run orthogonal to the<br />

direction of sensitization appear relatively bright. This artifact can be recognized<br />

with experience, although comparison with DW images obtained with the diffusion<br />

–sensitizing gradients applied in other directions can be helpful. One can eliminate<br />

this artifact completely by reviewing only images that have isotropic weighting.<br />

Another artifactual cause of hyperintense signal on DW scans is related to<br />

magnetic susceptibility artifacts. Most DW scans are made using echo-planer<br />

imaging techniques. These very rapid MRI methods are commonly used for DW<br />

images because DW scans are highly sensitive to patient motion.


- 48 -<br />

Review of literature<br />

One potential problem with echo-planar images is that they are very<br />

sensitive to susceptibility artifacts at soft tissue/ bone and soft tissue/ air interfaces<br />

.Foreign bodies (e.g., shunt catheters) may also produce susceptibility artifacts are<br />

usually easy to recognize because:<br />

They occur in locations (e.g., calvarium and skull base) that are prone to such<br />

artifacts.<br />

1- They are often flame-shaped.<br />

2- When sever, they can cause regional anatomic distortion in addition to<br />

causing increased signal intensity (Haaga et al, 2003).<br />

Stroke Mimics<br />

These syndromes generally fall into four categories: (1) Non ischemic<br />

lesions with no acute abnormality on routine or diffusion-weighted images; (2)<br />

ischemic lesions with reversible clinical deficits which may have imaging<br />

abnormalities; (3) Vasogenic edema syndromes which may mimic acute infarction<br />

clinically and on conventional imaging; (4) other entities with decreased diffusion<br />

patients (Gonzalez et al., 2006).<br />

Non ischemic Lesions with No Acute Abnormality on Routine or Diffusion-<br />

Weighted Images<br />

Non ischemic syndromes that present with signs and symptoms of acute<br />

stroke but have no acute abnormality identified on DWI or routine MR images<br />

include peripheral vertigo, migraines, seizures, dementia, functional disorders, and<br />

metabolic disorders. The clinical deficits associated with these syndromes are<br />

usually reversible. If initial imaging is normal and a clinical deficit persists, repeat<br />

DWI should be obtained. False-negative DWI and perfusion weighted images<br />

(PWI) occur in patients with small brainstem or deep gray nuclei lacunar<br />

infarctions patients (Gonzalez et al., 2006).<br />

Syndromes with Reversible Clinical Deficits that may have Decreased<br />

Diffusion<br />

Transient Ischemic Attack<br />

A transient ischemic attack (TIA) can be defined as a sudden loss of


- 49 -<br />

Review of literature<br />

neurological function lasting less than 24 hours due to occlusive thromboembolic<br />

cerebrovascular disease (Crisostomo et al, 2003).<br />

DWI demonstrates ischemic abnormalities in nearly half of clinically<br />

defined TIA patients. The lesions are usually less than 15mm in maximal diameter.<br />

The percentage of patients with positive DWI lesion increases with increasing total<br />

symptom duration. The compromised vascular territory detected on diffusion<br />

imaging and the clinical symptoms correlate. In nearly half the patients the<br />

diffusion MRI changes may be fully reversible, while in the remainder the findings<br />

herald the development of a parenchymal infarct (Kesavadas et al., 2003).<br />

Transient Global Amnesia<br />

Transient global amnesia (TGA) is a clinical syndrome characterized by<br />

sudden onset of profound memory impairment resulting in both retrograde and<br />

anterograde amnesia without other neurological deficits. The symptoms typically<br />

resolve in 3–4 h. Many patients with TGA have no acute abnormality on<br />

conventional or diffusion-weighted images .A number of studies, however, have<br />

reported punctuate lesions with decreased diffusion in the medial hippocampus, the<br />

parahippocampal gyrus, and the splenium of the corpus callosum (Gonzalez et al.,<br />

2006).<br />

Vasogenic Edema Syndromes<br />

Patients with these syndromes frequently present with acute neurologic<br />

deficits, which raise the question of acute ischemic stroke. Furthermore,<br />

conventional imaging cannot reliably differentiate cytotoxic from vasogenic edema<br />

because both types of edema produce T2 hyperintensity in gray and/or white<br />

matter. Diffusion MR imaging has become essential in differentiating these<br />

syndromes from acute stroke because it can reliably distinguish vasogenic from<br />

cytotoxic edema. While cytotoxic edema is characterized by decreased diffusion,<br />

vasogenic edema is characterized by elevated diffusion due to a relative increase in<br />

water in the extracellular compartment. Vasogenic edema is characteristically<br />

hypointense to slightly hyperintense on DWI images because these images have<br />

both T2 and diffusion contributions . Vasogenic edema is hyperintense on ADC<br />

maps and hypointense on exponential images while cytotoxic edema is


- 50 -<br />

Review of literature<br />

hypointense in ADC maps and hyperintense on exponential images patients<br />

(Gonzalez et al., 2006).<br />

Venous Infarction<br />

Cerebral venous sinus thrombosis (CVT) is a rare condition that affects<br />

fewer than 1 in 10,000 people. The pathophysiology of CVT is not completely<br />

clear. Venous obstruction results in increased venous pressure, increased<br />

intracranial pressure, decreased perfusion pressure, and decreased cerebral blood<br />

flow. Increased venous pressure may result in vasogenic edema from breakdown of<br />

the blood–brain barrier and extravasation of fluid into the extracellular space.<br />

Blood may also extravasate into the extracellular space. Severely decreased blood<br />

flow may also result in cytotoxic edema associated with infarction. Increases in<br />

CSF production and resorption have also been reported. Parenchymal findings on<br />

imaging correlate with the degree of venous pressure elevation .With mild to<br />

moderate pressure elevations, there is parenchymal swelling with sulcal effacement<br />

but without signal abnormality. As pressure elevations become more severe, there<br />

is increasing edema and development of intraparenchymal hemorrhage. Superior<br />

sagittal sinus thrombosis is characterized by bilateral parasagittal T2-hyperintense<br />

lesions involving cortex and subcortical white matter. Transverse sinus thrombosis<br />

results in T2-hyperintense signal abnormality involving temporal cortex and<br />

subcortical white matter. Deep venous thrombosis is characterized by T2-<br />

hyperintense signal abnormalities in the bilateral thalami and sometimes the basal<br />

ganglia. Hemorrhage is seen in up to 40% of patients with CVT and is usually<br />

located at gray white matter junctions or within the white matter.<br />

DWI has proven helpful in the differentiation of venous from arterial<br />

infarction and in the prediction of tissue outcome. T2-hyperintense lesions may<br />

have decreased diffusion, elevated diffusion or a mixed pattern. Lesions with<br />

elevated diffusion are thought to represent vasogenic edema and usually resolve.<br />

Lesions with decreased diffusion are thought to represent cytotoxic edema. Unlike<br />

arterial stroke, some of these lesions resolve and some persist. Resolution of<br />

lesions with decreased diffusion may be related to better drainage of blood through<br />

collateral pathways in some patients (Gonzalez et al., 2006).


- 51 -<br />

Review of literature


- 52 -<br />

Review of literature


2) Hemorrhagic Stroke<br />

- 53 -<br />

Review of literature<br />

Intraparenchymal Hemorrhage: Appearance and Evolution<br />

The classic pattern for temporal evolution of intracerebral hematomas on<br />

MRI at1.5 T is well known. However, determination of the age of hemorrhage is<br />

often inaccurate because of variations between individual patients. The change of<br />

signal intensity over time depends on many factors, such as the oxygenation state<br />

of hemoglobin, the status of red blood cell membrane, hematocrite, proteins and<br />

clot formation. Among these, the evolution of hemoglobin and red cell membrane<br />

integrity are most important. The transition of oxy-hemoglobin to deoxy-<br />

hemoglobinand thereafter to met-hemoglobin depends primarily upon oxygen<br />

tension in the vicinity of the lesion as well as inside the hematoma itself. In the<br />

hyperacute stage, oxy-hemoglobin will dominate initially, but transformation into<br />

deoxy-hemoglobin will soon take place and deoxy-hemoglobin will dominate after<br />

a few days- the acute hematoma. After approximately one week, deoxy<br />

hemoglobin will transform to met-hemoglobin. However, the rate of this oxidation<br />

to met-hemoglobin will depend on oxygen tension in the tissues, which may<br />

further complicate the temporal pattern of expected signal changes. Initially, met-<br />

hemoglobin will be found within intact red blood cells ( the early subacute stage),<br />

but when the red cell membrane start to rupture, met-hemoglobin will be found in<br />

the extracellular fluid space ( The late subacute stage) , which takes place about<br />

two weeks following hemorrhage. The final stage, the chronic stage, is the result of<br />

continous phagocytation of the breakdown products of hemoglobin, ferritin and<br />

hemosiderin, which starts about one month following hemorrhage. The products<br />

ferritin and hemosiderin, will remain within the phagocytic cells, which<br />

accumulate in the periphery of the hematoma, where they may remain for years,<br />

may be indefinitely, as a marker of an old hemorrhage (Moritani et al, 2005).<br />

Diffusion MR data would differ among the stages of evolving hematomas in<br />

that hematomas containing blood with intact cell membranes would have restricted<br />

diffusion compared with those hematomas in which RBC membranes have lysed.<br />

This hypothesis was based on several facts. First, the presence of intact cell


- 54 -<br />

Review of literature<br />

membranes restricts molecular diffusion .For instance, increased ADC values have<br />

been reported in cases of intratumoral necrosis, a pathologic environment defined<br />

by a lack of intact cell membranes. Second, an important requisite for the diffusion<br />

restriction observed in association with early ischemic infarction is the presence of<br />

intact cells, because the early reduction in diffusion is related to shifts of water<br />

from extracellular space to intracellular space. Once cells have lysed in the<br />

subsequent evolution of subacute to chronic infarction, measured diffusion<br />

increases. Hematomas composed of any and all of the evolutionary stages<br />

theorized to contain hemoglobin within intact RBCs (i.e., hyperacute, acute, and<br />

early subacute hematomas) show significantly reduced ADC values compared with<br />

the single hematoma state theorized to be comprised of lysed RBCs (i.e., ‘‘free’’<br />

methemoglobin in subacute-to-chronic hematomas). The ADC measurements in all<br />

of the intracellular hemoglobin states (intracellular oxyhemoglobin, intracellular<br />

deoxyhemoglobin, and intracellular methemoglobin) were statistically equivalent.<br />

These diffusion data support the theorized biophysical states of hemoglobin and<br />

the RBC put forth in the literature on evolving intracranial hematomas. Further<br />

analysis indicated that the ADC measurements of all early hematomas (including<br />

hyperacute, acute, and even early subacute) were significantly reduced compared<br />

with normal brain tissue. This restriction of diffusion is similar to the phenomenon<br />

observed in cases of early ischemic infarction, in which it is well documented that<br />

within minutes and for the first several days, the ADC value decreases and is<br />

therefore depicted as marked hypointensity on ADC maps. The precise biophysical<br />

explanation for the observed restriction of diffusion in early stages of intracranial<br />

hematomas is uncertain. Potential causes include, but are not limited to: 1)<br />

Shrinkage of extracellular space with clot retraction 2) a change in the osmotic<br />

environment once blood becomes extravascular, which alters the shape of the RBC<br />

-a phenomenon related to the formation of the fibrin network associated with clot<br />

3) a conformational change of the hemoglobin macromolecule within the RBC ;<br />

and the less likely possibility of 4) contraction of intact RBCs (thereby decreasing<br />

intracellular space) Any of these processes might alter the potential mobility of<br />

intracellular water protons. Regardless of the precise explanation for restricted


- 55 -<br />

Review of literature<br />

diffusion in early hematomas, our study indicates that the measured restriction of<br />

diffusion (reduced ADC value) in the hematoma itself would generate similar<br />

hypointensity on ADC maps (Atlas et al, 2000)<br />

Hyperacute Hematoma<br />

Results of DW imaging of hematomas at this stage have not been well<br />

characterized; however a hyperacute intraparenchymal hemorrhage is hyperintense<br />

on DW images, with a decreased apparent diffusion coefficient (ADC). The<br />

possible causes for the decreased ADC are shrinkage of the extracellular space due<br />

to clot retraction, changes in the concentration of hemoglobin and a high viscosity<br />

(Moritani et al, 2005).<br />

Acute hematoma<br />

Diffusion-weighted images of an acute hematoma show a marked<br />

hypointensity, caused by the magnetic field inhomogeneity created by the para-<br />

magnetic deoxy-hemoglobin. Although The ADC has been reported to be<br />

decreased, accurate calculations are often difficult (Moritani et al, 2005).<br />

Early subacute hematoma<br />

On DW imaging, intracellular met-hemoglobin shows hypointensity due to<br />

paramagnetic susceptibility effects and ADC measurements are not reliable due to<br />

susceptibility effects (Moritani et al, 2005).<br />

Late subacute hematoma<br />

It has been reported that late subacute hematomas are hyperintense on DW<br />

imaging. The ADC value for the late subacute hematoma is controversial<br />

(Moritani et al, 2005).<br />

Chronic hematoma<br />

Diffusion-weighted images are hyperintense in chronic hematomas. The<br />

ADC value has been reported to be increased, but this is often difficult to measure<br />

accurately due to paramagnetic susceptibility artifacts (Moritani et al, 2005)<br />

Hemorrhagic and Non hemorrhagic Stroke<br />

(a) Decreased ADC was demonstrated in the lesions of acute hemorrhagic<br />

stroke and in the lesions of acute non hemorrhagic stroke; (b) acute hemorrhagic


- 56 -<br />

Review of literature<br />

stroke was depicted as an area of hypointensity on T2-weighted EP images, in<br />

contrast to the normal to increased signal intensity in lesions of acute non<br />

hemorrhagic stroke; and (c) decreased ADC tended to persist longer in<br />

hemorrhagic stroke lesions than in non hemorrhagic stroke lesions (Ebisu et al,<br />

1997).<br />

Restriction of diffusion, even in the appropriate clinical setting of acute<br />

stroke by clinical assessment, should not necessarily be taken as specific for acute<br />

ischemic infarction. Second, several studies quantify regions of acute infarction on<br />

the basis of restricted diffusion on ADC maps. A potential pitfall to that<br />

quantitative assessment in that early hematomas might mask or mimic regions of<br />

acute infarction. This suggests that ADC maps should always be interpreted with<br />

conventional MR images for comparison (Atlas et al, 2000).<br />

Subarachnoid hemorrhage<br />

Computed tomography (CT) is still essential in the diagnosis of acute<br />

subarachnoid hemorrhages, as the sensitivity and usefulness of MR imaging is<br />

controversial. Fluid –attenuated inversion-recovery (FLAIR) imaging has a high<br />

sensitivity for subarachnoid hemorrhage.However, the sensitivity is low.<br />

It is often difficult to detect subarachnoid hemorrhage on DW images.<br />

However, DW images may be useful to visualize parenchymal injuries secondary<br />

to subarachnoid hemorrhage. Ischemic changes, probably related to subarachnoid<br />

hemorrhage, have showen hyperintensity on DW images (Moritani et al, 2005).<br />

Intraventricular Hemorrhage<br />

Intraventricular hemorrhage is well demonstrated on FLAIR, T1, T2 and<br />

proton density-weighted images. FLAIR has been reported to have highest<br />

sensitivity for detection of intraventricular hematomas. DW images can<br />

demonstrate intraventricular hemorrhages, but in general the GRE images have a<br />

higher sensitivity (Moritani et al, 2005).


- 57 -<br />

Review of literature


- 58 -<br />

Review of literature


- 59 -<br />

Review of literature


- 60 -<br />

Review of literature<br />

Perfusion is the steady-state delivery of blood (nutrients and oxygen) to<br />

tissue parenchyma representing coherent motion of water and cellular material.<br />

Since both perfusion-weighted imaging (PWI) and magnetic resonance<br />

angiography (MRA) map the flow of blood in vessels, there may be some<br />

confusion between the applications of these two techniques. MRA is useful in<br />

demonstrating the macroscopic vasculature (in both arteries and veins) and<br />

detecting morphological changes in blood vessels themselves such as stenosis,<br />

aneurysms, dissections and malformations. PWI detects microscopic flow at the<br />

capillary level and therefore is useful to investigate changes taking place at the<br />

cellular level (Davis et al, 2003).<br />

Perfusion is typically measured in ml/100gm of tissue /min, or units of<br />

CBF. Normal human gray matter is perfused at a rate of 50-60ml/100gm/min and<br />

is maintained in a narrow range by cerebral auto-regulation .Thus, low perfusion<br />

states might result in cellular ischemia, and high perfusion states might be<br />

associated with hypervascular lesions such as some tumors (Sorensen and Reimer,<br />

2000).<br />

Clinical Application of Perfusion Weighted<br />

There are three important parameters that are used to quantify and assess<br />

brain tissue perfusion: cerebral blood flow (CBF), cerebral blood volume (CBV)<br />

and mean transit time (MTT). CBV is defined simply as the amount of blood in a<br />

given amount of tissue at any time (ml/g) as compared to CBF, which represents<br />

the amount of blood moving through a certain amount of tissue per unit time (ml/g<br />

per min). The ratio of CBV and CBF is defined as the mean transit time (units<br />

minutes) (Davis et al, 2003).<br />

Imaging in Stroke<br />

Perfusion imaging techniques can be divided into two broad groups,<br />

depending upon the type of contrast mechanism:<br />

(i) signal monitoring using exogenous (injectable) contrast agents such as


- 61 -<br />

Review of literature<br />

gadolinium DTPA (Gd-DTPA; T1 shortening agent), oxygen 17 in H2O17 (T2<br />

shortening agent) or superparamagnetic agents (T2* shortening).<br />

(ii) Endogenous or inherent contrast agents such as arterial spin labelling<br />

technique (ASL) (Davis et al, 2003).The target of acute stroke therapy is that<br />

portion of the brain tissue that is ischemic and contributes to the neurologic deficit<br />

but is still viable and salvageable if appropriate blood flow is rapidly restored<br />

(Grandian et al, 2002).<br />

Viable tissue that is at risk for infarction is commonly called the ischemic<br />

penumbra or the tissue outside the (non viable) core of infarction that is<br />

nonetheless under perfused. It is postulated that the ischemic penumbra differs<br />

from viable tissue that is not at risk by the (decreased) amount of blood flow that it<br />

receives (Haaga et al, 2003).<br />

Depending on the effectiveness of collateral vessels, such tissue may persist<br />

as long as 17 hours after stroke onset in some patients or, inversely, may be<br />

irreversibly damaged within a few minutes after the arterial occlusion . Therefore,<br />

thrombolytic therapy should be individually tailored (Grandian et al, 2002).<br />

While diffusion-weighted MR imaging is most useful for detecting<br />

irreversibly infarcted tissue (Srinivasan et al, 2006), perfusion imaging findings<br />

provide information on the momentary hemodynamic state of brain tissue, as they<br />

reveal impaired tissue perfusion caused by blood vessel obstruction. Therefore,<br />

perfusion imaging findings may yield information about pathologically<br />

hypoperfused regions, even before genuine structural brain tissue damage has<br />

taken place (Wittsack et al, 2002).<br />

The pathophysiology of ischemia can be considered in four stages, based on<br />

autoregulatory physiology:<br />

First stage, normal hemodynamics.<br />

Second stage, characterized by initial decrease in perfusion pressure. Initially,<br />

autoregulatory vasodilatation preserves CBF, at the same time increasing CBV.<br />

MTT is also typically increased.<br />

Third stage, characterized by greater decrease in perfusion pressure and by an<br />

initial decrease in CBF value. CBV typically remains increased.


- 62 -<br />

Review of literature<br />

Fourth stage, characterized by further decrease in perfusion pressure and decrease<br />

in both CBF and CBV. This stage is associated with tissue ischemia and<br />

hemodynamic failure (Haaga et al, 2003).<br />

Reduced rCBF represents ischemic threat to brain tissue, increased rMTT<br />

represents the brain’s active response to that threat, and reduced rCBV may<br />

represent failure of that response to meet metabolic needs. In this view, regions of<br />

reduced rCBV represent tissue that is very likely destined for infarction, but<br />

regions of abnormal rCBF or rMTT may or may not proceed to infarction (Davis et<br />

al, 2003).<br />

Reliability<br />

In general, perfusion images are less sensitive than DWI images in the<br />

detection of acute stroke. Lesions missed on perfusion images include: (1) lesions<br />

with small abnormalities on DWI that are not detected because of the lower<br />

resolution of MR perfusion images, and (2) lesions with early reperfusion.<br />

Specificities for perfusion images range from 96% to 100%. Occasional false-<br />

positive scans occur when there is an ischemic, but viable hypoperfused region that<br />

recovers (Gonzalez et al., 2006).<br />

It has been proposed that MR perfusion imaging may complement DW<br />

imaging in depicting the region of penumbra. The premise is that DW imaging is<br />

thought to depict the region of core infarction, whereas the region of perfusion<br />

abnormality represents both the core and the penumbra. The difference between<br />

the extent of the diffusion abnormality and the extent of perfusion abnormality is<br />

thus thought to represent penumbra (Haaga et al, 2003).<br />

Addition of perfusion to diffusion information increases the sensitivity for<br />

detecting ischemic at-risk tissue and categorizing its vascular distribution to enable<br />

the best selection among therapeutic options. (Sunshine et al, 2001).<br />

Until now, recanalization treatment with using thrombolytics has been the<br />

only effective treatment for hyperacute ischemic stroke. The procedure is usually<br />

done within three hours after the onset of symptoms for the intravenous method,<br />

and it’s done within six hours for the intraarterial method. This quick intervention<br />

is based upon the hypothesis that there is a significant portion of tissue at risk of


- 63 -<br />

Review of literature<br />

irreversible injury that could be recovered if swift reperfusion is within these time<br />

periods. The area that can be salvaged by timely reperfusion is referred to as the<br />

“ischemic penumbra.” The aim of radiological imaging before recanalization<br />

treatment is to identify those patients who can most benefit from the treatment and<br />

to exclude those patients who are at a large degree of risk with using recanalization<br />

treatment (Lee et al., 2005).<br />

Proximal occlusions are much more likely to result in a diffusion–perfusion<br />

mismatch than distal or lacunar infarctions. Operationally, the diffusion<br />

abnormality is thought to represent the ischemic core and the region characterized<br />

by normal diffusion but abnormal perfusion is thought to represent the ischemic<br />

penumbra. Definition of the penumbra is complicated because of the multiple<br />

hemodynamic parameters that may be calculated from the perfusion MRI data<br />

(Gonzalez et al., 2006).<br />

The volume of penumbral tissue typically decreases with time as the infarct<br />

expands, but the duration of the potential therapeutic window is variable (Prosser<br />

et al, 2005).


- 64 -<br />

Review of literature<br />

Table 3: (Gonzalez et al., 2006) Lesion volumes of diffusion and perfusion. (DWI<br />

Diffusion-weighted imaging, PWI perfusion-weighted imaging).<br />

Pattern Cause Comment<br />

PWI but no DWI<br />

PWI>DWI<br />

PWI=DWI<br />

PWI


- 65 -<br />

Review of literature<br />

A diffusion lesion larger than the perfusion lesion or a diffusion lesion without a<br />

perfusion abnormality usually occurs with early reperfusion. Similarly, in this<br />

situation, there is usually no significant lesion growth (Gonzalez et al., 2006).<br />

An alternate approach for determining the extent of penumbra is to use<br />

perfusion maps alone to distinguish the core of infarction from the penumbra.<br />

Usually with the use of perfusion parameter value thresholds. The most commonly<br />

used perfusion parameter is the mean CBF value (Haaga et al, 2003).<br />

The variability in CBF ratios likely results from a number of different<br />

factors. Most importantly, the data obtained represent only a single time point in a<br />

dynamic process. One major factor is variability in timing of tissue reperfusion.<br />

Another factor is that normal average CBF in human parenchyma varies greatly,<br />

from 21.1 to 65.3 ml .100 g –1 .min –1 ,depending on age and location in gray matter<br />

versus white matter. Other factors affecting thresholds of tissue viability include<br />

variability methodologies, variability in initial and follow-up imaging times, and<br />

variability in post ischemic tissue responses.<br />

Low rCBV ratios are highly predictive of infarction. However, elevated<br />

rCBV is not predictive of tissue viability, and rCBV ratios for penumbral regions<br />

that do and do not infarct may not be significantly different. Some studies have<br />

demonstrated no statistically significant differences in MTT between infarct core<br />

and the two (viable and nonviable) penumbral regions, while others have<br />

demonstrated differences between all three regions or between the viable and<br />

nonviable penumbral regions. (Gonzalez et al., 2006).<br />

Correlation of Diffusion and Perfusion MRI with Clinical Outcome<br />

Attempts have been made to combine DWI and PWI by comparing lesion<br />

volumes identified by the 2 techniques. Diffusion-perfusion mismatches,” in which<br />

the lesion volumes identified by one modality are larger than those by the other,<br />

have been reported by several groups. Larger lesion enlargement of the acute DWI<br />

lesion volume in cases where the acute PWI volume is larger than the DWI lesion.<br />

In cases where the acute DWI lesion was larger than the PWI lesion, total lesion<br />

growth was reduced. Based on these observations, many have hypothesized that<br />

these DWI-PWI mismatches may allow identification of salvageable tissue in


individual patients (Wu et al, 2001).<br />

- 66 -<br />

Review of literature<br />

Now that thrombolytic therapy is commonly employed in acute stroke<br />

management, predicting the extent to which acute infarcts will grow in the days<br />

following presentation is increasingly important. Intravenous or intra-arterial<br />

thrombolysis is capable of saving substantial quantities of brain tissue that<br />

otherwise would have undergone infarction, resulting in marked reduction of<br />

permanent neurologic deficits. However, thrombolysis also carries considerable<br />

risk of catastrophic intracranial hemorrhage. The decision of whether or not to<br />

accept this risk and proceed with thrombolysis is a highly individualized one. In<br />

making that decision, clinicians, patients and their families would benefit from an<br />

informed estimation of how much brain tissue will be affected, and how severe<br />

neurological deficits are likely to be, if thrombolytic therapy is not initiated (Davis<br />

et al, 2003).<br />

Diffusion and perfusion MRI in predicting clinical outcome (Gonzalez et al.,<br />

2006)<br />

1. DWI, CBV, CBF, MTT, and TTP initial lesion volumes all correlate with acute and chronic<br />

neurologic assessment tests. It’s unclear which parameter is best.<br />

2. DWI initial lesion volume correlation with outcome scales is higher for cortical than for<br />

penetrator artery strokes.<br />

3. Patients with a mismatch (proximal stroke) usually have worse clinical outcomes compared<br />

to patients without a mismatch (distal or lacunar stroke).<br />

4. Size of the diffusion–perfusion mismatch volume correlates with clinical outcome.<br />

5. Amount of decrease in size of MTT abnormality volume following intravenous<br />

thrombolysis correlates with clinical outcome.


- 67 -<br />

Review of literature


- 68 -<br />

Review of literature<br />

One of the strengths of MRI for imaging patient tissue is its ability to<br />

acquire information regarding its function as well as its structure. The most<br />

common example is the examination of flowing blood within the vascular network<br />

using MR angiography (MRA). Moving tissue was shown to produce severe image<br />

artifacts. MRA uses flowing tissue such as blood to provide the primary source of<br />

signal intensity in the image. It provides visualization of the normal, laminar flow<br />

of blood within the vascular system and its disruptions due to pathologic<br />

conditions such as stenoses or occlusions. MRA can be of particular benefit in<br />

evaluating vessel patency. MRA techniques have the advantage over conventional<br />

X-ray-based angiographic techniques in that use of a contrast agent is not always<br />

required. Consequently, multiple scans may be performed if desired (e.g.,<br />

visualizing arterial then venous flow) (Brown and Semelka, 2003).<br />

Magnetic resonance angiography (MRA) is a set of vascular imaging<br />

techniques capable of depicting the extracranial and intracranial circulation. In the<br />

setting of acute stroke, these techniques are useful for determining stroke etiology<br />

and assessing vascular flow dynamics. Specifically, they are used to evaluate the<br />

severity of stenosis or occlusion as well as collateral flow. A typical stroke<br />

protocol includes two-dimensional (2D) and/or three-dimensional (3D) time of-<br />

flight (TOF) and contrast-enhanced MRA images through the neck and 3D TOF<br />

MRA images through the Circle of Willis (Gonzalez et al., 2006).<br />

MRA is broadly divided into non contrast and contrast-enhanced<br />

techniques. Non contrast MRA can be acquired with phase contrast (PC) or TOF<br />

techniques, and both can be acquired as 2D slabs or 3D volumes (Gonzalez et al.,<br />

2006).<br />

Clinical Application of MRA in Stroke<br />

Magnetic resonance angiography (MRA) uses the inherent motion<br />

sensitivity of MRI to visualize blood flow within vessels. There are two major<br />

classes of flow imaging methods that rely on the endogenous contrast of moving<br />

spins to produce angiographic images.


- 69 -<br />

Review of literature<br />

The time-of-flight (TOF) technique relies on flow dependent changes in<br />

longitudinal magnetization, whilst the phase-contrast (PC) technique relies on flow<br />

dependent changes in transverse magnetization. In addition there are contrast-<br />

enhanced MRA techniques that employ exogenous contrast agents, such as<br />

gadolinium chelates, to provide vascular contrast (McRobbie et al., 2006).<br />

MR angiography has recently been established as a valid and sensitive tool<br />

to demonstrate pathologic findings in extra- and intracranial arteries in acute stroke<br />

(Seitz et al, 2005).<br />

Extracranial Steno-occlusive diseases<br />

MRA of neck vessels is important in stroke management because<br />

extracranial atherosclerosis causes an estimated 20–30% of strokes (Gonzalez et<br />

al., 2006).<br />

Intracranial Steno-occlusive diseases<br />

In setting of acute stroke, information about the status of the intracranial<br />

arteries is critical to determination of stroke subtype and associated prognosis, and<br />

helps to determine the choice to employ thrombolytic therapy (Davis et al, 2003).<br />

In the setting of acute stroke, intracranial MRA can detect areas of stenosis<br />

and occlusion as well as determine collateral flow. Defining the location of<br />

intracranial vessel pathology is clinically important since an estimated 38% of<br />

patients with acute strokes have arterial occlusion seen on DSA (Gonzalez et al.,<br />

2006).<br />

The depiction of intracranial arterial stenosis by MRA requires a high<br />

sensitivity resolution technique, while the importance of an exact grading by MRA<br />

is not defined yet (Kummer and Back, 2006).<br />

Several studies report that intracranial MRA has a variable reliability of<br />

detecting occlusion and stenoses in the acute stroke setting (Gonzalez et al., 2006).<br />

In the diagnosis of intracranial stenosis, TOF MRA is definitely superior to<br />

PC-MRA protocols. Despite high anatomical resolution TOF-MRA does not allow<br />

an authentic quantification of intracranial vessel stenoses. Stenoses are commonly<br />

overestimated, especially in small vessels. Turbulences and velocity displacement


- 70 -<br />

Review of literature<br />

effects depending on flow velocity and the tortuosity of a vessel cause false<br />

positive stenoses. Typical localizations of pseudostenoses are the carotid siphon<br />

and the bent entrance into the carotid canal. In CE-MRA the contrast medium<br />

limits spin saturation effects and very short echo times reduce the negative signal<br />

effect of turbulent flow patterns, whereby false positive intracranial stenosis can be<br />

identified. Currently, the spatial resolution of CE-MRA as obtained with sufficient<br />

signal-to-noise ratio and without relevant venous contrast overlap in the limited<br />

acquisition time is still significantly lower than that of TOF-MRA. Therefore, CE-<br />

MRA should be used intracranially as an add-on and not as a substitute for TOF-<br />

MRA (Kummer and Back, 2006).<br />

Time of flight MRA also has limitations. Whereas spatial resolution is<br />

adequate for the primary branches of the Circle of Willis, it is less so in the more<br />

distal, smaller branches and the level of diagnostic confidence diminishes (Davis et<br />

al, 2003).<br />

The degree of collateral flow seen on DSA is an independent radiologic<br />

predictor of favorable outcome following thrombolytic treatment. TOF MRA,<br />

however, is limited in evaluation of collateral flow (Gonzalez et al., 2006).<br />

A dedicated analysis of collateral circulations, especially extra-<br />

intracranially, is still the domain of DSA as far as the exact depiction of anatomical<br />

connections is of importance. If the exact anastomotic vascular anatomy is not of<br />

primary interest, the collateral supply is better determined by MR perfusion<br />

techniques Kummer and Back, 2006).<br />

Arterial Dissection<br />

Vascular dissection is an important etiology of acute infarction, causing up<br />

to 20% of infarcts in young patients and an estimated 2.5% of infarcts in the<br />

overall population (Gonzalez et al., 2006).<br />

Dissections occur post-traumatically or spontaneously, in conjunction with<br />

hypertension or with migraine, often with fibromuscular dysplasia or with other<br />

connective tissue diseases (Kummer and Back, 2006).<br />

MRI reveals a crescentic intramural hyperintensity on both T1- and T2-<br />

weighted images (Davis et al, 2003).Acute dissections show luminal narrowing on


- 71 -<br />

Review of literature<br />

MRA and a flap can occasionally appear as a linear low signal defect on MRA<br />

(Gonzalez et al., 2006).<br />

Aneurysm<br />

Both MRA and CTA have been reported to be accurate tests for intracranial<br />

aneurysms, although the sensitivity diminishes greatly for smaller lesions. MRA is<br />

not a complete replacement for X-ray angiography. The accuracy of MRA might<br />

be sufficient to screen patients who are at risk for intracranial aneurysms (Davis et<br />

al, 2003).<br />

Vascular malformation<br />

MRI depicts the nidus of a vascular malformation, whereas MRA depicts<br />

the feeding arteries and draining veins. However, image quality is not sufficient to<br />

obviate the need for selective X-ray angiography prior to intervention. Contrast<br />

enhanced 3D MRA should provide more complete evaluation. When compared<br />

with DSA, the authors reported that CE 3DMRA consistently depicted AVM<br />

components and their orientation (Davis et al, 2003).<br />

Moya Moya<br />

The term moya moya refers to primary moya moya disease and moya moya<br />

pattern, associated with an underlying disease such as atherosclerosis or radiation<br />

therapy. There is an increased incidence of primary moya moya disease in Asians<br />

and in patients with neurofibromatosis or sickle cell disease. Pathologically, there<br />

is a progressive occlusive vasculopathy of the supraclinoid internal carotid artery<br />

with extension into the proximal anterior and middle cerebral arteries associated<br />

with characteristic dilated prominent collateral vessels. Pediatric patients with<br />

moya moya disease tend to develop symptoms from acute infarction while adults<br />

with moya moya disease more frequently present with symptoms from intracranial<br />

hemorrhage into the deep gray nuclei. MRA can depict stenoses and occlusion of<br />

the internal carotid, middle cerebral, and anterior cerebral arteries (Gonzalez et al.,<br />

2006).<br />

Vasculitis<br />

Patients with vasculitis develop ischemic and thrombotic infarctions. There


- 72 -<br />

Review of literature<br />

is also altered wall competence, which can result in dissection or vessel wall<br />

disruption with intracranial hemorrhage. MRA is clinically used to screen for<br />

vasculitis, but is less sensitive than DSA (Gonzalez et al., 2006)<br />

Although for a long time DSA was accepted as a diagnostic gold standard,<br />

MRI and MRA obviously provide the advantage not only to depict stenoses, but<br />

also to provide signs of inflammatory vessel disease. This provides new criteria for<br />

therapy monitoring. An appropriate examination consists of a CE-MRA, as well as<br />

acquisition of spin echo images (Kummer and Back, 2006).<br />

Fibromuscular Dysplasia<br />

Fibromuscular dysplasia (FMD) is a disease of medium-sized vessels,<br />

beginning in young to middle-aged patients with predominance among females<br />

(Kummer and Back, 2006).<br />

Fibromuscular dysplasia (FMD) is an uncommon idiopathic vasculopathy<br />

causing stenoses most often in the renal and internal carotid arteries, and patients<br />

with FMD of the neck vessels can present with infarcts or transient ischemic<br />

attacks (Gonzalez et al., 2006).<br />

FMD manifests through dissections or hyperplasia of the vessel walls with<br />

resulting stenosis. Stenosis and dilations with thinning of vascular walls alternate<br />

over longer distances, occasionally with aneurysms or luminal duplications. On<br />

DSA, this type of lesions was described as “string of beads”. The carotid artery is<br />

often affected bilaterally, mostly sparing the bifurcation. There is an elevated<br />

incidence of intracranial aneurysms and additionally arteriovenous fistulae can<br />

occur.<br />

MRA can provide evidence of dissections with high sensitivity and of<br />

alternating arterial dilatation and stenosed vessels. MRA may also to help identify<br />

the string of beads pattern. In source images of TOF-MRA, segmental changes in<br />

arterial wall thickness, however, are detected to only a limited extent (Kummer<br />

and Back, 2006).


- 73 -<br />

Review of literature


- 74 -<br />

Review of literature


- 75 -<br />

Review of literature<br />

MR spectroscopy enables non-invasive in vivo study of some phases of<br />

brain metabolism. It is based on the same principles as conventional MR, except<br />

that it envisages signal processing during and after sequence acquisition. Whereas<br />

in standard MR the signal intensity is the sum of the signals from all the hydrogen-<br />

containing molecules in a given volume, in spectroscopy the signal from a given<br />

nucleus is separated into its chemical components (Salvolini and Scarabino,<br />

2006).<br />

Clinical Application of MRS in Stroke<br />

The techniques used most often in clinical practice, are proton spectroscopy<br />

(1H MRS) and phosphor spectroscopy (3IP MRS)-those nuclei play the crucial role<br />

in metabolic turn over. The first gives information about various metabolic<br />

disorders (acethyl-aspartate acid- nervous cells marker, creatinin- related to<br />

metabolic changes, choline- element of cell membranes, lactates- markers of<br />

anaerobic metabolism). The second enables evaluation of energetic condition of<br />

the cells (phosphate compounds, mono-and biphosphate, phosphor-creatinin,<br />

adenosine triphosphate ATP) (Kaminska et al, 2007).<br />

However, not all metabolites contain phosphorus, and the MR application of<br />

this nucleus has been restricted to the study of energy and lipid metabolism. In the<br />

brain, [1H]-MRS has two great advantages: the proton provides 15 times more<br />

sensitivity than [31P]-MRS and almost every compound in living tissue contain<br />

hydrogen (Davis et al, 2003).<br />

Single Voxel Spectroscopy<br />

The practical use of in vivo MRS requires a method, which enables<br />

registering the spectrum of selected volume, called the "volume of interest" (VOI)<br />

or voxel.<br />

Multi- Voxel Spectroscopy (Chemical Shift Imaging-CSI)<br />

The CSI technique, which visualizes the chemical shift in one measurement<br />

period, it collects spectroscopic signals from multiple small voxels located within a<br />

large examined area. The registered signals form a map of spatial distribution of


- 76 -<br />

Review of literature<br />

the metabolites. The signals from particular voxels can be shown as spectra and<br />

further analyzed, like in the SVS (Kaminska et al, 2007).<br />

The normal [1H]-MRS spectrum<br />

Histochemical and cell culture studies have shown that specific cell types or<br />

structures have metabolites that give rise to particular [1H]-MRS peaks. A change<br />

in the signal intensity of the peaks may reflect loss or damage of the specific cell<br />

type or a change in the NMR visibility of the metabolite (Davis et al, 2003).<br />

A typical MRS spectrum of adult brain is shown in figure 41 (spectra vary<br />

for newborns, children under 8 years and elderly). The spectrum is read from right<br />

to left with the first and tallest peak assigned to neuronal marker NA (N-<br />

acetylaspartate) resonating at 2 ppm. The second tallest resonance is Cr (creatine<br />

including phosphocreatine) resonating at ~3 ppm. Adjacent to the Cr peak is the<br />

smaller but important Ch (choline) peak. The ratio Ch/Cr of about 0.5 is<br />

characteristic of brain gray matter. Peaks corresponding to glucose and myoinositol<br />

(ml) are also seen in the spectrum of the normal adult brain (Davis et al, 2003).<br />

N-acetyl aspartate (2.01 ppm)<br />

The methyl resonance of NAA produces a large, sharp peak at 2.01 ppm.<br />

NAA is almost exclusively confined to neurons in the human brain, where it is<br />

found predominantly in axons and nerve processes. These cells represent only 2–3<br />

% of the glial population in man. The NAA peak therefore acts as a marker of<br />

healthy neurons. Reduction in the size of the NAA peak provides a useful indicator<br />

of neuronal disease or loss, including infarction after stroke.<br />

In children, the interpretation of the NAA signal is complicated by<br />

increases in the concentration of NAA during development when it is thought to<br />

have a role in supplying acetyl groups for myelin synthesis. In adults, the<br />

concentration is known to vary in different areas in the brain. For example, the<br />

concentration of NAA is higher in grey than white matter, which is explained by<br />

the higher neuronal density in grey matter. This can be overcome in the study of<br />

stroke patients by comparing spectra from an area of suspected abnormality with<br />

spectra from a homologous region of the contralateral normal hemisphere (Davis et<br />

al, 2003).


Creatine (3.94 and 3.03 ppm)<br />

- 77 -<br />

Review of literature<br />

Both creatine and phosphocreatine have signals at 3.94 ppm (methylene<br />

singlet) and 3.03 ppm (methyl singlet) which makes it impossible to distinguish<br />

between the two compounds and therefore total creatine (Cr/PCr) signals are<br />

measured by [1H]-MRS. Other resonances seen at 3.00 ppm arise from the γ-<br />

amino butyric acid (GABA) and cytosolic macromolecules, which become<br />

incorporated into the Cr/PCr peak. Cr/PCr is found in both neurons and glial cells<br />

and acts as a phosphate buffer transport system and energy buffer within the cell.<br />

Little information can be gleaned about phosphocreatine metabolism because the<br />

signal comes from the sum of creatine and phosphocreatine. The complete absence<br />

of creatine signal probably reflects necrotic tissue (Davis et al, 2003).<br />

Choline (3.22 ppm)<br />

The trimethylamine resonance of choline-containing compounds is present<br />

at 3.22 ppm. In normal brain, the Cho peak is thought to consist predominantly of<br />

glycerolphosphocholine and phosphocholine.<br />

Both compounds are involved in membrane synthesis and degradation.<br />

Reduction in the choline peak has been proposed as a marker of membrane damage<br />

(Davis et al, 2003).<br />

Lipid/macromolecule resonances<br />

Lipid peaks are detectable by short echo proton spectroscopy but are not<br />

seen at long echo times because they have short T2 decay constants and so the<br />

signal is lost at longer repetition times.<br />

Lipid/macromolecule peaks have been assigned at 0.9, 1.3 and 1.45 ppm in<br />

normal appearing brain .Increase in these resonances have been reported in stroke<br />

and demyelination. The 0.9 and 1.3 ppm resonances are assigned to the methylene<br />

and methyl groups of lipid, respectively (Davis et al, 2003).


- 78 -<br />

Review of literature<br />

Fig.41 (Davis et al, 2003) Short echo time proton spectra (TE=30 ms,<br />

TR=2020 ms) obtained normal hemisphere.<br />

[H] MRS changes in cerebral ischemia<br />

MRS has been suggested to be more sensitive than MRI in detecting<br />

hypoxic damage. It has been proposed that MRS may be able to detect cerebral<br />

ischemia within seconds of its onset as compared to DWI that provides warning as<br />

early as 1 h after the onset (Davis et al, 2003).<br />

The most striking changes in patients with acute cerebral infarction on MRS<br />

is the appearance of lactate with reduction in NAA and total Cr/PCr within the<br />

infarct compared to the contralateral hemisphere. Large variations in the initial<br />

concentrations of Cho have been observed in the region of infarction (Davis et al,<br />

2003).<br />

The presence of a significant quantity of blood in the brain disrupts the<br />

homogeneity of the magnetic field due to the presence of the iron from the<br />

hemoglobin molecule, and spectroscopy cannot be carried out in patients with<br />

large intracerebral hemorrhages. However, it is possible to sufficiently shim the<br />

magnetic field away from areas of hemorrhagic transformation or subarachnoid<br />

hemorrhage and acquire spectra of acceptable line widths. Petechial hemorrhage<br />

may result in spectra with broader line widths due to field inhomogeneity, but the


spectra may still be of acceptable quality (Davis et al, 2003).<br />

Lactate (doublet at 1.33 ppm)<br />

- 79 -<br />

Review of literature<br />

The detection of lactate on MRS, an end product of glycolysis, is a<br />

particularly useful measure of anaerobic metabolism. Lactate is not detected by<br />

MRS in normal brain. The concentration of lactate rises when the glycolytic rate<br />

exceeds the tissue’s capacity to catabolize it or remove it from the bloodstream.<br />

The rise in brain lactate that results from the mismatch between glycolysis and<br />

oxygen supply has been demonstrated by numerous [1H]-MRS experiments,<br />

making it an important hallmark for the detection of cerebral ischemia. The<br />

persistence of lactate for weeks or months after stroke onset is a common<br />

observation. Removal of lactate depends on the permeability of the blood–brain<br />

barrier (BBB) and diffusion of the metabolite through the damaged tissue. A fall in<br />

lactate concentration has been shown to occur during a period of hyperemia.<br />

Spectroscopic imaging studies have demonstrated that lactate is not confined to<br />

areas of infarction determined by T2-weighted imaging and in one study was even<br />

found in the contralateral hemisphere. In this study, the lactate levels in regions<br />

adjacent to infarcts as shown by T2 changes were not significantly different from<br />

those found in the infarcted brain. It seems likely that this is the result of diffusion<br />

of lactate out of the infarct, but the extent to which there is local production of<br />

lactate in peri-infarct tissue remains uncertain (Davis et al, 2003).<br />

Unfortunately, measurements within the core and penumbra of an infarct are<br />

not definitely different; moreover, the presence of Lac has a differential diagnosis<br />

including abscess. Lac evolution and/or resolution may be observed with or<br />

without an associated infarct (Gonzalez et al., 2006).<br />

NAA (2.01 ppm)<br />

Decreased NAA is most consistent with neuronal degradation, may occur<br />

very early in ischemia, is likely irreversible (as opposed to disease processes such<br />

as demyelination), and may continue for days to weeks even after the cessation of<br />

ischemia (Gonzalez et al., 2006).<br />

The continuing fall in NAA concentration over the course of the first week<br />

after stroke onset cannot be explained simply by an increase in edema because


- 80 -<br />

Review of literature<br />

changes in the concentration of water within the infarct have been corrected for by<br />

using the contralateral water signal as the internal standard. It has been suggested<br />

that NAA is actively degraded by enzymes within the injured neurones in the first<br />

few days or hours following infarction. This remains a possibility but it seems<br />

unlikely that enzymes would remain active for up to 7–10 days within an ischemic<br />

neurone. The gradual decline in NAA concentration and the persistence of lactate<br />

within the region of infarction over a period of a number of days may suggest a<br />

period of ongoing ischemia and has implications in the timing of therapeutic<br />

intervention. Alternatively, it may reflect breakdown of NAA, removal by<br />

phagocytosis or diffusion, or alteration in the physicochemical surrounding of the<br />

metabolite associated with cell dissolution so that it is no longer MRS visible<br />

(Davis et al, 2003).<br />

Correlation of NAA concentration to infarct size (volume) allows a better<br />

prediction of morbidity/ outcome than does either NAA concentration or infarct<br />

volume alone (Gonzalez et al., 2006).<br />

Cr/PCr (3.94 and 3.03 ppm)<br />

Initial reductions in Cr/PCr are identified following infarction and further<br />

reductions have been demonstrated up to 10 days following the time of onset. The<br />

pathological correlate is thought to be gliosis of the tissue. The reduction in NAA<br />

in the infarct region is more marked than the reduction in Cr/PCr and this is<br />

thought to reflect the increased sensitivity of neurons to ischemia, compared to<br />

glial tissue (Davis et al, 2003).<br />

Choline (3.22 ppm)<br />

The choline peak has been shown to be increased, decreased and stays the<br />

same following cerebral infarction. The changes in the choline peak are thought to<br />

reflect changes in the MR visibility of the choline containing compounds that make<br />

up the cell membrane. (Davis et al, 2003) have shown a fall and then a late rise,<br />

maximal 3 months after onset of stroke.This may represent loss of membrane<br />

function, followed by late gliosis (Davis et al, 2003).<br />

Glutamate and other amino acids<br />

It would be useful to be able to measure the concentration of excitotoxic


- 81 -<br />

Review of literature<br />

amino acids, such as glutamate and glutamine, which are released in ischemia and<br />

may be responsible for neuronal injury in the penumbra. Glutamate and glutamine<br />

have strongly coupled spins and the chemical shift of 2.1–2.5 ppm at 1.5T overlaps<br />

with the NAA peak. Other amino acids, such as GABA, aspartate and alanine are<br />

also difficult to detect because of their low concentrations and/or overlap with<br />

more intense resonances from other compounds (Davis et al, 2003).<br />

Lipids/macromolecules<br />

Signals from lipids pose particular problems in [1H]-MRS by obscuring the<br />

resonances from the methyl group of lactate. It has been postulated that membrane<br />

lipids in the brain under normal conditions are not mobile enough to generate sharp<br />

spectral peaks in vivo. During ischemia, degradation of the membrane leads to the<br />

release of free fatty acids that produce well defined, but broad, resonances in the<br />

region of the lactate peak with resulting difficulties in the accurate quantification<br />

of lactate. The ‘metabolite nulling’ technique has been used to separate the lipid<br />

from the lactate signal (Davis et al, 2003).<br />

Advantages of MR spectroscopy:<br />

1. Potentially quantitative if external standards are used in addition to<br />

measurements on the patient (however, this may increase the time of the<br />

examination and further delay diagnosis and treatment of stroke – a<br />

liability).<br />

2. May be easily combined with conventional MR imaging, MR angiography,<br />

PWI and diffusion weighted imaging; these techniques are complementary.<br />

3. May be repeated immediately.<br />

4. No ionizing radiation exposure.<br />

5. Could theoretically improve differentiation in specific situations where the<br />

differential diagnosis includes tumor.<br />

6. Could potentially provide a surrogate marker for guiding stroke<br />

Liabilities<br />

thrombolysis (Gonzalez et al., 2006).<br />

1. The patient must have MR compatibility (that is, no pacemaker, cochlear<br />

implant, neurostimulator, etc.).


2. Motion artifact obviates interpretation.<br />

3. Low spatial resolution.<br />

4. Perfusion data are unavailable.<br />

5. Typically, the entire brain is not imaged.<br />

- 82 -<br />

Review of literature<br />

6. Not all sites will have MR scanners readily available; furthermore, not all<br />

medical facilities will be able to offer this service round the clock (Gonzalez<br />

et al., 2006).


Patients and Methods<br />

- 83 -<br />

Patients & Methods<br />

This study was performed in the period from October 2007 till October<br />

2009 in Zagazig University Hospitals. Patients we referred from Emergency<br />

Unit, General Medicine and Neurology Departments. It included 50 stroke<br />

patients, 32 males and 18 females. Their ages ranged from 19 to 90 years.<br />

All patients were subjected to the following:<br />

1- Full history taking (from the patients or their relatives).<br />

*Accurate onset of neurological manifestation.<br />

*History of hypertension, diabetes mellitus, trauma or bleeding tendency.<br />

*Previous attacks of stroke.<br />

*Past history of allergy against contrast media like Gd-DTPA.<br />

2- Clinical assessment (by neurologist).<br />

3- Laboratory assessment (if needed) as:<br />

*Arterial blood gases to exclude electrolyte imbalance.<br />

*Blood sugar to exclude diabetic coma.<br />

*Prothrombin time.<br />

4- Computed tomography was performed on GE Hi-speed using axial cuts<br />

then the patients are divided into two groups according to the CT findings:<br />

a) Hemorrhagic stroke patients including 11 patients with intracerebral,<br />

intraventricular or subarachnoid hemorrhage.<br />

b) Ischemic stroke patients including 39 patients with or without evidence<br />

of infarction at CT.<br />

5- MR Imaging using Philips Achieva system (1.5 T).<br />

Ischemic stroke group<br />

These patients are chronologically classified into several groups:<br />

hyperacute (1 st 6 hours), Acute (6-24 hours), Early subacute (1-7 days), late<br />

subacute (1-2 weeks) and chronic (>2weeks) stages.


- 84 -<br />

Patients & Methods<br />

Then all these patients (39 patients) were subjected to the following MRI<br />

sequences:<br />

1) Axial T1WI (TR148-597/TE2-15).<br />

2) Axial T2WI (TR4400-4800/TE110).<br />

3) Coronal T2WI (TR4400-4800/TE110).<br />

4) Axial Fluid attenuation inversion recovery (FLAIR) (TR6000/TE120-<br />

TI2000) for all ischemic stroke patients (39 patients).<br />

The previous sequences are done with slice section 5 mm with 1mm gap and<br />

Field of view (FOV) 230mm.<br />

5) Diffusion Weighted image (DWI) for all ischemic stroke patients (39<br />

patients):<br />

*The patients position is the same examining position of the brain MRI in the<br />

supine position with head coil.<br />

*Diffusion-weighted images were obtained using a multisection, single-shot<br />

echo-planar imaging sequence.<br />

*Section thickness was 5 mm with a gap of 1 mm. The number of sections was<br />

set to include the whole brain (average, 22). Matrix size was256 x128 ,(3100-<br />

3400/98-99/1 [TR/TE/excitation]), flip angle 90 degree and FOV 230mm .<br />

*Sensitization gradients are sequentially applied in the X, Y and Z planes.3 sets<br />

of images corresponding to sequential application of the sensitization gradient<br />

(b=1000) in the X, Y and Z planes, and a last set corresponding to the average<br />

images at b=1000. Typically, only 2 sets of images are used in clinical practice:<br />

the b=0 T2W images and the b=1000 average images. Scanning time was 1<br />

minute 15 seconds.<br />

*Apparent diffusion coefficient (ADC) maps were automatically calculated by<br />

MRI machine soft ware and included in the sequence.


- 85 -<br />

Patients & Methods<br />

*Post Processing of DWI: region of interest (ROI) was selected in the area of<br />

infarction and comparative normal brain tissue.<br />

6) Perfusion weighted images (PWI) : for 17 (8 hyperacute, 8 acute and 1<br />

chronic) ischemic stroke patients.<br />

*The patients position is the same examining position of the brain MRI in the<br />

supine position with head coil.<br />

Perfusion-weighted images were obtained following administration of a bolus<br />

of gadopentetate dimeglumine 15 ml, the injection was performed by MRcompatible<br />

power injector at a speed of 4 mL/s followed by 20 ml of normal<br />

saline delivered via a large-bore cannula in the antecubital fossa. Imaging<br />

sequence at 26/19/1 (TR/TE/excitation) and flip angle of 7 degrees was used.<br />

(30) sections were obtained. Section thickness was3.5 mm with a 0 mm gap<br />

(matrix, 256 x 128; FOV 220 mm). Images were obtained at 60 time points per<br />

section with average scanning time 1 minute 24 seconds.<br />

Postprocessing of perfusion data: PWI images are transferred to workstation<br />

then color maps (TTP, MTT, T0) are generated automatically then the<br />

concentration time curve is obtained which represent the changes occurred.<br />

Firstly, ROI puts in the contralateral normal hemisphere as control normal<br />

region, and then multiple ROIs put in the different region of the lesion and<br />

surrounding it to see the extension of the lesion.<br />

Then obtaining of the quantitative measures of each ROI automatically or from<br />

the intensity time curve.<br />

7) Post contrast T1WI :for 11 patients of ischemic stroke patients.


- 86 -<br />

Patients & Methods<br />

8) 2D & 3D time of flight MR angiography (TOF MRA) :TR:23-25, TE:7 ,<br />

flip angle 20 degree, FOV 170-250 mm , slice thickness 1mm with gap 0.5 mm<br />

for 37 patients of ischemic stroke patients.<br />

9) MR venography (MRV): for 1 patient of hemorrhagic infarction.<br />

10) MR spectroscopy (MRS): for 34 patients of ischemic stroke patients.<br />

MRS was performed using the head coil. After global shiming performed with<br />

standered nonselective shiming sequence, the volume of interest (VOI) was<br />

localized in the infarcted area, taking care to avoid the inclusion of normal<br />

tissue or cerebrospinal fluid, and in the corresponding non affected<br />

contralateral region. The water proton signal was suppressed by a preceding<br />

chemical shift-selective radiofrequency pulse. The proton specta were acquired<br />

by means of a double spine-echo sequence. After Fourier tansformation and<br />

zero order phase correction, the areas under the peaks were obtained by<br />

numerical integration. Baseline correction was performed for the purpose of<br />

presentation. Resonance were assigned as follows:<br />

*Lactate (Lac) containing compounds at 1.3 ppm.<br />

*Choline (Cho) containing compounds at 3.2 ppm.<br />

*Creatine-phosphocreatine (Cr) containing compounds at 3.02 ppm.<br />

*N-acetyle aspartate(NAA) containing compounds at 2.02 ppm.<br />

Correlation between DWI and PWI lesions:<br />

*In aid of the DWI (b0 and b1000) for detection of the site of the<br />

infracted area, on PWI ;the ROIs put in the center of the lesions (infracted<br />

core, if present) and surrounding area (penumbra) and measure T0, TTP<br />

and MTT of each ROI then compared with the normal ROI measures.


- 87 -<br />

Patients & Methods<br />

*DWI lesion volumes were determined by manually tracing the edge of<br />

the hyperintense signal on each slice of the trace DWI scans obtained at<br />

b=1000 s/mm 2 . The areas of hyperintensity were summed and multiplied<br />

with the slice thickness plus interslice gap to calculate the volume of the<br />

DWI abnormality. Then, volume of perfusion deficit measured from the<br />

TTP map in comparison with the contralateral side by the same method.<br />

DWI lesion volume is compared by PWI lesion volume. If the lesion<br />

volume in PWI is larger than DWI (DWI/PWI mismatch) subtraction of<br />

the DWI volume from PWI volume to obtain the penumbra volume.<br />

Hemorrhagic stroke group<br />

These patients are classified into several groups according to the site of<br />

hemorrhage : Intracerebral hemorrhage (7 patients), Subarachnoid hemorrhage<br />

(2 patients), Intraventricular hemorrhage (1 patient) and Intracerebral<br />

hemorrhage with intraventricular extension (1 patient).They are<br />

chronologically classified into hyperacute (1st 24 hours), acute stage for 2–3<br />

days, early subacute stage for 3–7 days, late subacute stage up to 2 weeks and<br />

chronic stage more than 2 weeks. Then these patients are subjected to the<br />

following MRI sequences:<br />

1) Axial T1WI .<br />

2) Axial T2WI .<br />

3) Axial Fluid attenuation inversion recovery (FLAIR).<br />

4) Axial Gradient images (T2*) (TR250/TE12).<br />

5) Diffusion Weighted image (DWI).<br />

6) 2D & 3D time of flight MR angiography (TOF MRA) for all hemorrhagic<br />

stroke patients and Phase contrast (Pc) MRA in 1 case of subarachnoid<br />

hemorrhage.


RESULTS<br />

RESULTS<br />

This study was performed on 50 patients .They were 32 males<br />

(64%) and 18 females (36%).Their ages ranged from 19to 90 years.<br />

Table (4):Age & Sex distribution among our patients.<br />

Age group<br />

(years)<br />

Age<br />

Patient<br />

NO.<br />

10- 1 2<br />

20- 1 2<br />

30- 1 2<br />

40- 8 16<br />

50- 11 22<br />

60- 19 38<br />

70- 7 14<br />

80- 2 4<br />

sex<br />

% sex Patient<br />

NO.<br />

Male<br />

Female<br />

Total 50 100 50 100<br />

The most common age group was 60-70 years (19 patients-38%) with<br />

mean age 59.6±14.1 years.<br />

-88-<br />

32<br />

18<br />

%<br />

64<br />

32


Table (5) Clinical presentation of our patients<br />

Clinical presentation No of<br />

patients<br />

RT sided hemiparesis 9 18<br />

RT sided hemiparesis with aphasia 4 8<br />

RT sided hemiplegia 10 20<br />

RT sided hemiplegia with aphasia 5 10<br />

LT sided hemiparesis 6 12<br />

LT sided hemiplegia 10 20<br />

Quadriparesis 4 8<br />

Headache with neck rigidity 2 4<br />

-89-<br />

RESULTS<br />

%


RESULTS<br />

Most of our cases were exposed to different risk factors<br />

including Hypertension, Ischemic heart disease, Diabetes mellitus,<br />

Hyperlipidaemia and smoking . Multiple risk factors can coexist in<br />

same patient.<br />

Table (6): Risk factors among our patients<br />

Risk factors No. Of patient %<br />

Hypertension 15<br />

Ischemic heart disease<br />

Ischemic heart disease with<br />

atrial fibrillation<br />

Diabetes mellitus<br />

Hyperlipidaemia<br />

Smoking 6<br />

7<br />

30<br />

14<br />

5 10<br />

Hypertension was the most common risk factor seen in 15 patients<br />

(30% of patients).<br />

9<br />

7<br />

-90-<br />

18<br />

14<br />

12


RESULTS<br />

Our patients were classified into 2 groups; Ischemic and Hemorrhagic<br />

stroke.<br />

Table (7): Classification of our patient:<br />

Type of stroke No. of patients %<br />

Ischemic stroke<br />

Hemorrhagic stroke<br />

Total 50 100<br />

39<br />

11<br />

Ischemic stroke patients were 39 patients (78%) however hemorrhagic<br />

stroke were 11 patients (22%).<br />

-91-<br />

78<br />

22


Ischemic stroke patients were classified into several groups<br />

according to the age of stroke.<br />

RESULTS<br />

Table (8):Chronological classification of ischemic stroke patients:<br />

Stage of infarction No. of patients %<br />

Hyperacute (1 st 6 hours). 10 25.6<br />

Acute (6-24 hours). 12 30.8<br />

Early subacute (1-7 days). 4 10.3<br />

Late subacute(1-2 weeks). 5 12.8<br />

Chronic (> 2 weeks). 8 20.5<br />

Total 39 100<br />

-92-


RESULTS<br />

Our ischemic stroke patients are subjected to different MRI<br />

sequences.<br />

Type of infarction<br />

Table (9): Different MRI sequences in ischemic stroke patients.<br />

No. of<br />

patient<br />

T1 T2 FLAIR MRA DWI PWI PCT1 MRV MRS<br />

Hyperacute 10 10 10 10 10 10 8 5 - 10<br />

Acute 12 12 12 12 12 12 8 4 - 10<br />

Early subacute 4 4 4 4 4 4 - - - 4<br />

Late subacute 5 5 5 5 3 5 - - 1 2<br />

Chronic 8 8 8 8 8 8 1 1 - 8<br />

Total<br />

%<br />

39 39 39 39 37 39 17 10 1 34<br />

100 100 100 100 95 100 46 26 2.6 87<br />

Perfusion weighted imaging (PWI) was done in only 17<br />

patients to show the presence or absence of the ischemic but<br />

still viable tissue (ischemic penumbra).<br />

Postcontrast T1(PCT1) was done in 10 patients only and<br />

showed arterial enhancement in 5 patients out of them.<br />

-93-


Stage of<br />

stroke Intensity Pt<br />

Hyperacute<br />

(10)<br />

RESULTS<br />

Table (10): Appearance of different ischemic stroke stages at<br />

different MRI sequences.<br />

*Not appear<br />

*Subtle intense<br />

Acute (12) *Not appear<br />

Early<br />

subacute(4)<br />

Late<br />

subacute(5)<br />

*Subtle intense<br />

*hyperintense<br />

T2WI FLAIR DWI(b0) DWI(b1000) Exponential image ADC<br />

No<br />

3<br />

9<br />

1<br />

2<br />

7<br />

Intensity Pt<br />

*Not appear<br />

*Subtle intense<br />

No<br />

6<br />

4<br />

Intensity Pt<br />

*Not appear<br />

*Subtle<br />

intense<br />

*Hyperintense 11 *Not appear<br />

*Subtle<br />

intense<br />

*hyperintense<br />

No<br />

1<br />

7<br />

3<br />

9<br />

2<br />

Intensity Pt<br />

*hyperintense<br />

ẽ brightness<br />

*hyperintense<br />

ẽ brightness<br />

No<br />

Intensity Pt<br />

No<br />

Intensity Pt<br />

No<br />

10 *hyperintense 10 hypointense 10<br />

12 *hyperintense 12 hypointense 12<br />

*hyperintense 4 *Hyperintense 4 *Hyperintense 4 *hyperintense 4 *hyperintense 4 hypointense 4<br />

*hyperintense 5 *Hyperintense 5 *Hyperintense 5 *hyperintense 5 *isointense 5 isointense 5<br />

Chronic(8) *hyperintense 8 *Hyperintense 8 *Hyperintense 8 *isointense<br />

-94-<br />

*hypointense<br />

6<br />

2<br />

*hypointense 8 hyperintense 8


stroke<br />

Table (11): Different ADC values in hyperacute ischemic<br />

Patient No ADC in infarction<br />

(10 -6 mm 2 /sec)<br />

ADC of contralateral<br />

normal area<br />

(10 -6 mm 2 /sec)<br />

1 354.62 814.95<br />

2 473.25 838.38<br />

3 480.54 959.51<br />

4 494 747.47<br />

5 370.72 731.85<br />

6 428.25 834.19<br />

7 405.49 722.03<br />

8 415.11 754.09<br />

9 320.82 790.60<br />

10 501.67 809.14<br />

RESULTS<br />

The mean ACD at the infarction site in hyperacute stage<br />

was 423.5410±63.7865 (mean±SD) while the mean ADC of the<br />

contralateral normal area was 800.2200±69.9746. There is a<br />

highly statistical significant difference between the ADC in<br />

infarction and contralateral normal site using T-test; t=15.383,<br />

p< 0.001.<br />

-95-


Table (12): Different ADC values in acute ischemic stroke<br />

Patient No ADC in infarction<br />

(10 -6 mm 2 /sec)<br />

ADC of contralateral<br />

normal area<br />

(10 -6 mm 2 /sec)<br />

1 314.46 839.77<br />

2 363.02 823.91<br />

3 351.07 1155.22<br />

4 377.90 926.17<br />

5 367.72 1023.41<br />

6 236.71 668.48<br />

7 356.07 808.01<br />

8 319.14 739.22<br />

9 401.76 803.72<br />

10 235.25 762.23<br />

11 229.56 878.66<br />

12 342.11 911.87<br />

RESULTS<br />

The mean ACD at the infarction site in acute stage was 324.5642±59.5444<br />

while the mean ADC of the contralateral normal area was 869.9808±132.9708.<br />

There is a highly statistical significant difference between the ADC in infarction<br />

and contralateral normal site using T-test; t=15.555, p< 0.001.<br />

-96-


RESULTS<br />

Table (13): Different ADC values in early and late subacute ischemic<br />

stroke<br />

stage Patient<br />

No<br />

ADC in infarction<br />

(10 -6 mm 2 /sec)<br />

ADC of contralateral<br />

normal area<br />

(10 -6 mm 2 /sec)<br />

Early subacute 1 373.63 661.61<br />

Early subacute 2 434.03 689.42<br />

Early subacute 3 667.94 822.73<br />

Early subacute 4 617.03 788.46<br />

Late subacute 1 872.54 816.92<br />

Late subacute 2 743.32 751.44<br />

Late subacute 3 831.25 823.69<br />

Late subacute 4 894.67 911.95<br />

Late subacute 5 851.29 834.55<br />

The mean ACD at the infarction site in early subacute stage was<br />

525.6575±144.9676 while the mean ADC of the contralateral normal area was<br />

740.5550±77.2328. There is a highly statistical significant difference between the<br />

ADC in infarction and contralateral normal site using T-test; t=6.340, p= 0.008.<br />

The mean ACD at the infarction site in late subacute stage was<br />

838.6140±58.2862 while the mean ADC of the contralateral normal area was<br />

827.7100±57.2120. There is no statistical significant difference between the ADC<br />

in infarction and contralateral normal site using T-test; t=0.862, p= 0.437.<br />

-97-


Table (14): Different ADC values in chronic ischemic stroke<br />

Patient No ADC in infarction<br />

(10 -6 mm 2 /sec)<br />

ADC of contralateral<br />

normal area<br />

(10 -6 mm 2 /sec)<br />

1 1647.25 757.88<br />

2 1312.49 838.20<br />

3 3023.26 785.62<br />

4 1597.53 719.24<br />

5 1675.18 856.23<br />

6 1241.05 794.64<br />

7 1164.29 958.45<br />

8 2845.90 908.23<br />

The mean ACD at the infarction site in chronic stage was<br />

RESULTS<br />

1813.3688±719.4211 while the mean ADC of the contralateral normal<br />

area was 827.3112±79.2877. There is a highly statistical significant<br />

difference between the ADC in infarction and contralateral normal site<br />

using T-test; t=3.845, p= 0.006.<br />

-98-


RESULTS<br />

Table(15): Correlation between Mean ADC at the infarction<br />

and contralateral normal site in different ischemic stroke stages using<br />

Paired T test.<br />

Stage Patient<br />

No<br />

ADC in<br />

infarction<br />

[x - ±SD]<br />

(10 -6 mm 2 /sec)<br />

Hyperacute 10 423.5410±<br />

63.7865<br />

Acute 12 324.5642±<br />

Early<br />

subacute<br />

Late<br />

subacute<br />

4<br />

5<br />

59.5444<br />

525.6575±<br />

144.9676<br />

838.6140±<br />

58.2862<br />

Chronic 8 1813.3688±<br />

x - : is the mean<br />

719.4211<br />

SD: is the standered deviation.<br />

-99-<br />

ADC in<br />

contralateral<br />

normal site<br />

[x - ±SD]<br />

(10 -6 mm 2 /sec)<br />

800.2200±<br />

69.9746<br />

869.9808±<br />

132.9708<br />

740.5550±<br />

77.2328<br />

827.7100±<br />

57.2120<br />

827.3112±<br />

79.2877<br />

Paired T-<br />

test<br />

P value<br />

15.383


RESULTS<br />

Table(16):ADC and rADC correlation with different stages of infaction<br />

using ANOVA (f):<br />

ADC in infarction<br />

[x - ±SD]<br />

(10 -6 mm 2 /sec)<br />

rADC in infarction<br />

[x - ±SD]<br />

Hyperacute 423.5410± 63.7865 0.5308± 0.0899<br />

Acute 324.5642± 59.5444 0.3765± 0.06954<br />

Early subacute 525.6575± 144.9676 0.7002± 0.1231<br />

Late subacute 838.6140± 58.2862 1.0135± 0.03422<br />

normal site".<br />

Chronic 1813.3688± 719.4211 2.2094± 0.8806<br />

ANOVA (f) 28.193 28.280<br />

P value


RESULTS<br />

Fig 42(a)Scatterplot between the groups (stages) of ischemic stroke and<br />

ADC, (b)Scatterplot between the stages of ischemic stroke and rADC(ADC ratio)<br />

where 1 is hyperacute , 2 is acute stage ,3 is early subacute, 4 is late subacute and<br />

5 is chronic stages.The rADC starts to decrease (


RESULTS<br />

Most of hyperacute and acute ischemic stroke patients (17 patients)<br />

are subjected to PWI.<br />

17 patients.<br />

Table (17): Relation between lesion volumes in DWI & PWI in<br />

Pattern No of patient<br />

PWI > DWI (diffusion<br />

perfusion mismatch with<br />

penumbrae)<br />

PWI = DWI (matched)<br />

Hyperacute (5 patients)<br />

Acute (3 patients)<br />

Hyperacute (2 patients)<br />

Acute (3 patients)<br />

Chronic (1 patient)<br />

8<br />

6<br />

Arterial<br />

distribution<br />

ICA(1 patient)<br />

MCA(6patients)<br />

PCA(1 patient)<br />

ICA(1 patient)<br />

MCA(3patients)<br />

PCA(1 patient)<br />

ACA(1 patient)<br />

PWI < DWI Acute 2 MCA(2patients)<br />

DWI but no PWI Hyperacute 1 Pontine branch 0f<br />

basilar artery (1<br />

Total 17<br />

-102-<br />

patient)


RESULTS<br />

Table (18): Mean transit time (MTT) values in patients with<br />

diffusion perfusion mismatch.<br />

Patient<br />

No.<br />

Stage MTT at<br />

penumbra(sec)<br />

MTT at normal<br />

contralateral<br />

area(sec)<br />

1 Hyperacute 19 11.5<br />

2 Hyperacute 22 16<br />

3 Hyperacute 13 6<br />

4 Hyperacute 16.5 10.2<br />

5 Hyperacute 17.7 9.4<br />

6 Acute 18 11<br />

7 Acute 19.6 11.4<br />

8 Acute 15.2 9.3<br />

The mean MTT at penumbra in diffusion perfusion<br />

mismatched cases was 17.6250±2.7675 while the mean MTT of<br />

the contralateral normal area was 10.6000±2.8087. There is a<br />

highly statistical significant difference between the MTT at<br />

penumbra and contralateral normal site using T-test; t=21.329,<br />

p< 0.001.<br />

-103-


Table (19): Time to peak (TTP) values in patients with<br />

diffusion perfusion mismatch.<br />

Patient<br />

No.<br />

Stage TTP at<br />

penumbra(sec)<br />

RESULTS<br />

TTP at normal<br />

contralateral<br />

area(sec)<br />

1 Hy.peracute 49 44<br />

2 Hyperacute 72 60<br />

3 Hyperacute 32.5 26<br />

4 Hyperacute 38.2 28.3<br />

5 Hyperacute 45.9 36.4<br />

6 Acute 42.5 30<br />

7 Acute 41.4 32.6<br />

8 Acute 36.8 30.5<br />

The mean TTP at penumbra in diffusion perfusion<br />

mismatched cases was 44.7875±12.1561 while the mean TTP of<br />

the contralateral normal area was 35.9375±11.2246. There is a<br />

highly statistical significant difference between the mean TTP<br />

at penumbra and contralateral normal site using T-test; t=9.154,<br />

p< 0.001.<br />

-104-


RESULTS<br />

Table(20): Correlation between Mean MTT and TTP at the<br />

infarction and contralateral normal site using Paired T test.<br />

Stage Patient<br />

No<br />

Penumbra<br />

[x - ±SD]<br />

MTT (sec) 8 17.6250±<br />

2.7675<br />

TTP (sec) 8 44.7875±<br />

12.1561<br />

-105-<br />

contralateral<br />

normal site<br />

[x - ±SD]<br />

10.6000±<br />

2.8087<br />

35.9375±<br />

11.2246<br />

Paired T-<br />

test<br />

P value<br />

21.329


RESULTS<br />

Table (21): MRA findings in different types of ischemic stroke<br />

patients (37 patients).<br />

Type of stroke<br />

Hyperacute<br />

Positive<br />

(occlusion)<br />

8<br />

MRA<br />

Negative Total<br />

2<br />

10<br />

Distribution(+ve)<br />

Both ICAs (1 patient)<br />

Rt ICA (1 patient)<br />

Rt MCA(1 patient)<br />

Lt MCA(3 patients)<br />

Rt PCA(1 patients)<br />

Both PCA(1 patient)<br />

Acute 4 8 12 Rt MCA(1 patient)<br />

Lt MCA(3 patients)<br />

Early subacute 2 2 4 Rt MCA(2 patients)<br />

Late subacute 1 2 3 Lt MCA<br />

Chronic - 8 8 -<br />

Total 15 22 37 -<br />

MRA was positive (occlusion) in 15 out of 37 patients mainly<br />

in the hyperacute (8 out of 10 patients) and acute (4 out of 8 patients)<br />

stages.<br />

-106-


RESULTS<br />

Table (22): Post contrast T1WI findings in different types of<br />

ischemic stroke patients (11 patients).<br />

Type of stroke<br />

Hyperacute<br />

Acute<br />

Chronic<br />

Positive<br />

+ ++<br />

1<br />

2<br />

-<br />

2<br />

-<br />

-<br />

PCT1<br />

Negative Total<br />

Total 3 2 5 10<br />

Postcontrast T1(PCT1) was done in 10 patients only and<br />

showed arterial enhancement in 5 patients out of them (2 patients<br />

showed marked enhancement and 3 patients showed mild<br />

enhancement)<br />

-107-<br />

2<br />

2<br />

1<br />

5<br />

4<br />

1


RESULTS<br />

Table (23): MR spectroscopic findings in ischemic stroke patients(34<br />

patients).<br />

Stage No Lactate iNAA/cNAA iCr/cCr iCho/cCho<br />

Hyperacute 1 + 0.09 0.46 0.92<br />

Hyperacute 2 + 0.21 0.08 0.76<br />

Hyperacute 3 + 0.02 0.62 0.45<br />

Hyperacute 4 + 0.81 1.05 0.88<br />

Hyperacute 5 + 0.43 0.31 0.94<br />

Hyperacute 6 + 0.19 0.37 0.77<br />

Hyperacute 7 + 0.01 0.07 0.21<br />

Hyperacute 8 + 0.68 1.03 0.73<br />

Hyperacute 9 + 0.32 0.23 0.85<br />

Hyperacute 10 + 0.56 0.92 0.91<br />

Acute 11 + 0.72 0.77 0.42<br />

Acute 12 + 0.61 1.00 1.13<br />

Acute 13 + 0.01 0.48 0.44<br />

Acute 14 + 0.08 0.63 0.07<br />

Acute 15 + 0.37 0.09 0.36<br />

Acute 16 + 0.45 0.95 1.06<br />

Acute 17 + 0.21 0.08 0.74<br />

-108-


Acute 18 + 0.17 1.12 0.96<br />

Acute 19 + 0.05 0.01 0.81<br />

Acute 20 + 0.57 0.69 1.21<br />

Early subacute 21 + 0.46 0.65 0.86<br />

Early subacute 22 + 0.7 0.32 0.62<br />

Early subacute 23 - 0.51 0.43 0.46<br />

Early subacute 24 + 0.32 0.08 1.21<br />

Late subacute 25 - 0.82 0.02 0.91<br />

Late subacute 26 + 0.91 0.89 0.84<br />

Chronic 27 + 0.24 0.81 0.27<br />

Chronic 28 + 0.52 0.64 0.48<br />

Chronic 29 - 0.34 0.43 1.72<br />

Chronic 30 + 0.83 1.13 0.92<br />

Chronic 31 - 0.64 0.77 1.09<br />

Chronic 32 - 0.76 0.61 1.56<br />

Chronic 33 - 0.21 1.27 1.66<br />

Chronic 34 + 0.92 0.93 0.88<br />

RESULTS<br />

iNAA indicates N-acetylaspartate from infarcted area; cNAA:N-acetylaspartate<br />

from contralateral area, iCr: creatine-phosphocreatine from infarcted area, cCr:<br />

creatine-phosphocreatine from contralateral area, iCho: choline-containing<br />

compounds from infarcted area, cCho: choline-containing compounds from<br />

contralateral area, Lac: lactate; +: present; -: absent.<br />

-109-


RESULTS<br />

Table(24):MR spectroscopic lactate findings correlation with different<br />

stages of infaction using Chi-Sguare test (X 2 ):<br />

Stage Hyperacute Acute Early<br />

Patient<br />

No.<br />

Lactate<br />

+ve<br />

Lactate<br />

-ve<br />

subacute<br />

Late<br />

subacute<br />

Chronic<br />

10 10 4 2 8<br />

10<br />

100%<br />

10<br />

100%<br />

3<br />

75%<br />

0 0 1<br />

25%<br />

X 2 11.637<br />

P value 0.020<br />

1<br />

50%<br />

1<br />

50%<br />

4<br />

50%<br />

4<br />

50%<br />

There is a statistical significant difference in lactate level<br />

among different stroke stages (p


RESULTS<br />

Table(25):MR spectroscopic findings correlation with different stages of<br />

infaction using ANOVA (f):<br />

Patient<br />

No.<br />

Mean iNAA/cNAA<br />

[x - ±SD]<br />

Mean iCr/cCr<br />

[x - ±SD]<br />

Hyperacute 10 0.3320± 0.2801 0.5140±<br />

0.3743<br />

Acute 10 0.3270± 0.2593 0.5830±<br />

Early<br />

subacute<br />

Late<br />

subacute<br />

0.4071<br />

4 0.3400± 0.1971 0.3700±<br />

0.2371<br />

2 0.8650± 0.0636 0.4550±<br />

0.6152<br />

Chronic 8 0.5575± 0.2735 0. 8238±<br />

0.2782<br />

Mean iCho/cCho<br />

[x - ±SD]<br />

0.7420± 0.2356<br />

0.7200± 0.3816<br />

0. 7875± 0.3261<br />

0.8750± 0.0495<br />

1.0725± 0.5422<br />

ANOVA (f) 2.730 1.368 1.176<br />

P value 0.048 0.269 0.342<br />

There is a statistical significant difference in NAA level among<br />

different stroke stages(p


Table (26): LSD (least significant difference) for Mean iNAA/cNAA<br />

comparison between different ischemic stroke stages.<br />

Hyperacute Acute Early<br />

subacute<br />

RESULTS<br />

Late<br />

subacute<br />

Acute >0.05 - - -<br />

Early subacute >0.05 >0.05 - -<br />

Late subacute 0.05 >0.05<br />

LSD revealed that late subacute stage is statistically significant higher<br />

than hyperacute, acute and early subacute stages.<br />

-112-


patients).<br />

RESULTS<br />

Table (27):Classification of hemorrhagic stroke patients (11<br />

Type of hemorrhage No %<br />

ICH<br />

SAH<br />

IVH<br />

Total 12<br />

8<br />

2<br />

2<br />

72.7<br />

The number of hemorrhagic stroke patients was 11 patients<br />

however the number of lesions was 12 as there is one patient had ICH<br />

with intraventricular extension.<br />

-113-<br />

18<br />

18


Stage of<br />

hematoma<br />

Hyperacute<br />

(1 st 24<br />

hours)<br />

Acute (1 st<br />

2-3 days)<br />

Early<br />

subacute<br />

(3-7days)<br />

Late<br />

subacute<br />

(1-2<br />

weeks)<br />

Chronic<br />

(>2weeks)<br />

Table (28): Signal intensity of cases of intracerebral hematoma in<br />

different MRI sequences.<br />

No Degree<br />

of<br />

edema<br />

2 + iso ↑<br />

2 + Iso<br />

T1 T2 GRE (T2 * )<br />

with↓rim<br />

with↓rim<br />

heterogenous<br />

↑<br />

FLAIR<br />

↑<br />

with↓rim<br />

RESULTS<br />

DWI<br />

b 0 b<br />

↑ with↓<br />

rim<br />

1000<br />

ADC<br />

map<br />

↑ ↓<br />

↓ ↓ ↓ ↓ ↓ ↓<br />

1 + ↑ ↓ ↓ ↓ ↓ ↓ ↓<br />

2 - ↑ ↑ ↑ ↑ ↑ ↑ ↑<br />

1 - ↓with↑<br />

center<br />

↓with↑<br />

center<br />

Iso=intermediate signal intensity.<br />

↑=high signal intensity.<br />

↓=low signal intensity.<br />

-114-<br />

↓with↑<br />

center<br />

↓with↑<br />

center<br />

↓with↑<br />

center<br />

↓ ↑


RESULTS<br />

Table (29): Detection of subarachnoid hemorrhage by different MRI<br />

sequences.<br />

Patient<br />

No.<br />

1 st<br />

patient<br />

2 nd<br />

patient<br />

onset T1 T2 GRE<br />

(T2 * )<br />

FLAIR<br />

DWI<br />

b 0 b<br />

1000<br />

ADC<br />

map<br />

5 days ++ + + ++ + + +<br />

6 days - - - + - - -<br />

-115-


RESULTS<br />

Table (30):Signal intensity of cases of intraventricular in different<br />

MRI sequences.<br />

Patient<br />

No.<br />

1 st<br />

patient<br />

2 nd<br />

patient<br />

onset T1 T2 GRE<br />

(T2 * )<br />

FLAIR<br />

b<br />

0<br />

DWI<br />

b<br />

1000<br />

ADC<br />

hyperacute iso ↑ ↑ ↑ ↑ ↑ ↓<br />

map<br />

Acute iso ↓ ↓ ↓ ↓ ↓ ↓<br />

Iso=intermediate signal intensity.<br />

↑=high signal intensity.<br />

↓=low signal intensity.<br />

-116-


-117-<br />

RESULTS<br />

Table (31): MRA findings in different types of hemorrhagic stroke.<br />

Site of hemorrhage<br />

Non<br />

filling<br />

Positive<br />

MRA<br />

spasm aneurysm Total<br />

Negative Total<br />

Intracerebral(8) 1 2 - 3 5 8<br />

Subarachnoid(2) - 1 1 2 - 2<br />

Intraventricular(1) - - - - 1 1<br />

Total 1 3 1 5 6 11


Stage of<br />

stroke<br />

Hyper-<br />

acute(10)<br />

Acute (12)<br />

Early subacute<br />

(4)<br />

Late subacute<br />

(5)<br />

Chronic<br />

(8)<br />

Table (10): Appearance of different ischemic stroke stages at different MRI sequences.<br />

T2WI<br />

Intensity<br />

Not appear<br />

Subtle intense<br />

Not appear<br />

Subtle intense<br />

hyperintense<br />

hyperintense<br />

hyperintense<br />

hyperintense<br />

Pt.<br />

No.<br />

9<br />

1<br />

2<br />

7<br />

3<br />

4<br />

5<br />

8<br />

Intensity<br />

Not appear<br />

FLAIR<br />

Subtle intense<br />

Hyperintense<br />

Hyperintense<br />

Hyperintense<br />

Hyperintense<br />

Pt.<br />

No.<br />

4<br />

6<br />

11<br />

4<br />

5<br />

8<br />

Intensity<br />

Not appear<br />

DWI(b0)<br />

Subtle intense<br />

Not appear<br />

Subtle intense<br />

Hyperintense<br />

Hyperintense<br />

Hyperintense<br />

Hyperintense<br />

-94-<br />

Pt.<br />

No<br />

9<br />

1<br />

2<br />

7<br />

3<br />

4<br />

5<br />

8<br />

DWI(b1000)<br />

Intensity<br />

hyperintense ẽ<br />

brightness<br />

hyperintense ẽ<br />

brightness<br />

hyperintense<br />

hyperintense<br />

isointense<br />

hypointense<br />

Pt.<br />

No.<br />

10<br />

12<br />

4<br />

5<br />

2<br />

6<br />

Exponential<br />

image<br />

Intensity<br />

hyperintense<br />

hyperintense<br />

hyperintense<br />

isointense<br />

hypointense<br />

Pt.<br />

No.<br />

10<br />

12<br />

4<br />

5<br />

8<br />

RESULTS<br />

ADC<br />

Intensity<br />

hypointense<br />

hypointense<br />

hypointense<br />

Isointense<br />

hyperintense<br />

Pt.<br />

No<br />

10<br />

12<br />

4<br />

5<br />

8


-94-<br />

RESULTS


Illustrative cases<br />

- 118 -<br />

Illustrative cases<br />

Case 1 (Fig.43): Hyperacute ischemic stroke with D/P mismatch and<br />

penumbra.<br />

Female patient, 90 years old presented with quadriplegia.<br />

MRI was done 6 hours after clinical presentation.<br />

(a) T1WI is normal. T2WI (b) and FLAIR (c) show subtle increase signal<br />

intensity involving most of the left cerebral hemisphere.<br />

(d) DWI (b1000) shows abnormal signal intensity lesion involving most<br />

of the left cerebral hemisphere and a small area in the right and right<br />

basal ganglia regions displaying high signal intensity with brightness<br />

(restricted diffusion).<br />

(e) Exponential image shows hyperintense signal in the same area.<br />

(f) ADC map shows low signal intensity of the same area.<br />

(g-k) perfusion maps show a larger lesion in the left cerebral hemisphere<br />

with central dead area and surrounding zone of hypoperfusion (delayed<br />

TTP and increased MTT).<br />

(l) Time signal intensity curve shows: ROIs reveal normal one (area 1)<br />

showing T0=37 sec, TTP=44 sec and MTT=11.5 sec.The central area<br />

(area 2) shows flat curve with no cerebral blood flow. The peripheral area<br />

(area 3) shows wide curve with delayed TTP=49 sec (delay 5 sec from<br />

normal curve), increase MTT=19 sec (increase 7.5 sec from normal<br />

curve).<br />

(m) axial and (n) coronal 3D TOF MRA show non filling of both ICAs.<br />

Right posterior cerebral artery (PCA) is slightly compansatory<br />

hypertrophied to supply the right cerebral hemisphere through anastmosis<br />

with the right posterior communicating artery. The left posterior cerebral<br />

artery is not good visualized.<br />

(o) MR spectroscopy (MRS) of the infarction site shows presence of<br />

lactate, decreased level of NAA, creatine and choline.


- 119 -<br />

Illustrative cases


- 120 -<br />

Illustrative cases


- 121 -<br />

Illustrative cases


- 122 -<br />

Illustrative cases<br />

Case 2 (Fig.44): Hyperacute ischemic stroke with D/P mismatch and<br />

penumbra.<br />

Male patient, 72 years old presented with right sided hemiplegia.<br />

MRI was done 5 hours after clinical presentation.<br />

(a) Axial CT is completely normal.<br />

Axial T1 (b) and T2WI (c) are completely normal.<br />

(d) Axial FLAIR shows subtle intensity at the left high parietal region.<br />

DWI-b0 (e) is normal however DWI-b1000 (f) shows hyperintense lesion<br />

with brightness at the left high parietal region. The infarction shows<br />

hyperintensity at exponential image (g) and low signal intensity at ADC<br />

map (h).<br />

Perfusion Weighted Images (i-m) show larger lesion in the left parital<br />

region with central dead area and peripheral are of hypoperfusion.<br />

(n) Time signal intensity curve shows 3ROIs. ROI 1 shows normal curve<br />

with TTP 60 sec and MTT 16 sec. ROI2 (central dead area) shows<br />

flattened curve with no cerebral blood flow. ROI 3 (peripheral<br />

hypoperfused area) shows wide curve with TTP 72 sec and MTT 22 sec<br />

[delayed TTP 12 sec and increased MTT 6 sec in comparison with the<br />

normal curve].<br />

Axial (o) and coronal (p) 3D TOF MRA show obstruction of some of the<br />

cortical branches of the left Middle Cerebral Artery (MCA) and non<br />

filling of the right Anterior Cerebral Artery (ACA).<br />

(q) MRS of the infarction show presence of small amount of lactate,<br />

reduction of the level of NAA, creatine and choline.


- 123 -<br />

Illustrative cases


- 124 -<br />

Illustrative cases


- 125 -<br />

Illustrative cases


Case 3 (Fig.45): Hyperacute ischemic stroke with D/P matching.<br />

Male patient, 59 years old presented with left sided hemiplegia.<br />

- 126 -<br />

Illustrative cases<br />

Radiological examination was done 5 hours after clinical presentation.<br />

(a) Axial CT is completely normal.<br />

Axial T1 (b) and T2WI (c) and FLAIR (d) are completely normal.<br />

(e) DW images-b0 are completely normal however DWI-b1000 (f) show<br />

multiple infarcts displaying high signal intensity with brightness<br />

(restricted diffusion) at the right temporal, right midbrain and both<br />

thalamic regions.<br />

At exponential images (g) the lesions are hyperintense however they are<br />

of low signal intensity at ADC maps (h).<br />

PW images (I1-5) show the infarcted region as seen on DWI(D/P<br />

matching).<br />

Time signal intensity curve (I6) shows the infarcted area (area 2)<br />

represented by flat curve with no CBF and the normal contralateral area<br />

(area 2) represented by normal curve with T0=26sec, TTP=32 sec and<br />

MTT=9sec.<br />

Axial and coronal 3D TOF MRA (j) show occlusion of right Posterior<br />

cerebral artery (PCA) with non filling of right posterior communicating<br />

artery.<br />

MRS at the infarction site (k) shows presence of lactate and reduction of<br />

NAA, creatine and choline levels.


- 127 -<br />

Illustrative cases


- 128 -<br />

Illustrative cases


- 129 -<br />

Illustrative cases


Case 4 (Fig. 46): Hyperacute ischemic stroke with D/P matching.<br />

Male patient, 74 years old presented with left sided wakness.<br />

Radiological examination was done 6 hours after clinical presentation.<br />

(a) Axial CT is completely normal.<br />

Axial T1W images (b&c) are normal.<br />

- 130 -<br />

Illustrative cases<br />

T2WI (d) and FLAIR (e) show subtle area of increased signal intensity at the<br />

right basal ganglia (right lentiform).<br />

DW image-b0 (f) is normal however DWI-b1000 (g-h) show high signal<br />

intensity area with brightness (restricted diffusion) at the right basal ganglia<br />

and periventricular regions mainly at the right side.<br />

At exponential images (i-j) the infarction is hyperintense however they are of<br />

low signal intensity at ADC maps (k-l).<br />

Post contrast T1W images (m-n) show right cortical vascular enhancement at<br />

the disterbution of right MCA.<br />

PW images (o-s) show the infarction size as seen on DWI (D/Pmatching).<br />

Time signal intensity curve (t) shows ROIs revealing :ROI 1 on the infarcted<br />

area is represented by flat curve with no cerebral blood flow ,ROI 3 on the<br />

contralateral normal area revealing normal curve with T0=24 sec, TTP=30.2<br />

sec and MTT=11.7 sec and ROI 2 on the normal area surrounding the<br />

infarction revealing normal curve with T0=22.9 sec, TTP=30.4 sec and<br />

MTT=11.4 sec. Axial (u) and coronal (v) 3D TOF MRA show obstruction of<br />

the right ICA with filling of the right MCA and ACA from contralateral ICA<br />

through the circle of Wills associated with attenuation of right ACA and MCA<br />

and their branches.<br />

MRS (v) of the infarcted area showspresence of lactate, reduction of NAA and<br />

creatine levels with slight increase in choline level.


- 131 -<br />

Illustrative cases


- 132 -<br />

Illustrative cases


- 133 -<br />

Illustrative cases


- 134 -<br />

Illustrative cases


- 135 -<br />

Illustrative cases<br />

Case 5 (Fig. 47): Hyperacute ischemic stroke DWI lesion without PWI<br />

abnormality.<br />

Female patient, 80 years old presented with coma.<br />

Radiological examination was done 6 hours after clinical presentation.<br />

(a) Axial CT is completely normal.<br />

Axial T1W images (b) , T2WI (c) and FLAIR (d) are normal.<br />

DW image-b0 (e) is normal however DWI-b1000 (f) shows high signal<br />

intensity area with brightness (restricted diffusion) at the left side of pons.<br />

At exponential images (g) the infarction is hyperintense however it is of<br />

low signal intensity at ADC maps (h).<br />

On PW images (i-m) there is no apparent perfusion abnormality.<br />

Time signal intensity curve (n) : ROI 1at the infarction site on DWI<br />

shows normal curve with TTP=30 sec and MTT=11 sec and ROI 2 on<br />

normal site on DWI shows normal curve with TTP=31 sec and MTT=10<br />

sec.<br />

Coronal 3D TOF MRA (o) shows no apparent abnormality .<br />

MRS (p) of the infarcted area showspresence of lactate, reduction of<br />

NAA and creatine levels with normal choline level.


- 136 -<br />

Illustrative cases


- 137 -<br />

Illustrative cases


- 138 -<br />

Illustrative cases


Case 6 (Fig.48): Hyperacute ischemic stroke .<br />

- 139 -<br />

Illustrative cases<br />

Female patient, 80 years old presented with right sided hemiplegia.<br />

Radiological examination was done 6 hours after clinical presentation.<br />

(a) Axial CT is completely normal.<br />

Axial T1W image (b) is completely normal.<br />

T2WI (c) and FLAIR (d) and DW image-b0 (e) show mild<br />

hyperintensity of the left basal ganglia (left lentiform).<br />

DWI-b1000 (f) show high signal intensity area with brightness (restricted<br />

diffusion) at the left basal ganglia.<br />

At exponential images (g) the infarction is hyperintense however they are<br />

of low signal intensity at ADC maps (h).<br />

Post contrast coronal T1W images (i-j) show left cortical vascular<br />

enhancement at the disterbution of left MCA.<br />

Axial non enhanced CT cuts at the level of the left MCA (k-l) show<br />

hyperattenuation along the proximal segment of the left MCA(hyperdense<br />

MCA sign) denoting dense thrombus.<br />

Axial 2D TOF MRA (m) shows absence of flow of the left MCA.<br />

Axial (n) and coronal (o) 3D TOF MRA show obstruction of the<br />

proximal segment of the left MCA (M1).<br />

MRS (p) of the infarcted area shows presence of lactate, reduction of<br />

NAA and creatine levels with normal choline level.


- 140 -<br />

Illustrative cases


- 141 -<br />

Illustrative cases


- 142 -<br />

Illustrative cases


Case 7 (Fig.49): Hyperacute ischemic stroke .<br />

Male patient, 69 years old presented with coma.<br />

- 143 -<br />

Illustrative cases<br />

Radiological examination was done 6 hours after clinical presentation.<br />

(a) Axial non enhanced CT cuts are completely normal.<br />

Axial T1W images (b) are normal.<br />

T2WI (c) and FLAIR (d) and DW images-b0 (e) show small faintly<br />

intense lesions at both thalami.<br />

DW images-b1000 (f) show multiple hyperintense lesions with brightness<br />

(restricted diffusion) at right occipital, midbrain and both thalamic<br />

regions.<br />

At exponential images (g) the infarctions are hyperintense however they<br />

are of low signal intensity at ADC maps (h).<br />

Axial (i) and coronal (j) 3D TOF MRA show non filling of the distal<br />

basilar and right posterior cerebral artery (PCA). The left PCA is not well<br />

visualized (filling from the left posterior communicating artery). The<br />

proximal segment of the left anterior cerebral (A1) is not visualized.<br />

MRS (k) of the infarcted site shows presence of lactate, slight reduction<br />

of NAA and creatine levels with normal choline level.


- 144 -<br />

Illustrative cases


- 145 -<br />

Illustrative cases


- 146 -<br />

Illustrative cases


Case 8 (Fig.50): Acute ischemic stroke with D/P matching.<br />

Male patient, 65 years old presented with rigt sided wakness.<br />

- 147 -<br />

Illustrative cases<br />

Radiological examination was done 12 hours after clinical presentation.<br />

(a) Axial CT shows faint ill defined hypodense area at the left basal<br />

ganglia (left caudate nucleus).<br />

Axial T1W image (b) shows faint hypointense area at the left caudate<br />

nucleus.<br />

Axial T2WI (c) ,FLAIR (d) and DW images-b0 (e) show subtle area of<br />

increased signal intensity at the left basal ganglia .<br />

DWI-b1000 (f) shows hyperintense infarction with brightness (restricted<br />

diffusion) at the left basal ganglia(left caudate nucleus and lentiform) .<br />

At exponential images (g) the infarction displays high signal intensity<br />

however on ADC map (h) it displays low signal intensity.<br />

PW images (i-m) show the infarction size as seen on DWI<br />

(D/Pmatching).<br />

Time signal intensity curve (n) : ROI 2 on the infarcted area is<br />

represented by flat curve with no cerebral blood flow ,ROI 1 on the<br />

contralateral normal area revealing normal curve with T0=24.9 sec,<br />

TTP=31.8 sec and MTT=10 sec and ROI 3 on the normal area<br />

surrounding the infarction revealing normal curve with T0=26.6 sec,<br />

TTP=33.1 sec and MTT=9.8 sec.<br />

Post contrast T1WI (o) is normal.<br />

Coronal 3D TOF MRA (p) shows no abnormality.<br />

MRS (q) of the infarcted area showspresence of lactate, reduction of<br />

NAA and creatine and choline levels.


- 148 -<br />

Illustrative cases


- 149 -<br />

Illustrative cases


- 150 -<br />

Illustrative cases


Case 9 (Fig. 51 ): Acute ischemic stroke.<br />

Male patient, 47 years old presented with right sided wakness.<br />

- 151 -<br />

Illustrative cases<br />

Radiological examination was done 14 hours after clinical presentation.<br />

Axial T1W image (a) shows small faint hypointense area at the left basal<br />

ganglia.<br />

Axial T2WI (b) ,FLAIR (c) and DW images-b0 (d) show mildly intense<br />

small lesion at the left basal ganglia.<br />

The lesion is markedly hyperintense with brightness on DWI-b1000 (e)<br />

and hyperintense on exponential images (f). On ADC map (g) it displays<br />

low signal intensity.<br />

PW images (k-h) show the infarction size as seen on DWI<br />

(D/Pmatching).<br />

Time signal intensity curve (l) : ROI 1 on the infarcted area is represented<br />

by flat curve with no cerebral blood flow however ROI 2 on the<br />

contralateral normal area revealing normal curve with T0=25.5 sec,<br />

TTP=31.9 sec and MTT=9.2 sec .<br />

Post contrast T1WI (m) is normal.<br />

Axial (n) and coronal (o) 3D TOF MRA show no abnormality.<br />

MRS (p) of the infarcted area shows presence of lactate, reduction of<br />

NAA and creatine levels with normal choline level.


- 152 -<br />

Illustrative cases


- 153 -<br />

Illustrative cases


- 154 -<br />

Illustrative cases


Case 10 (Fig. 52): Acute ischemic stroke .<br />

Female patient, 45 years old presented with left sided wakness.<br />

- 155 -<br />

Illustrative cases<br />

Radiological examination was done 14 hours after clinical presentation.<br />

(a) Axial CT shows subtle small hypodense area at the right lateral aspect<br />

of pons.<br />

Axial T1W image (b) is normal.<br />

Axial T2WI (c) ,FLAIR (d) and DW images-b0 (e) show subtle intensity<br />

at the right lateral aspect of pons .<br />

DWI-b1000 (f) shows small bright hyperintense infarction (restricted<br />

diffusion) at the right lateral aspect of pons).<br />

At exponential images (g) the infarction displays high signal intensity<br />

however on ADC map (h) it displays low signal intensity.<br />

Coronal 3D TOF MRA (i) shows no abnormality.<br />

MRS (j) of the infarcted area showspresence of lactate, reduction of<br />

NAA,normal creatine and increased choline levels.


- 156 -<br />

Illustrative cases


- 157 -<br />

Illustrative cases


Case 11 (Fig.53): Acute ischemic stroke .<br />

Female patient, 60 years old presented with dysarthria.<br />

- 158 -<br />

Illustrative cases<br />

Radiological examination was done 15 hours after clinical presentation.<br />

(a-b) Axial CT cuts are normal.<br />

Axial T1W image (c) is normal.<br />

Axial T2WI (d) ,FLAIR (e) and DW images-b0 (f) show mild<br />

hyperintensity at the right high parietal region .<br />

DWI-b1000 (g) shows bright hyperintense infarction (restricted diffusion)<br />

at the right high parietal region).<br />

At exponential images (h) the infarction displays high signal intensity<br />

however on ADC map (i) it displays low signal intensity.<br />

Axial 3D TOF MRA (j) shows no apparent abnormality.<br />

MRS (k) of the infarcted area shows presence of lactate, reduction of<br />

NAA and creatine levels with normal choline level.


- 159 -<br />

Illustrative cases


- 160 -<br />

Illustrative cases


Case 12 (Fig.54): Subacute ischemic stroke .<br />

Male patient, 40 years old presented with ataxia.<br />

- 161 -<br />

Illustrative cases<br />

Radiological examination was done 8 days after clinical presentation.<br />

Axial T1W image (a) shows small hypointense area at the right cerebellar<br />

hemisphere adjacent to the fourth ventricle.<br />

The infarction shows high signal intensity at axial T2WI (b) ,FLAIR (c)<br />

and DW images-b1000 (d).<br />

It displays nearly intermediate signal intensity at exponential images (e)<br />

and ADC map (f).


- 162 -<br />

Illustrative cases


Case 13 (Fig.55): Chronic ischemic stroke .<br />

Male patient, 27 years old presented with left sided hemiplegia.<br />

- 163 -<br />

Illustrative cases<br />

Radiological examination was done 1 month after clinical presentation.<br />

(a) Axial CT shows an ill defined hypodense area of infarction at the<br />

right basal ganglia.<br />

The infarction is of low signal intensity at axial T1WI (b) and high signal<br />

intensity at axial T2WI (c) ,FLAIR (d) and DW images-b0 (e) however it<br />

is isointense on DWI-b1000 (f) , low signal intensity at exponential image<br />

(g) and high signal intensity on ADC map (h).<br />

Coronal 3D TOF MRA (i) shows no apparent abnormality.<br />

MRS (k) of the infarcted area shows presence of lactate, reduction of<br />

NAA and creatine levels with increase in choline level.


- 164 -<br />

Illustrative cases


- 165 -<br />

Illustrative cases


Case 14 (Fig.56): Chronic ischemic stroke .<br />

Male patient, 65 years old presented with left sided weakness.<br />

- 166 -<br />

Illustrative cases<br />

Radiological examination was done 6 monthes after clinical presentation.<br />

(a) Axial CT shows a well defined hypodense area of infarction at the<br />

right high patietal region.<br />

The infarction is of low signal intensity at axial T1WI (b) and high signal<br />

intensity at axial T2WI (c) ,FLAIR (d) and DW images-b0 (e). At DWIb1000<br />

(f) the infarction displays low to intermediate signal intensity<br />

however at exponential image (g) it displays low signal intensity and high<br />

signal intensity at ADC map (h).<br />

PW images (i-m) show the non perfused infarcted area.<br />

Time signal intensity curve (n) : ROI 2 on the infarcted area is<br />

represented by flat curve with no cerebral blood flow ,ROI 1 on the<br />

contralateral normal area revealing normal curve with T0=26.1 sec,<br />

TTP=33.9 sec and MTT=11.8 sec and ROI 3 on the normal area<br />

surrounding the infarction revealing normal curve with T0=26.1 sec,<br />

TTP=34.1 sec and MTT=12.3 sec.<br />

Post contrast axial T1WI (o) shows no vascular or parenchymal<br />

enhancement.<br />

Coronal 3D TOF MRA (o) shows no abnormality.<br />

MRS (p) of the infarcted area shows presence of lactate, reduction of<br />

NAA and creatine levels with increase in choline level.


- 167 -<br />

Illustrative cases


- 168 -<br />

Illustrative cases


- 169 -<br />

Illustrative cases


Case 15 (Fig.57): Hyperacute intracerebral hemorrhagic stroke .<br />

Male patient, 70 years old presented with right sided hemiplegia.<br />

- 170 -<br />

Illustrative cases<br />

Radiological examination was done 7 hours after clinical presentation.<br />

(a) Axial non enhanced CT shows fresh blood density at both basal<br />

ganglia regions associated with intraventricular extension.<br />

MRI through the level of the left basal ganglia hematoma shows<br />

abnormal signal intensity lesion at the left basal ganglia surrounded by<br />

mild perifocal oedema, causing effacement of the left lateral ventricle and<br />

associated with intraventricular extension displaying the same signl<br />

intensity.<br />

The lesion displays intermediate to low signal intensity at axial T1WI (b),<br />

high signal intensity surrounded by low signal intensity rim at T2WI (cd)<br />

and FLAIR (f) and heterogenous signal intensity at GRE image (e).<br />

The lesion displays high signal intensity at DWI-b1000 (g) and<br />

exponential image (h) and low signal intensity on ADC map (i).<br />

Axial (j) and coronal (k) 3D TOF MRA show non visualization of the<br />

left MCA with non good visualization of both lenticulostriate arteries.


- 171 -<br />

Illustrative cases


- 172 -<br />

Illustrative cases


Case 16 (Fig.58): Acute intracerebral hemorrhagic stroke .<br />

Male patient, 19 years old presented with right sided hemiplegia.<br />

MRI examination was done 3 days after clinical presentation.<br />

- 173 -<br />

Illustrative cases<br />

It shows abnormal signal intensity lesion (hematoma) at the left parieto-<br />

occipital region, surrounded by mild perifocal oedema,and causing mass<br />

effect in the form of effacement of the left lateral ventricle .<br />

It displays low signal intensity with high signal intensity peripheral zone<br />

(peripheral intracellular methemoglobin of early subacute hemorrhage) at<br />

axial T1WI (a) and low signal intensity at T2WI (b) , FLAIR (c) and<br />

DWI-b0 (d). It also displays low signal intensity at DWI-b1000 (e) due to<br />

magnetic susceptability effect and low signal intensity on ADC map (f).<br />

Axial (g) and coronal (h) 3D TOF MRA show the hematoma as well as<br />

attenuation os the distal cortical segment of left MCA.


- 174 -<br />

Illustrative cases


- 175 -<br />

Illustrative cases


- 176 -<br />

Illustrative cases<br />

Case 17 (Fig.59): Late subacute intracerebral hemorrhagic stroke .<br />

Male patient, 41 years old presented with right sided weakness.<br />

MRI examination was done 13 days after clinical presentation.<br />

It shows abnormal signal intensity lesion (hematoma) at the left basal<br />

ganglia, surrounded by mild perifocal oedema.<br />

It displays high signal intensity at axial T1 (a) , T2WI (b) , FLAIR (c) ,<br />

DWI-b0 (d) , DWI-b1000 (e) and ADC map (f).


- 177 -<br />

Illustrative cases


- 178 -<br />

Illustrative cases<br />

Case 18 (Fig.60):Subarachnoid hemorrhage (SAH) with rupture<br />

inracranial aneurysm.<br />

Female patient, 59 years old presented with sever headche, vomiting and<br />

confusion.<br />

Axial CT examination (a-b) done 4 hours after presentation shows fresh<br />

blood density at the subarachnoid space of the basal cisterns associated<br />

with mild hydrocephalic changes.<br />

MRI examination done 6 days after clinical presentation the<br />

subarachnoid hemorrhage can not be seen at all MR sequence except<br />

FLAIR image (e) in which it displays high signal intensity.<br />

Mild hydrocephalic changes are seen associated with trans-ependymal<br />

leak displaying high signal intensity at T2WI (d), FLAIR (e) , DWI -b0<br />

(f) and ADC map (i) and low signal intensity at T1WI (c) , DW-b1000 (g)<br />

and exponential images (h).<br />

Coronal (k-m) and axial (n) 3D TOF MRA show aneurysmal dilatation<br />

of the supraclinoid portion of the left ICA.<br />

Digital Subtraction angiography (o-r) shows aneurysmal dilatation of the<br />

supraclinoid portion of the left ICA.


- 179 -<br />

Illustrative cases


- 180 -<br />

Illustrative cases


- 181 -<br />

Illustrative cases


Case 19 (Fig.61):Subarachnoid hemorrhage .<br />

Male patient, 34 years old presented with seizors.<br />

- 182 -<br />

Illustrative cases<br />

Axial CT examination (a-b) done 5 hours after presentation shows fresh<br />

blood density at the subarachnoid space mainly of the right sylvian<br />

fissure associated with mild hydrocephalic changes.<br />

MRI examination done 5 days after clinical presentation.<br />

The SAH shows high signal intensity at T1W images (c-d) and FLAIR<br />

images (g-h) and low signal intensity at T2WI (e) and Gradient (f).<br />

It displays low signal intensity on DWI-b0 (i), high signal intensity at<br />

DWI-b1000 (j) and exponential images (k) however on ADC it shows<br />

low signal intensity (l) as well as mild hydrocephalic changes.<br />

Axial 2D TOF MRA (m) shows normal flow in the right MCA without<br />

aneurysmal dilatation.<br />

Coronal 3D phase contrast MRA (n) and 3D TOF MRA (o) show<br />

attenuation (spasm) of the right MCA without aneurysmal dilatation.<br />

Digital Subtraction angiography (p-q) is normal.


- 183 -<br />

Illustrative cases


- 184 -<br />

Illustrative cases


- 185 -<br />

Illustrative cases


DISCUSSION<br />

- 186 -<br />

DISCUSSION<br />

Stroke can be defined as an acute central nervous system injury with<br />

an abrupt onset (Srinivasan et al.,2006).<br />

Stroke is a leading cause of mortality and morbidity in the developed<br />

world. The goals of an imaging evaluation for acute stroke are to<br />

establish a diagnosis as early as possible and to obtain accurate<br />

information about the intracranial vasculature and brain perfusion for<br />

guidance in selecting the appropriate therapy (Srinivasan et al.,2006).<br />

Although strokes are much more common in people over 65, and many<br />

people believe that strokes only happen to old folks, strokes can occur at<br />

any age, including infancy, childhood, adolescence and early adulthood<br />

(Caplan, 2006).<br />

Our study included 50 patients, 32males(64%) and 18 females(36%),<br />

their ages ranged from 19 to 90 years, they are classified into 8 groups<br />

according to their ages with 10 years interval between each group. Stroke<br />

patients were most commonly seen in 60-70 years age group. Their<br />

number were 19 patients (38%). This is followed by 50- 60 age group in<br />

which their number was 11 patients (22%) then 40- 50 age group in<br />

which their number was 8 patients (16%) and 70- 80 age group in which<br />

their number was 7 patients(14%).<br />

This result was in agreement with the study of Somay et al., 2005<br />

study about cerebrovacular stroke risk factors and stroke subtypes in<br />

different age groups included 401 patients. They were 199 males and 202<br />

females. The most common age groups were 71-80 and 61-70years,<br />

however the difference in sex prevalence between our study and their<br />

study may be caused by the small number of patients in our study. We are


- 187 -<br />

DISCUSSION<br />

also in agreement with Provenzale and Barboriak, 1997 regarding the<br />

patient age who stated that Strokes occur relatively infrequently in young<br />

adults (i.e., individuals who are 15-45 years old).<br />

The most common risk factors in our study were hypertension (15<br />

cases-30%), ischemic heart disease (11 cases-22%), Diabetes mellitus (9<br />

cases-18%) and hyperlipidemia (7 case-14%) and smoking (6 cases-<br />

12%).This is consistent with Somay et al., 2005 in which the<br />

hypertension (60.1%) was the most common risk factor followed by<br />

ischemic heart disease (40.1%), hypercholesterolemia (29.2%), smoking<br />

(31.4%) and Diabetes mellitus (21.4%) and with Eguchi et al., 2003 who<br />

stated that Diabetes mellitus (DM), is a major risk factor for stroke and<br />

also with Smoller et al., 2000 who reported that Hypertension is a major<br />

risk factor for stroke and heart disease among both men and women.<br />

The first task of imaging is to divide the strokes into ischemic (85%)<br />

or hemorrhagic (15%) subtypes. Distinction of hemorrhage versus<br />

infarction is the initial critical branch point in acute stroke triage, and<br />

directs care toward medical therapy or tailored intervention such as<br />

endovascular aneurysm coiling or thrombolysis. Hemorrhage is most<br />

efficiently excluded by CT, but can also be reliably assessed using MR<br />

imaging, which includes both T2*-weighted and FLAIR sequences<br />

(Rowley, 2001).<br />

Our patients are subjected to CT scanning then divided into two<br />

main groups; ischemic stroke (39 patients, 78%) and hemorrhagic stroke<br />

(11 patients, 22%). This is consistent with Srinivasan et al., 2006 who<br />

stated that acute ischemia constitutes approximately 80% of all strokes<br />

and is an important cause of morbidity and mortality and Haaga et al,<br />

2003 who said that cerebral infarction accounts for approximately 85% of


- 188 -<br />

DISCUSSION<br />

all strokes and with (Weissleder et al., 2003) who reported that 80% of<br />

strokes are due to cerebral ischemia.<br />

Temporal evolution of strokes is typically categorized into<br />

hyperacute (0–6 h), acute (6–24 h), subacute (24 h to approximately 2<br />

weeks), and chronic stroke (>2 weeks old) (Gonzalez, 2006).<br />

The first group, ischemic stroke patients (39 patients), are<br />

chronological divided into five stages. The hyperacute stage (1 st 6 hours)<br />

including 10 patients, the acute stage (6-24hours) including 12 patients,<br />

the early subacute stage(1-7 days) including 4 patients, late subacute<br />

stage(1-2 weeks) including 5 patients and chronic stage(>2weeks)<br />

including 8 cases. All these patients are subjected to different MRI<br />

techniques.<br />

Multiparametric stroke imaging combining diffusion (DWI) and<br />

perfusion-weighted MRI (PWI), MR angiography (MRA), and<br />

conventional MR sequences is increasingly used as the primary<br />

diagnostic imaging modality in major stroke centers (Fiehler et al, 2003).<br />

We had 39 ischemic stroke patients, all of them are subjected to<br />

conventional MRI and DWI, 37 patients are subjected to MRA, 34<br />

patients to MRS, 17 patient to PWI, 11 patients to Post contrast T1WI<br />

and only 1 case is subjected to MRV examination, however all<br />

hemorrhagic stroke patients are subjected to conventional MRI , DWI and<br />

MRA examination.<br />

A growing body of evidence accumulated during recent years has<br />

documented the superiority of magnetic resonance (MR) imaging as<br />

compared with computed tomography (CT) in the clinical setting of acute<br />

ischemic stroke not only in the diagnosis of hyperacute ischemic and<br />

hemorrhagic stroke but also in the proper selection of patients who might<br />

benefit from reperfusion therapy (Rovira et al., 2004).


- 189 -<br />

DISCUSSION<br />

Conventional computed tomography (CT) and MR imaging cannot<br />

be used to reliably detect infarction at the earliest time points. The<br />

detection of hypoattenuation on CT scans and hyperintensity on T2-<br />

weighted MR images requires a substantial increase in tissue water. DW<br />

images are very sensitive and specific for the detection of hyperacute and<br />

acute infarctions, with a sensitivity of 88-100% and a specificity of 86-<br />

100%. A lesion with decreased diffusion is strongly correlated with<br />

irreversible infarction (Schaefer et al., 2000).<br />

Diffusion-weighted MR imaging appears to be sensitive to an early<br />

pathoghysiologic process in cerebral infarction. The loss of adenosine<br />

triphosphate, which causes a subsequent shift of water from the<br />

extracellular space into the intracellular space and possibly an increase in<br />

the intracellular viscosity.<br />

Superior contrast-to-noise ratio (CNR) of acute stroke lesions on<br />

diffusion-weighted MR imaging compared with CT and conventional MR<br />

imaging (Gonzalez et al.,1999).<br />

FLAIR improved the conspicuity of ischemic lesions in comparison<br />

with T2 and T1, especially when lesions were located in the cortical<br />

ribbon were CSF contamination and different MR properties of GM can<br />

impair lesion conspicuity (Schaller, 2007).<br />

Our results are in agreement with Schaefer et al.,2000, Gonzalez et<br />

al.,1999 and Schaller, 2007 as we had 10 cases of hyperacute ischemic<br />

stroke, one case of them is diagnosed by T2WI and 9 cases not<br />

diagnosed. On FLAIR images 6 cases shows subtle increase of signal<br />

intensity and 4 cases not diagnosed however the ten cases are diagnosed<br />

by DWI (hyperintense with brightness). We also had 12 cases of acute<br />

ischemic stroke , 3 of them are diagnosed on T2WI (hyperintense ), 7<br />

cases showed faint hyperintensity and 2 cases not diagnosed however on


- 190 -<br />

DISCUSSION<br />

FLAIR images 11 cases are diagnosed (hyperintense) and one case not<br />

appear but in DWI the all 12 cases are diagnosed (hyperintense with<br />

brightness).These results are also in agreement with Wilcock et al., 1999<br />

and Karonen et al, 1999 who reported that Diffusion weighted imaging<br />

demonstrates changes in acute infarction before conventional T2<br />

weighted or CT images become positive and with Perkins et al.,2003<br />

who concluded that DWI nearly doubles the likelihood of detecting acute<br />

ischemic stroke lesions compared with FLAIR for all etiologies and in all<br />

anatomic locations.<br />

It is important to understand that DW image has T2-weighted<br />

contrast, the DW image can be divided by the echo-planer spin-echo T2-<br />

weighted (or b=0 sec/mm2) image to give an "exponential<br />

image"(Schaefer et al.,2000).<br />

A detailed understanding of the serial evolution of the brain’s MRI<br />

relaxation and diffusion parameters is clinically important for the<br />

diagnosis and treatment of stroke patients. For example, it can be used to<br />

help estimate the age of a lesion, to establish rational time windows for<br />

stroke treatment, or perhaps to provide alternative outcome measures or<br />

“surrogate end points (Liu et al, 2007).<br />

In humans, decreased diffusion in ischemic brain tissue is observed<br />

as early as 30 min after vascular occlusion. The ADC continues to<br />

decrease with peak signal reduction at 1–4 days. This decreased diffusion<br />

is markedly hyperintense on DWI (a combination of T2 and diffusion<br />

weighting), less hyperintense on exponential images, and hypointense on<br />

ADC images. The ADC returns to baseline at 1–2 weeks. This is<br />

consistent with the persistence of cytotoxic edema (associated with


- 191 -<br />

DISCUSSION<br />

decreased diffusion) as well as cell membrane disruption, and the<br />

development of vasogenic edema (associated with increased diffusion).<br />

At this point, a stroke is usually mildly hyperintense on the DWI images<br />

due to the T2 component and isointense on the ADC and exponential<br />

images. Thereafter, the ADC is elevated secondary to increased<br />

extracellular water, tissue cavitation, and gliosis. There is slight<br />

hypointensity, isointensity or hyperintensity on the DWI images<br />

(depending on the strength of the T2 and diffusion components),<br />

increased signal intensity on ADC maps, and decreased signal on<br />

exponential images (Gonzalez et al., 2006).<br />

This is consistent with our study as we found decreased ADC in<br />

the hyperacute stage with more decrease in the acute stage and it started<br />

to increase to reach near normal levels in the late subacute stage with<br />

more increase in the chronic stage, by other means the rADC (ratio<br />

between ADC in infarction and contralateral normal site) is 0.5308±<br />

0.0899 in the hyperacute, 0.3765± 0.06954 in the acute stage, 0.7002±<br />

0.1231in the early subacute, 1.0135± 0.03422 in late subacute and<br />

2.2094± 0.8806 in chronic stage.<br />

In our study, we found that patients with hyperacute stroke (in the<br />

first 6 hours-10 patients) the infarction could be visualized in DWI b<br />

1000 (hyperintensity with brightness), ADC map (hypointensity) and<br />

exponential images (hyperintensity) however 9 patients couldn’t be seen<br />

on the DWI b0 or T2-wieghted image . In the FLAIR 4 patients out of<br />

them were not appear completely while the remaining 6 patients showed<br />

slight intensity.<br />

Patients with acute stroke (6-24 hours-12 patients) the infarction<br />

could be visualized in DWI b 1000 (hyperintensity with brightness), ADC<br />

map (hypointensity) and exponential images (hyperintensity) however on


- 192 -<br />

DISCUSSION<br />

the DWI b0 and T2-wieghted image 2 patients couldn’t be seen, 7 cases<br />

showed faint hyperintensity and 3 patients are hyperintense . In the<br />

FLAIR 1 patients out of them was not appear completely while the<br />

remaining 11 patients showed hyperintensity.<br />

This is consistent with Schaefer et al., 2000 who observed<br />

restricted diffusion associated with acute ischemia 30 minutes after a<br />

witnessed ictus. The ADC continues to decrease and is most reduced at 8-<br />

32hours. The ADC remains markedly reduced for 3-5 days. This<br />

decreased diffusion is markedly hyperintense on DW images and<br />

hypointense on ADC images and with Yang et al, 1999 who reported that<br />

acute infarcts appear hyperintense on DWI because of a reduction in the<br />

apparent diffusion coefficient (ADC) of water within minutes after the<br />

onset of ischemia. We are also in agreement with Keir and Wardlaw,<br />

2000 who stated that DWI works on the principle that sensitizing a<br />

standard MR image to diffusion weighting identifies regions of abnormal<br />

water movement, occurring very early after onset of ischemia, resulting in<br />

increased (bright) signal intensity. This is consistent also with Neumann-<br />

Haefelin et al 1999 and Krueger et al, 2000 who reported that with DWI<br />

it is possible to identify severely ischemic brain regions within minutes to<br />

hours after stroke onset. A decrease in the apparent diffusion coefficient<br />

of water (ADC), apparent as hyperintensity on DW images, indicates a<br />

restriction in the diffusional movement of water.<br />

In our study patients with early subacute stroke (1-7 days-4<br />

patients) showed hyperintensity in DWI (b1000 and b0),exponential<br />

image, T2-weighted and FLAIR images but hypointensity on ADC maps.<br />

Patients with late subacute stroke (1-2 weeks-5 patients) showed<br />

hyperintensity in DWI (b1000 and b0), T2-weighted and FLAIR images<br />

and isointense signal at exponential image and ADC maps.


- 193 -<br />

DISCUSSION<br />

Chronic stroke patients (>2weeks-8 patients) showed hyperintensity<br />

on T2-weighted, FLAIR images and DWI ( b0).On DWI ( b1000) 6 cases<br />

showed hyperintensity however 2 cases were isointense. The all 8 cases<br />

were hyperintense on ADC maps.<br />

These agree with Schaefer et al.,2000 who said that the ADC<br />

returns to baseline at 1-4 weeks. This most likely reflects persistence of<br />

cytotoxic oedema(associated with decreased diffusion) and development<br />

of vasogenic oedema and cell membrane disruption, leading to increased<br />

extracellular water (associated with increased diffusion). At this point, an<br />

infarction is usually mildly hyperintense due to the T2 component on DW<br />

images and isointense on the ADC images. Thereafter, diffusion is<br />

elevated as a result of continued increase in extracellular water, tissue<br />

caviation and gliosis. This elevated diffusion is characterized by slight<br />

hypointensity, isointensity or hyperintensity on DW images (depending<br />

on the strength of the T2 and ADC maps.<br />

These also agree with Huang et al., 2001 who reported that when<br />

the signal intensity changes on DW images were studied as a single<br />

parameter, abnormally high signal intensity was constantly observed at<br />

the acute and the subacute stages of cerebral infarction. This abnormal<br />

signal intensity increased with time and declined 2 weeks after symptom<br />

onset. Lansberg et al., 2001 also reported that it is well accepted that<br />

ADC values decline rapidly after the onset of ischemia and subsequently<br />

increase, the observed time course of the ADC increase to supernormal<br />

values has varied from 24 hours to 17 days.<br />

While diffusion-weighted MR imaging is most useful for detecting<br />

irreversibly infarcted tissue (Srinivasan et al, 2006), perfusion imaging<br />

findings provide information on the momentary hemodynamic state of<br />

brain tissue, as they reveal impaired tissue perfusion caused by blood


- 194 -<br />

DISCUSSION<br />

vessel obstruction. Therefore, perfusion imaging findings may yield<br />

information about pathologically hypoperfused regions, even before<br />

genuine structural brain tissue damage has taken place (Wittsack et al,<br />

2002).<br />

Perfusion-weighted imaging (PWI) and diffusion weighted imaging<br />

(DWI) have been used with increasing frequency in acute ischemic<br />

stroke. PWI demonstrates areas of decreased blood flow, whereas DWI<br />

reveals regions of acute bioenergetic compromise ( Loh et al, 2005).<br />

The combination of DW imaging, perfusion- weighted imaging, and<br />

MR angiography enables identification of tissue at risk of infarction,<br />

which is characterized by the mismatch between an area of restricted<br />

diffusion and a larger area with ischemic but still viable tissue on<br />

perfusion-weighted images (Saur et al., 2003).<br />

A variety of hemodynamic images may constructed from perfusion<br />

data, including relative cerebral blood volume(rCBV), relative cerebral<br />

blood flow(rCBF), mean transit time (MTT)and time to peak (TTP) maps<br />

(Schaefer et al.,2000).<br />

Cerebral blood volume (CBV) is defined simply as the amount of<br />

blood in a given amount of tissue at any time (ml/g) as compared to CBF,<br />

which represents the amount of blood moving through a certain amount<br />

of tissue per unit time (ml/g per min). The ratio of CBV and CBF is<br />

defined as the mean transit time (units minutes) (Davis et al, 2003). Time<br />

to peak (TTP) maps of perfusion imaging show the arrival time of the<br />

contrast agent which is the same for the gray and white matter in the brain<br />

(Wittsack et al., 2002).<br />

In our study we used MTT and TTP maps for the correlation<br />

between the lesion size on perfusion weighted images and its size on DW<br />

images.


- 195 -<br />

DISCUSSION<br />

It is difficult to name one perfusion map type that could, even when<br />

combined with DW images, be used to predict infarct growth with<br />

satisfactory accuracy. Although the initial lesion volume on rCBV maps<br />

correlates best with the final infarct size and on average is closest to it,<br />

the initial rCBV map in many patients lead to underestimation of the final<br />

infarct size. Estimating the final infarct size with the initial rCBF finding<br />

seems to be more accurate. However, some infarcts grow to the area<br />

where the only initially detected imaging abnormality is prolonged MTT<br />

(Karonen et al., 2000).<br />

In an acute stroke setting, TTP maps offer two advantages over<br />

other images derived from perfusion imaging. First, they involve the least<br />

amount of post processing, and thus allows the clinical information to be<br />

available within 20 minutes. Second, TTP maps provide a relatively clear<br />

picture of the location and extent of the lesion (Wittsack et al., 2002).<br />

The volume of sever ischemia determined at a TTP delay of some 6<br />

seconds relative to the non affected hemisphere helped to predict the<br />

definite infarct lesion (Seitz et al.,2005).<br />

When most patients of hyperacute and acute infarction are evaluated<br />

by DW and perfusion-weighted MR imaging, their images usually<br />

demonstrate one of three patterns: A lesion is smaller on DW images than<br />

the same lesion on perfusion –weighted image; a lesion on DW images is<br />

equal to or larger than that on perfusion weighted images; or a lesion is<br />

depicted on DW images but not demonstrable on perfusion-weighted<br />

images (Schaefer et al.,2000).<br />

Schaefer et al.,2000 found that the large-vessel stroke lesion (such<br />

as in the proximal portion of the middle cerebral artery), the abnormality<br />

as depicted on perfusion-weighted images is frequently larger than the<br />

lesion as depicted on DW images, the peripheral region characterized by


- 196 -<br />

DISCUSSION<br />

normal diffusion and decrease perfusion, usually progress to infarction<br />

unless there is early reperfusion. Thus, in the acute setting, perfusion-<br />

weighted imaging in combination with DW imaging helps identify an<br />

operational "ischemic penumbra" or area at risk for infarction.<br />

On the other hand, with small-vessel infarction (perforator<br />

infarctions and distal middle cerebral artery infarctions) the initial lesion<br />

volumes on perfusion-weighted and DW images are usually similar, and<br />

the DWI lesion volume increases only slightly with time. A lesion larger<br />

on DW images than on perfusion weighted images or a lesion visible on<br />

DWI but not on perfusion weighted images usually occurs with early<br />

reperfusion. In this situation, the lesion on DW images usually does not<br />

change substantially over time.<br />

These are in agreement with our results as 8 patients of hyperacute<br />

and acute stroke in our series showed PWI> DWI, 7 patients of them<br />

were suffering from lesions in the distribution of large cerebral blood<br />

vessels . 6 patients of hyperacute, acute and chronic stroke showed<br />

DWI=PWI , their lesion were in the distribution of small vessels. 2<br />

patients showed DWI>PWI , and 1 patient showed lesion visible on DWI<br />

but not on perfusion weighted images ,their lesion were in the distribution<br />

of small vessels with reperfusion.<br />

These are also in agreement with Gonzalez et al., 2006 who stated that<br />

Proximal occlusions are much more likely to result in a diffusion–<br />

perfusion mismatch than distal or lacunar infarctions and with Neumann-<br />

Haefelin et al, 2000 who concluded that the total PWI/DWI mismatch is<br />

often larger in patients with ipsilateral carotid stenosis .70% than in<br />

patients without carotid stenosis.


- 197 -<br />

DISCUSSION<br />

In cases with diffusion perfusion mismatch and ischemic penumbra we<br />

found increase in the MTT and delay in TTP in the penumbra region<br />

compared with the normal contralateral site (MTT at the penumbra is<br />

17.6250± 2.7675 sec and 10.6000± 2.8087 sec at normal site, TTP at the<br />

penumbra is 44.7875± 12.1561sec and 35.9375± 11.2246 sec at normal<br />

site) denoting statistically significant difference between ischemic and<br />

contralateral normal site, p


- 198 -<br />

DISCUSSION<br />

the more distal, smaller branches and the level of diagnostic confidence<br />

diminishes (Davis et al, 2003).<br />

In acute stroke imaging (


- 199 -<br />

DISCUSSION<br />

marked vascular enhancement was observed in 2 patients of hypercaute<br />

stroke however mild enhancement was detected in 3 patients [ 2 cases of<br />

hyperacute stroke and 1 case of acute stroke] .So vascular enhancement<br />

was noted in hyperacute and acute stages not the chronic stages and this<br />

is consistent with Gonzalez et al., 2006.<br />

Magnetic Resonance Spectroscopy (MRS) has become one of the<br />

most important imaging techniques of CNS over 10 years, mainly<br />

because this method is enables noninvasive measurement of metabolites<br />

which play a central role in cellular metabolism (Kaminska et al, 2007).<br />

MRS has been suggested to be more sensitive than MRI in detecting<br />

hypoxic damage. It has been proposed that MRS may be able to detect<br />

cerebral ischemia within seconds of its onset as compared to DWI that<br />

provides warning as early as 1 h after the onset. The most striking<br />

changes in patients with acute cerebral infarction on MRS is the<br />

appearance of lactate with reduction in NAA and total Cr/PCr within the<br />

infarct compared to the contralateral hemisphere. Large variations in the<br />

initial concentrations of Cho have been observed in the region of<br />

infarction (Davis et al, 2003).<br />

34 cases of ischemic stroke patients are subjected to MR<br />

spectroscopic examination to the lesion (infarction) and the contralateral<br />

normal area (10 patients of hyperacute stroke, 10 patients of acute stroke,<br />

6 patients of subacute stroke and 8 patients of chronic stroke).<br />

Lactate is not normally detected within the brain and, as the end<br />

product of glycolysis, is a particularly useful measure of metabolism. The<br />

concentration of lactate rises when the glycolysis rate exceeds the tissue's<br />

capacity to catabolise it or remove it from the blood stream. The rise in<br />

brain lactate that results from the mismatch between glycolysis and<br />

oxygen supply, making it the hallmark for the detection of cerebral


- 200 -<br />

DISCUSSION<br />

ischemia. The persistence of lactate weeks or months following stroke<br />

onset has been observed. Removal of lactate depends on tissue perfusion,<br />

the permeability of the blood brain barrier (BBB) and diffusion of the<br />

metabolite through the damaged tissue (Saunders, 2000 ).<br />

In our study the lactate is present in all hyperacute(100%),<br />

acute cases(100%), 3 cases of early subacute(75%) , 1 cases of late<br />

subacute(50%) and 4 cases of chronic stroke(50%) with statistical<br />

significant difference in lactate level among different stroke stages<br />

(p


- 201 -<br />

DISCUSSION<br />

This is consistent with our study as all our patients in different stroke<br />

stages showed decrease in the level of NAA. We also in agreement with<br />

Graham et al, 2001, Pereira et al, 1999, Mathews et al., 1995, , Graham<br />

et al,1993 and Graham et al, 1992 who observed that lnfarcts were<br />

characterized by significantly increased lactate and significantly<br />

decreased NAA compared with contralateral brain regions. Our study is<br />

consistent with Gideon et al, 1992 who concluded that the NAA content<br />

in infarcted brain tissue is considerably reduced compared with normal<br />

brain tissue.<br />

Initial reductions in Cr/PCr are identified following infarction and<br />

further reductions have been demonstrated up to 10 days following the<br />

time of onset. The pathological correlate is thought to be gliosis of the<br />

tissue. The reduction in NAA in the infarct region is more marked than<br />

the reduction in Cr/PCr and this is thought to reflect the increased<br />

sensitivity of neurons to ischemia, compared to glial tissue (Davis et al,<br />

2003) and (Saunders, 2000 ).<br />

These are consistent with our study as 28 patients showed reduction<br />

in Cr/PCr and 6 patients were normal or showed slight increase of Cr/PCr<br />

level.<br />

The trimethylamine resonance of choline-containing compounds is<br />

present at 3.22 ppm. In normal brain, the Cho peak is thought to consist<br />

predominantly of glycerolphosphocholine and phosphocholine.<br />

Both compounds are involved in membrane synthesis and degradation.<br />

Reduction in the choline peak has been proposed as a marker of<br />

membrane damage. The choline peak has been shown to be increased,<br />

decreased and stays the same following cerebral infarction. The changes<br />

in the choline peak are thought to reflect changes in the MR visibility of<br />

the choline containing compounds that make up the cell membrane. We


- 202 -<br />

DISCUSSION<br />

have shown a fall and then a late rise, maximal 3 months after onset of<br />

stroke. This may represent loss of membrane function, followed by late<br />

gliosis (Davis et al, 2003).<br />

In our study the choline level was decreased in 22 out of 26 cases of<br />

early ischemic stroke (hyperacute, acute and subacute stages) however in<br />

chronic stage choline level is increased in 4 cases and decreased in 4<br />

cases.<br />

When comparing MRS findings in different ischemic stroke stages we<br />

found that there is a statistical significant difference in NAA level among<br />

different stroke stages(p


- 203 -<br />

DISCUSSION<br />

extension, 2 cases of subarachnoid hemorrhage and 1 case of<br />

intraventricular hemorrhage .<br />

The ability to detect acute hemorrhage by MRI is related to the<br />

oxygen saturation of hemoglobin and its degradation. As a hematoma<br />

ages, the hemoglobin passes through several forms (oxyhemoglobin,<br />

deoxyhemoglobin, and methemoglobin) prior to red cell lysis and<br />

breakdown into ferritin and hemosiderin (Davis et al, 2003).<br />

Based on the most mature form of the hemoglobin present in the clot,<br />

five stages of evolving ICH have been described. The hyperacute stage<br />

lasts for 24 h, acute stage for 2–3 days, early subacute stage for 3–7 days,<br />

late subacute stage up to 2 weeks and chronic stage more than 2 weeks.<br />

The evolution of an ICH is a complex and dynamic process and the<br />

estimation of the time of onset based on MRI findings may be inaccurate<br />

(Kummer and Back, 2006).<br />

It was initially believed that on MRI an ICH of less than 24 h<br />

duration could not be distinguished reliably either from a mass lesion or<br />

from an acute infarct, because of an unspecific pattern of isointensity in<br />

T1 and slight hyperintensity on T2 imparted by the predominance of<br />

oxyhemoglobin. However, due to a reduced oxygen tension at the<br />

periphery of the clot, some deoxygenation of hemoglobin occurs<br />

immediately after the bleeding, and several studies have demonstrated<br />

that the resulting susceptibility effects can be visualized very early on T2-<br />

and particularly on T2*-weighted images as a hypointense rim . It was<br />

reported that the ability of high field MRI to reliably detect a spontaneous<br />

ICH as early as 20 min after onset and schematized the appearance of the<br />

hyperacute hemorrhage as consisting of three distinct concentric regions.<br />

The center of the hemorrhage appears isointense or heterogeneously<br />

hyperintense on T2- and T2*-weighted images, probably reflecting the


- 204 -<br />

DISCUSSION<br />

presence of intact oxyhemoglobin. The periphery is hypointense on T2-<br />

and T2*-weighted images reflecting the susceptibility effects due to<br />

deoxyhemoglobin, and finally there is an outer rim that appears<br />

hypointense on T1-weighted images and hyperintense on T2- and T2*-<br />

weighted images, suggestive of edema (Kummer and Back, 2006).<br />

Diffusion MR data would differ among the stages of evolving<br />

hematomas in that hematomas containing blood with intact cell<br />

membranes would have restricted diffusion compared with those<br />

hematomas in which RBC membranes have lysed. This hypothesis was<br />

based on several facts. First, the presence of intact cell membranes<br />

restricts molecular diffusion (Atlas et al, 2000).<br />

Results of DW imaging of hematomas at this stage have not been<br />

well characterized; however a hyperacute intraparenchymal hemorrhage<br />

is hyperintense on DW images, with a decreased apparent diffusion<br />

coefficient (ADC). The possible causes for the decreased ADC are<br />

shrinkage of the extracellular space due to clot retraction, changes in the<br />

concentration of hemoglobin and a high viscosity (Moritani et al, 2005).<br />

These are consistent with our study as we had 2 cases of hyperacute<br />

hemorrhagic stroke. At conventional MRI the lesions showed<br />

intermediate signal intensity at T1, slightly high signal intensity at T2WI,<br />

FLAIR and Gradient images. The periphery of the lesions showed low<br />

signal intensity at T2WI, FLAIR and Gradient (T2*) images. On DW<br />

images the lesions showed high signal intensity (restricted diffusion) and<br />

low signal intensity on ADC maps. We are also in agreement with<br />

Linfante et al, 1999 who reported that hyperacute hematomas have<br />

increased signal intensity present on FLAIR and T2WI, and the<br />

hypointensity present on T1WI and with Fiebach et al, 2004 and Patel<br />

et al, 1996 who concluded that Intracranial hemorrhage less than 24 hours


- 205 -<br />

DISCUSSION<br />

old has been postulated to be in the oxyhemoglobin phase and would be<br />

isointense on T1-weighted images and slightly hyperintense on T2-<br />

weighted images, with additional feature in all the cases is a peripheral<br />

rim of hypointensity on both T1- and T2-weighted images.<br />

In the absence of rebleeding, deoxygenation of the clot is completed<br />

in most cases by 24 h. At that time deoxyhemoglobin becomes the<br />

predominant blood breakdown product in the lesion. Due to its<br />

paramagnetic properties and its compartmentalization within still intact<br />

RBCs, deoxyhemoglobin exerts a strong susceptibility effect, which<br />

makes acute hematomas appear distinctly and characteristically dark on<br />

T2 weighted SE images, and more so on T2* weighted GRE images . On<br />

T1-weighted images, acute ICHs still appear isointense or mildly<br />

hypointense, as in the hyperacute stage. The parenchyma surrounding the<br />

clot exhibits a halo due to vasogenic edema and initial inflammatory<br />

reaction. The region of edema has long T1 and T2 relaxation times and<br />

appears hypointense on T1-weighted images and hyperintense on T2-<br />

weighted images (Kummer and Back, 2006).<br />

Diffusion-weighted images of an acute hematoma show a marked<br />

hypointensity, caused by the magnetic field inhomogeneity created by the<br />

para-magnetic deoxy-hemoglobin. Although The ADC has been reported<br />

to be decreased, accurate calculations are often difficult (Moritani et al,<br />

2005).<br />

These is consistent with our results as we had 2 cases of acute<br />

intracerebral hematomas. At T1WI they showed intermediate signal<br />

intensity with hyperintense peripheral rim however at they showed low<br />

signal intensity at T2-weighted, FLAIR and Gradient images. They<br />

showed low signal intensity on DW images and ADC maps.


- 206 -<br />

DISCUSSION<br />

This is also in agreement with Schaefer et al.,2000 who reported<br />

that hemorrhage containing deoxyhemoglobin, intracellular<br />

methemoglobin and hemosiderin and hypointense on DW images because<br />

of magnetic susceptibility and with Kang et al., 2001 who observed that<br />

all acute hemorrhagic cases showed marked hypointensity on DW, T2-<br />

weighted, FLAIR and Gragient images. We are also in agreement with<br />

Schellinger et al, 1999 who concluded that the key substrate for MRI<br />

visualization of hemorrhage is deoxyhemoglobin, which causes a signal<br />

loss in T2-weighted imaging (T2-WI) because of paramagnetic<br />

susceptibility effects, although usually not within the first 12 to 24 hours.<br />

The appearance of methemoglobin in the clot marks the transition<br />

from the acute to the subacute phase. In the early subacute phase,<br />

methemoglobin is still compartmentalized within red blood cells and<br />

hence generates local field gradients that maintain a T2 shortening effect<br />

similar to that induced by deoxyhemoglobin. Therefore, in the early<br />

subacute phase relaxivity effects and susceptibility effects coexist, i.e. the<br />

clot appears bright in T1, and dark in T2 and especially in T2*. This stage<br />

is expected to last for approximately 1 week (Kummer and Back, 2006).<br />

On DW imaging, intracellular met-hemoglobin shows hypointensity<br />

due to paramagnetic susceptibility effects and ADC measurements are not<br />

reliable due to susceptibility effects (Moritani et al, 2005).<br />

In our study we had 1case of early subacute hemorrhage. The case<br />

showed high signal intensity on T1, low signal intensity at T2-weighted,<br />

FLAIR and Gradient images. The patient showed low signal intensity on<br />

DW images and ADC maps.<br />

This is consistent also with Kang et al., 2001 who observed that all<br />

early subacute hemorrhagic cases showed marked hypointensity on DW,<br />

T2-weighted, FLAIR and Gradient images and with Schaefer et al.,2000


- 207 -<br />

DISCUSSION<br />

who reported that hemorrhage containing deoxyhemoglobin, intracellular<br />

methemoglobin and hemosiderin and hypointense on DW images because<br />

of magnetic susceptibility.<br />

As the clot ages, progressive fragmentation and osmotic phenomena<br />

damage RBC membranes until they eventually lyse. Hemolysis releases<br />

methemoglobin into the extracellular fluid compartment of the<br />

hematoma. The resulting dilution of methemoglobin in the extracellular<br />

fluids eliminates the biological field gradients, and with them the<br />

susceptibility phenomena, an effect that is observed initially at the<br />

periphery of the clot. When all methemoglobin is extracellular, the T2-<br />

shortening induced signal loss is no longer observed, so that the late<br />

subacute ICH is predominantly hyperintense in both T1- and T2-weighted<br />

images (Kummer and Back, 2006).<br />

It has been reported that late subacute hematomas are hyperintense<br />

on DW imaging. The ADC value for the late subacute hematoma is<br />

controversial (Moritani et al, 2005).<br />

Extracellular methemoglobin has a higher ADC than does normal<br />

brain tissue, which indicates that the mobility of water in the extracellular<br />

space is increased. The prolongation of the T2 component of fluid with<br />

extracellular methemoglobin results in hyperintensity on DW images<br />

(Schaefer et al.,2000).<br />

This is consistent with our study as we had 2 cases of late subacute<br />

hematoma that showed hyperintense signal at T1, T2, FLAIR , Gradient ,<br />

DW images and ACD maps.<br />

We are in agreement with Kang et al., 2001 who observed that all<br />

cases of late subacute hematomas showed marked hyperintensity at<br />

diffusion-weighted, T1-, T2-weighted and FLAIR images.


- 208 -<br />

DISCUSSION<br />

A chronic ICH initially shows a hypointense peripheral hemosiderin<br />

rim completely surrounding a central area hyperintense in all sequences<br />

due to the persistence of extracellular methemoglobin. The hemosiderin<br />

rim is less conspicuous on T1-weighted sequences and more conspicuous<br />

on T2- and T2*-weighted sequences (Kummer and Back, 2006).<br />

Diffusion-weighted images are hyperintense in chronic hematomas.<br />

The ADC value has been reported to be increased, but this is often<br />

difficult to measure accurately due to paramagnetic susceptibility artifacts<br />

(Moritani et al, 2005).<br />

Hemorrhage containing deoxyhemoglobin, intracellular<br />

methemoglobin and hemosiderin and hypointense on DW images because<br />

of magnetic susceptibility (Schaefer et al.,2000).<br />

We are in agreement with Kummer and Back, 2006 and Schaefer et<br />

al.,2000 as we had 1 case of chronic intracerebral hematoma. It showed<br />

low signal intensity with small high signal intensity center at T1-, T2-<br />

weighted, FLAIR and Gradient images. It showed low signal intensity on<br />

DW images with increased ADC value.<br />

Subarachnoid Hemorrhage (SAH) is responsible of 3% of all acute<br />

strokes and of 5% of stroke-related deaths. Rupture of intracranial<br />

saccular aneurysm accounts for about 85% of cases of SAH (Kummer<br />

and Back, 2006).<br />

Ninety per cent of non-traumatic SAHs are caused by ruptured<br />

Berry aneurysms ( Howlett and Ayers, 2004).<br />

Our results are in agreement with Kummer and Back, 2006 and<br />

Howlett and Ayers, 2004 as we had 2 cases of subarachnoid hemorrhage<br />

4 % (out of 50 patients with stroke). One of them is caused by rupture of<br />

intracranial aneurysm (50%). The difference in incidence of ruptured


- 209 -<br />

DISCUSSION<br />

intracranial aneurysms as a causative factor is due to the small number of<br />

SAH cases in our study.<br />

Conventional T1–WI and T2–WI were shown to be ineffective in<br />

the diagnosis of acute SAH. The detection of SAH may be improved by<br />

using PD–WI with shorter repetition times (TR) to visualize the T1-<br />

effect. FLAIR sequences have been reported to be suitable for the<br />

diagnosis of not only subacute but also acute SAH (Davis et al, 2003).<br />

We are in agreement with Davis et al, 2003 as Our 2 cases were<br />

subacute. On case was detected by all pulse sequences, however the other<br />

case was detected by FLAIR images only.<br />

It is often difficult to detect subarachnoid hemorrhage on DW<br />

images. However, DW images may be useful to visualize parenchymal<br />

injuries secondary to subarachnoid hemorrhage. Ischemic changes,<br />

probably related to subarachnoid hemorrhage, have shown hyperintensity<br />

on DW images (Moritani et al, 2005).<br />

Most intraventricular hemorrhages (IVHs) are secondary to ICH or<br />

SAH. Isolated IVH is rare and usually due to the rupture of underlying<br />

vascular malformations. Since the CSF in ventricles has a higher content<br />

in oxygen and glucose, the layered form evolves more slowly compared<br />

to the clotted form. The latter evolves at a rate similar to ICH. However,<br />

in the chronic stage, unlike ICH, IVHs do not show hemosiderin<br />

formation. Among the conventional sequences, proton density depicts<br />

IVH better than T1- and T2-weighted images. In the acute stage FLAIR<br />

may have better sensitivity than CT scan to demonstrate IVH. On FLAIR,<br />

in the initial 48 h IVH appears hyperintense (Kummer and Back, 2006).<br />

DW images can demonstrate intraventricular hemorrhages, but in general<br />

the GRE images have a higher sensitivity (Moritani et al, 2005).


- 210 -<br />

DISCUSSION<br />

We had 2 cases of intraventricular hemorrhage. One of them is<br />

associated with hyperacute intracerebral hematoma and showed the same<br />

signal intensity. The other case was acute and showed intermediate signal<br />

intensity at T1 and low signal intensity at all other pulse sequences.<br />

MR angiography has recently been established as a valid and<br />

sensitive tool to demonstrate pathologic findings in extra- and intracranial<br />

arteries in acute stroke (Seitz et al, 2005).<br />

Magnetic resonance angiography (MRA) was done to all our<br />

hemorrhagic stroke cases (11 patients), 5 patients showed positive finding<br />

[spasm in 2 cases and non filling in 1 case of intracerebral hematoma –<br />

aneurysm in 1 case and spasm in 1 case of SAH].<br />

The presence of a significant quantity of blood in the brain disrupts<br />

the homogeneity of the magnetic field due to the presence of the iron<br />

from the hemoglobin molecule, and spectroscopy cannot be carried out in<br />

patients with large intracerebral hemorrhages (Davis et al, 2003). So MR<br />

spectroscopic study was not included in our hemorrhagic stroke cases.


Summary and Conclusion<br />

- 211 -<br />

Summary& Conclusion<br />

Despite the efforts being made to develop effective treatments,<br />

ischemic stroke remains a common cause of death and disability in the<br />

industrialized world. It has been claimed that new imaging methods could<br />

provide valuable information in developing new therapies.<br />

Any magnetic resonance (MR) imaging protocol for acute ischemic<br />

stroke must be able to depict early ischemic changes in a sensitive manner.<br />

This can be achieved with diffusion-weighted (DW) and perfusion-<br />

weighted (PW) MR imaging. DW MR imaging depicts infarcted tissue<br />

within 5 minutes after the occlusion of the feeding vessel. PW imaging is<br />

able to depict hypoperfused brain tissue around the infarcted core. The<br />

mismatch between lesion sizes at PW and DW imaging (perfusion-diffusion<br />

mismatch) in the acute phase has been considered as an estimate of the<br />

ischemic penumbra and is helpful in selecting patients for different<br />

treatment groups (Karonen et al.,2000).<br />

Our study included 50 patients, 32males (64%) and 18 females (36%),<br />

their ages ranged from 19-90 with mean age 59.6±14.1 years. The most<br />

common risk factors in our study were hypertension (15 cases-30%)<br />

followed by ischemic heart disease (11 cases-22%).<br />

Our patients are divided into two main groups; ischemic stroke (39<br />

patients, 78%) and hemorrhagic stroke (11 patients, 22%).<br />

The first group, ischemic stroke patients (39 patients), are<br />

chronological divided into five stages. The hyperacute stage (1 st 6 hours)<br />

including 10 patients, the acute stage (6-24hours) including 12 patients, the<br />

early subacute stage (1-7 days) including 4 patients, late subacute stage(1-2


- 212 -<br />

Summary& Conclusion<br />

weeks) including 5 patients and chronic stage(>2weeks) including 8 cases.<br />

All these patients are subjected to different MRI techniques.<br />

On DWI we found decreased ADC in the hyperacute stage with<br />

more decrease in the acute stage and it started to increase to reach near<br />

normal levels in the late subacute stage with more increase in the<br />

chronic stage.<br />

Most of hyperacute and acute ischemic stroke patients (17 patients)<br />

were evaluated by perfusion weighted images (PWI). We used MTT and<br />

TTP maps for the correlation between the lesion size on perfusion weighted<br />

images and its size on DW images.<br />

Eight (8) patients of hyperacute and acute stroke in our series showed<br />

PWI> DWI, 7 patients of them were suffering from lesions in the<br />

distribution of large cerebral blood vessels . 6 patients of hyperacute, acute<br />

and chronic stroke showed DWI=PWI , their lesion were in the distribution<br />

of small vessels. 2 patients showed DWI>PWI , and 1 patient showed lesion<br />

visible on DWI but not on perfusion weighted images ,their lesion were in<br />

the distribution of small vessels with reperfusion.<br />

Postcontrast T1 was done in 11 patients. Vascular enhancement was<br />

positive in 5 patients out of them. Vascular enhancement was noted in<br />

hyperacute and acute stages not the chronic stages.<br />

Thirty four (34) cases of ischemic stroke patients are subjected to MR<br />

spectroscopic examination. Lactate was present in all hyperacute, acute<br />

cases, and most of subacute and chronic stages. Decrease in the level of<br />

NAA in all our patients in different stroke stages, reduction in Cr/PCr in<br />

most patients and varied level of choline amoung patients.


- 213 -<br />

Summary& Conclusion<br />

Our study included 11 patients of hemorrhagic stroke, they were 8<br />

cases with intracerebral hemorrhage; one of them showed intraventricular<br />

extension, 2 cases of subarachnoid hemorrhage and 1 case of<br />

intraventricular hemorrhage.<br />

We had 2 cases of hyperacute hemorrhagic stroke. At conventional<br />

MRI the lesions showed intermediate signal intensity at T1, slightly high<br />

signal intensity at T2WI, FLAIR and Gradient images. The periphery of the<br />

lesions showed low signal intensity at T2WI, FLAIR and Gradient (T2*)<br />

images. On DW images the lesions showed high signal intensity and low<br />

signal intensity on ADC maps. We had also 2 cases of acute intracerebral<br />

hematomas. At T1WI they showed intermediate signal intensity with<br />

hyperintense peripheral rim however at they showed low signal intensity at<br />

T2-weighted, FLAIR and Gradient images. They showed low signal<br />

intensity on DW images and ADC maps. We had 1case of early subacute<br />

hemorrhage. The case showed high signal intensity on T1, low signal<br />

intensity at T2-weighted, FLAIR and Gradient images. The patient showed<br />

low signal intensity on DW images and ADC maps. We had 2 case of late<br />

subacute hematoma that showed hyperintense signal at T1, T2, FLAIR,<br />

Gradient, DW images and ACD maps. We had 1 case of chronic<br />

intracerebral hematoma. It showed low signal intensity with small high<br />

signal intensity center at T1-, T2- weighted, FLAIR and Gradient images. It<br />

showed low signal intensity on DW images with increased ADC value.<br />

We had 2 cases of subarachnoid hemorrhage 4 % (out of 50 patients<br />

with stroke). One of them is caused by rupture of intracranial aneurysm<br />

(50%). Our 2 cases were subacute. On case was detected by all pulse<br />

sequences, however the other case was detected by FLAIR images only.<br />

We had 2 cases of intraventricular hemorrhage. One of them is<br />

associated with hyperacute intracerebral hematoma and showed the same


- 214 -<br />

Summary& Conclusion<br />

signal intensity. The other case was acute and showed intermediate signal<br />

intensity at T1 and low signal intensity at all other pulse sequences.<br />

MRA showed positive data in 5 out of 11 patients.<br />

Conclusion:<br />

*Diagnosis of ischemic stroke has been improved by the use of magnetic<br />

resonance(MR) imaging.<br />

*Diffusion Weighted Images (DWI) is superior to CT and conventional<br />

MRI sequences in early detection of hyperacute and acute ischemic stroke.<br />

*The combination DWI with Perfusion Weighted Images (PWI) enables<br />

identification of tissue at risk of infarction which is characterized by the<br />

mismatch between an area of restricted diffusion and a larger area with<br />

ischemic but still viable tissue on perfusion-weighted images.<br />

* MRA can pinpoint the cause of infarction mainly in large proximal<br />

vessels than distal smaller ones more in hyperacute and acute ischemic<br />

stages.<br />

*Contrast-enhanced T1-weighted images can show arterial enhancement in<br />

hyperacute and strokes due to slow or collateral flow.<br />

*The most striking changes in patients with cerebral infarction on MRS is<br />

the appearance of lactate with reduction in NAA and total Cr/PCr with<br />

variations in the concentrations of Cho.<br />

*Initial CT is important in early cases of stroke to diagnose Intracranial<br />

hemorrhage.<br />

*Based on the most mature form of the hemoglobin present in the clot, five<br />

stages of evolving ICH have been described. Each has specific appearance<br />

on conventional MRI and DW images.<br />

*MR angiography is a sensitive tool to demonstrate pathologic findings in<br />

extra- and intracranial arteries in hemorrhagic stroke as occlusion, spasm or<br />

intracranial aneurysms.


REFERENCES<br />

- 215 -<br />

References<br />

Akopov S. and Whitman G.T.(2002) : Hemodynamic Studies<br />

in Early Ischemic Stroke Serial Transcranial Doppler and Magnetic<br />

Resonance Angiography Evaluation. Stroke ;33:1274-1279.<br />

Atlas S.W., DuBois P., Singer M.B.and Lu D.(2000):<br />

Diffusion Measurment in Intracranial Hematomas: Implications for<br />

MR Imaging of Acute Stroke.AJNR;21:1190-1194.<br />

Beaucbamp N.J., Ulug A.M., Passe T.J. and Van Zijl<br />

P.C.M. (1998): MR Diffusion Imaging in Stroke: Review and<br />

Controversies. RadioGraphics; 18:1269-1283.<br />

Brant W.E. and Helms C.A.(2007): Fundamentals of<br />

Diagnostic Radiology, 2 nd edition, Lippincott Williams& Wilkins;<br />

chapter 2 Pp 30-44.<br />

Brown J.J., Hesselink J.R. and Rothrock J.F.(1988): MR<br />

and CT of Lacunar lnfarcts. AJR ;151:367-372.<br />

Brown M.A. and Semelka R.C.(2003): MRI-Basic Principles<br />

and Applications, 3 rd edition, Wiley-Liss, A John Wiley& Sons, Inc.,<br />

Hoboken, New Jersey, Canada;151-169.<br />

Butler P., Mitchell A.W.M. and Ellis H. (1999): Applied<br />

Radiological Anatomy, Cambridge University press, New York,<br />

Melbourne, Madrid, Cape Town, Singapore, Sao Paulo; chapter 2 Pp<br />

17-60.<br />

Caplan L.R. (2006): Stroke, Aan Press , American Academy<br />

of Neurology; chapter 1 Pp 4-20.


- 216 -<br />

References<br />

Chen P.E., Simon J.E., Hill M.D., Sohn C.H., Dickhoff P.,<br />

Morrish W.F., Sevick R.J.and Frayne R.(2006): Acute Ischemic<br />

Stroke: Accuracy of Diffusion-Weighted MR Imaging-Effects of b<br />

Value and Cerebrospinal Fluid Suppression. Radiology; 238(1):232-<br />

239.<br />

Crisostomo R.A., Garcia M.M.,and Tong D.C. (2003):<br />

Detection of Diffusion-Weighted MRI Abnormalities in Patients With<br />

Transient Ischemic Attack Correlation With Clinical Characteristics.<br />

Stroke; 34:932-937.<br />

Davis S., Fisher M. and Warach S.(2003): Magnetic<br />

Resonance Imaging In Stroke, Cambridge University Press, New<br />

York, Melbourne, Madrid, Cape Town, Singapore, Sao Paulo; 55-158.<br />

Debrey S.M., Yu H., Lynch J.K., Olof Lovblad K., Wright<br />

V.L., Janket S.J.D. , Baird A.E. (2008) : Diagnostic Accuracy of<br />

Magnetic Resonance Angiography for Internal Carotid Artery<br />

Disease. A Systematic Review and Meta-Analysis. Stroke; 39:2237-<br />

2248.<br />

Duijn J.H., Matson G.B., Maudsley A.A., Hugg J.W. and<br />

Weiner M.W.(1992): Human Brain Infarction: Proton MR<br />

Spectroscopy. Radiology; 183:711-718.<br />

Ebisu T, Tanaka C, Umeda M, Kitamura M, Fukunaga M,<br />

Aoki I, Sato H, Higuchi T, Narusa S, Horikawa Y and Ueda S.<br />

(1997) :Hemorrhagic and Non hemorrhagic Stroke: Diagnosis with<br />

Diffusion-Weighted and T2-Weighted Echo-planer MR Imaging.<br />

Radiology; 203: 823-828.


- 217 -<br />

References<br />

Eguchi K., Kario K.and Shimada K. (2003): Greater Impact<br />

of Coexistence of Hypertension and Diabetes on Silent Cerebral<br />

Infarcts. Stroke.; 34: 2471-2474.<br />

Federico F., Simone I.L., Lucivero V., Giannini P.,<br />

Laddomada G., Mezzapesa D.M.,and Tortorella C.(1998):<br />

Prognostic Value of Proton Magnetic Resonance Spectroscopy in<br />

Ischemic Stroke. Arch Neurol;55:489-494.<br />

Fiebach J.B., Schellinger P.D., Gass A., Kucinski T., Siebler<br />

M., Villringer A., Ölkers P., Hirsch J.G., Heiland S., Wilde P.,<br />

Jansen O., Röther J., Hacke W., and Sartor K. (2004): Stroke<br />

Magnetic Resonance Imaging Is Accurate in Hyperacute Intracerebral<br />

Hemorrhage A Multicenter Study on the Validity of Stroke Imaging.<br />

Stroke; 35:502-507.<br />

Fiehler J., Knudsen K., Kucinski T., Kidwell C.S., Alger<br />

J.R., Thomalla G., Eckert B., Wittkugel O., Weiller C., Zeumer<br />

H.,and Röther J. (2004) : Predictors of Apparent Diffusion<br />

Coefficient Normalization in Stroke Patients. Stroke; 35:514-519.<br />

Gazzaniga M.S., Ivry R.B. and Mangun G.R. (2002):<br />

Cognitive Neuroscience The biology of the Mind, 2 nd edition, Norton<br />

& Company; chapter 3.<br />

Gideon P., Henriksen D., Sperling B., Christiansen P., Olsen<br />

T.S., Jorgensen H.S., and Soborg P.A.(1992) : Early Time Course<br />

of JV-Acetylaspartate, Creatine and Phosphocreatine, and Compounds<br />

Containing Choline in the Brain After Acute Stroke. A Proton<br />

Magnetic Resonance Spectroscopy Study. Stroke ; 23:1566-1572.


- 218 -<br />

References<br />

Gillard J.H., Barker P.B., van Zijl P.C.M., Bryan R.N.,and<br />

Oppenheimer S.M.(1996): Proton MR Spectroscopy in Acute Middle<br />

Cerebral Artery Stroke. AJNR; 17:873–886.<br />

Gonzalez R.G., Hirsch J.A., Koroshetz W.J., Lev M.H. and<br />

Schaefer P.(2006) : Acute ischemic stroke: Imaging and Intervention,<br />

Springer, Berlin, Heidelberg, New York ; 246-284.<br />

Gonzalez R.G., Schaefer P.W., Buonanno F.S., Schwamm<br />

L.H., Budzik R.F., Rordorf G., Wang B., Sorensen A.G. and<br />

Koroshetz WJ. (1999) :Diffusion Weighted MR Imaging: Diagnostic<br />

Accuracy in patients Imaged within 6 Hours of Stroke Symptom<br />

Onset. Radiology;210: 155-162.<br />

Graham G.D., Blamire A.M., Howseman A.M., Rothman<br />

D.L., Fayad P.B., Brass L.M., Petroff O.A.C., Shulman R.G., and<br />

Prichard J.W. (1992) : Proton Magnetic Resonance Spectroscopy of<br />

Cerebral Lactate and Other Metabolites in Stroke Patients. Stroke ;<br />

23:333-340.<br />

Graham G.D. ,Blamire A.M., Rothman D.L., Brass L.M.,<br />

Fayad P.B., Petroff O.A.C.,and Prichard J.W. (1993): Early<br />

Temporal Variation of Cerebral Metabolites After Human Stroke. A<br />

Proton Magnetic Resonance Spectroscopy Study. Stroke; 24:1891-<br />

1896.<br />

Graham G.D., Hwang J.H., Rothman D.L.,and Prichard<br />

J.W. (2001) : Spectroscopic Assessment of Alterations in<br />

Macromolecule and Small-Molecule Metabolites in Human Brain<br />

After Stroke. Stroke. ;32:2797-2802.<br />

Grandian C.B., Duprez T.P., Smith A.M., Oppenheim C.,<br />

Peeters A., Robert A.R. and Cosnard G.(2002): Which MR-derived<br />

Perfusion Parameters are the Best Predictors of Infarct Growth in


- 219 -<br />

References<br />

Hyperacute Stroke? Comparative Study between Relative and<br />

Quantitative Measurments.Radiology;223: 361-370.<br />

Haaga J.R., Lanzieri C.F. and Gilkeson R.C.(2003):<br />

Computed Tomography And Magnetic Resonance Imaging of the<br />

whole body, 4 th edition, Mosby, St Louis , Baltimore, Berlin, Boston,<br />

Carlsbad, Chicago, London, Madrid, Naples, New York, Philadelphia,<br />

Sydney, Tokyo, Toronto; vol 1 chapter 7 Pp 246-284.<br />

Howlett D.C. and Ayers B., (2004): The hands-on guide to<br />

imaging, Blackwell Publishing, Oxford, London, Edinburg, Malden,<br />

Paris, chapter 12 Pp 195-213.<br />

Huang I.J., Chen C.Y., Chung H.W., Chang D.C., Lee C.C.,<br />

Chin S.C. and Liou M.(2002) : Time Course of Cerebral Infarction<br />

in the Middle Cerebral Arterial Territory: Deep Watershed versus<br />

Territorial Subtypes On Diffusion-Weighted MR Images.Radiology;<br />

221:35-42.<br />

Jansen J.F.A., Backes W.H., Nicolay K., Kooi M.E. (2006):<br />

1H MR Spectroscopy of the Brain: Absolute Quantification of<br />

Metabolites. Radiology; 240 (2):318-332.<br />

Kaminska K., Walecki J., Grieb P., Bogorodzki P.(2007):<br />

Magnetic resonance spectroscopy-State of art and future. Polish<br />

Journal of Radiology; 72(1):71-75.<br />

Kang B.K., Na D.G., Ryoo J.W., Byun H.S., Roh H.G. and<br />

Pyeun Y.S.(2001): Diffusion-Weighted MR Imaging of Intracerebral<br />

Hemorrhage. Korean J Radiol ;2(4):183-191.<br />

Karonen J.O., Liu Y., Vanninen R.L., Ostergaard L.,<br />

Partanen P.L.K., Vainio E.J., Vanninen E.J., Nuutinen J.,<br />

Roivainen R., Soimakallio S., Kuikka J.T. and Aronen H.J.(2000):


- 220 -<br />

References<br />

Combined Perfusion and Diffusion Weighted MR Imaging in Acute<br />

ischemic Stroke during the 1 st Week: A Longitudinal Study.<br />

Radiology; 217:886-894.<br />

Karonen J.O., Vanninen R.L., Liu Y., Østergaard L.,<br />

Kuikka J.T., Nuutinen J., Vanninen E.J., Partanen P.L.K., Vainio<br />

P.A., Korhonen K., Perkio J., Roivainen R., Sivenius J.,and<br />

Aronen H.J. (1999) : Combined Diffusion and Perfusion MRI With<br />

Correlation to Single-Photon Emission CT in Acute Ischemic Stroke<br />

Ischemic Penumbra Predicts Infarct Growth.Stroke;30:1583-1590.<br />

Keir S.L. ,and Wardlaw J. M. (2000) : Systematic Review of<br />

Diffusion and Perfusion Imaging in Acute Ischemic Stroke.<br />

Stroke;31:2723-2731.<br />

Kesavadas C., Fiorelli M., Gupta A.K., Pantano P., Bozzao<br />

L.and Kapilamoorthy T.R.(2003) : Diffusion weighted magnetic<br />

resonance imaging in acute ischemic stroke. Indian Journal of<br />

Radiology and Imaging ;13(4): 433-440.<br />

Kim H.J., Choi C.G., Lee DH, Lee J.H., Kim S.J. and Suh<br />

D.C. (2005): High b-Value Diffusion MR Imaging of Hyperacute<br />

Ischemic Stroke at 1.5T. AJNR; 26:208-215.<br />

Kim H.S., Lee D.H., Ryu C.W., Lee J.H., Choi C.G., Kim<br />

S.J.and Suh D .C. (2006): Multiple Cerebral Microbleeds in<br />

Hyperacute Ischemic Stroke: Impact on Prevalence and Severity of<br />

Early Hemorrhagic Transformation After Thrombolytic Treatment.<br />

AJR; 186:1443–1449.<br />

Krueger K., Kugel H., Grond M., Thiel A., Maintz D.,and<br />

Lackner K. (2000) : Late Resolution of Diffusion-Weighted MRI<br />

Changes in a Patient With Prolonged Reversible Ischemic


- 221 -<br />

References<br />

Neurological Deficit After Thrombolytic Therapy. Stroke; 31:2715-<br />

2718.<br />

Kummer R.V. and Back T. (2006): Magnetic Resonance<br />

Imaging in Ischemic Stroke, Springer, Berlin, Heidelberg, New York<br />

;23-249.<br />

Lansberg MG, Thijs VN, O'Brien MW, Ali JO, Cresping<br />

AJ, Tong DC, Moseley ME and Albers GW.(2001): Evolution of<br />

Apparent Diffusion Coefficient, Diffusion-Weighted and T2-<br />

Weighted Signal intensity of Acute Stroke. AJNR;22:637-644.<br />

Lee D. H., Kang D.W., Ahn J.S., Choi C. G, Kim S.J.,and<br />

Suh D.C. (2005) : Imaging of the Ischemic Penumbra in Acute<br />

Stroke. Korean J Radiol ;6:64-74.<br />

Linfante I., Llinas R.H., Caplan L.R., and Warach S. (1999)<br />

: MRI Features of Intracerebral Hemorrhage Within 2 Hours. From<br />

Symptom Onset. Stroke;30:2263-2267.<br />

Liney G.(2005) : MRI from A to Z,A Definitive Guide for<br />

Medical <strong>Prof</strong>essionals, Cambridge, New York, Melbourne, Madrid,<br />

Cape Town, Singapore, São Paulo: Pp6.<br />

Liu Y., D’Arceuil H.E., Westmoreland S., He J., Duggan M.,<br />

Gonzalez R.G. , Pryor J. ,and de Crespigny A.J. (2007): Serial<br />

Diffusion Tensor MRI After Transient and Permanent Cerebral<br />

Ischemia in Nonhuman Primates. Stroke; 38:138-145.<br />

Liu Y., Karonen J.O., Vanninen R.L., Nuutinen J., Koskela<br />

A., Soimakallio S., Aronen H.J. (2004): Acute Ischemic Stroke:<br />

Predictive Value of 2D Phase-Contrast MR Angiography—Serial<br />

Study with Combined Diffusion and Perfusion MR Imaging.<br />

Radiology ; 231:517–527.


- 222 -<br />

References<br />

Loh P.S., Butcher K.S., Parsons M.W., MacGregor L.,<br />

Desmond P.M., Tress B.M. ,and Davis S.M. (2005) : Apparent<br />

Diffusion Coefficient Thresholds Do Not Predict the Response to<br />

Acute Stroke Thrombolysis. Stroke. ;36:2626-2631.<br />

Makkat S., Vandevenne J.E., Verswijvel G., Ijsewijn T.,<br />

Grieten M., Palmers Y., De Schepper A.M. and Parizel P.M.<br />

(2002):Signs of Acute Stroke Seen on Fluid-Attenuated Inversion<br />

Recovery MR Imaging. AJR;179: 237–243.<br />

Mathews V.P., Barker P.B., Blackband S.J., Chatham J.C.<br />

and Bryan1 R.N. (1995): Cerebral Metabolites in Patients with Acute<br />

and Subacute Strokes: Concentrations Determined by Quantitative<br />

Proton M R Spectroscopy. AJR;165: 633-638.<br />

McRobbie D.W., Moore E.A., Graves M.J. and Prince M.R.<br />

(2006): MRI From Picture to Proton, Second edition, CAMBRIDGE<br />

UNIVERSITY PRESS, New York, Melbourne, Madrid, Cape Town,<br />

Singapore, Sao Paulo, chapter 13 Pp 258-281.<br />

Mori S. (2007): Introduction to Diffusion Tensor Imaging, 1 st<br />

edition, Elsevier B.V., Amsterdam, Oxford; chapter 4 Pp 33-40.<br />

Moritani T., Ekholm S.,and Westesson P.L. (2005):<br />

Diffusion-Weighted MR Imaging of the Brain, Springer, Berlin,<br />

Heidelberg, New York ,chapter 6 Pp 55-68.<br />

Mullins M.E. (2006): Modern Emergant Stroke Imaging:<br />

Pearls, Protocols, and Pitfalls. Radilogic Clinics of North America;<br />

44: 41-62.<br />

Mullins M.E., Schaefer P.W., Sorensen A.G., Halpern E.F.,<br />

Ay H., He J., Koroshetz W.J. and Gonzalez R.G.(2002): CT and<br />

Conventional and Diffusion –Weighted MR Imaging in Acute stroke:


- 223 -<br />

References<br />

Study in 691 patients at presentation to the Emergancy Department.<br />

Radiology 224:353-360.<br />

Nazliel B., Starkman S., Liebeskind D.S., Ovbiagele B.,<br />

Kim D., Sanossian N., Ali L., Buck B., Villablanca P., Vinuela F.,<br />

Duckwiler G., Jahan R.and Saver J.L.(2008) : A Brief Prehospital<br />

Stroke Severity Scale Identifies Ischemic Stroke Patients Harboring<br />

Persisting Large Arterial Occlusions. Stroke; 39:2264-2267.<br />

Neumann-Haefelin T., Wittsack H.J., Fink G.R., Wenserski<br />

F., Li T.Q., Seitz R.J., Siebler M., Modder U.,and Freund H.J.<br />

(2000): Diffusion- and Perfusion-Weighted MRI Influence of Severe<br />

Carotid Artery Stenosis on the DWI/PWI Mismatch in Acute Stroke.<br />

Stroke;31:1311-1317.<br />

Neumann-Haefelin T., Wittsack H.J., Wenserski F., Siebler<br />

M., Seitz R.J., Modder U., and Freund H.J.(1999) : Diffusion- and<br />

Perfusion-Weighted MRI The DWI/PWI Mismatch Region in Acute<br />

Stroke. Stroke; 30:1591-1597.<br />

Oppenheim C., Grandin C., Samson Y., Smith A., Duprez<br />

T., Marsault C.,and Cosnard G.(2001) : Is There an Apparent<br />

Diffusion Coefficient Threshold in Predicting Tissue Viability in<br />

Hyperacute Stroke?. Stroke. ;32:2486-2491.<br />

Patel M.R., Edelman R.R.,and Warach S.(1996) : Detection<br />

of Hyperacute Primary Intraparenchymal Hemorrhage by Magnetic<br />

Resonance Imaging. Stroke; 27:2321-2324.<br />

Pereira A.C., Saunders D.E., Doyle V.L., Bland J.M., Howe<br />

F.A., Griffiths J.R., and Brown M.M. (1999) : Measurement of<br />

Initial N-Acetyl Aspartate Concentration by Magnetic Resonance<br />

Spectroscopy and Initial Infarct Volume by MRI Predicts Outcome in


- 224 -<br />

References<br />

Patients With Middle Cerebral Artery Territory Infarction. Stroke;<br />

30:1577-1582.<br />

Perkins C.J., Kahya E., Roque C.T., Roche P.E.,and<br />

Newman G.C.(2001) : Fluid-Attenuated Inversion Recovery and<br />

Diffusion- and Perfusion-Weighted MRI Abnormalities in 117<br />

Consecutive Patients With Stroke Symptoms. Stroke ;32:2774-2781.<br />

Prosser J., Butcher K., Allport L., Parsons M., MacGregor<br />

L., Desmond P., Tress B. and Davis S. (2005): Clinical-Diffusion<br />

Mismatch Predicts the Putative Penumbra With High Specificity.<br />

Stroke; 36:1700-1704.<br />

Provenzale J.M. and Barboriak D.P.(1997) : Brain Infarction<br />

in Young Adults: Etiology and Imaging Findings. AJR ;169:1 161-1<br />

168.<br />

Provenzale J.M. and Sorensen A.G. (1999) : Diffusion-<br />

Weighted MR Imaging in Acute Stroke: Theoretic Considerations and<br />

Clinical Applications. AJR ;173:1459-1467.<br />

Rajeshkannan R., Moorthy S., Sreekumar K.P. , Rupa R.<br />

and Prabhu N.K. (2006):Clinical Application of Diffusion Weighted<br />

MR Imaging:A Review. Ind J Radiol Imag;16(4): 705-710.<br />

Reimer P.,Parizel P.M. and Stichnoth F.A.(2006): Clinical<br />

MR Imaging, A practical Approach, 2 nd edition, Springer, Berlin<br />

Heidelberg New York; chapter 4 Pp 65-76.<br />

Rima K., Rohit G., Anjali P., and Veena C. (2003) : Role of<br />

diffusion weighted MR images in early diagnosis of cerebral<br />

infarction. Indian Journal of Radiology and Imaging ;13(2) : 213-217.<br />

Rovira A., Orellana P., Sabý´n J., Arenillas J.F., Aymerich<br />

X., Grive E., Molina C.,and Rovira-Gols A. (2004): Hyperacute


- 225 -<br />

References<br />

Ischemic Stroke:Middle Cerebral Artery Susceptibility Sign at Echo-<br />

planar Gradient-Echo MR Imaging. Radiology; 232:466–473.<br />

Rowley H.A. (2001): The Four Ps of Acute Stroke Imaging:<br />

Parenchyma, Pipes, Perfusion, and Penumbra. AJNR; 22:599-601.<br />

Ryan S., McNicholas M., Eustace S. (2004):Anatomy for<br />

Diagnostic Imaging, 2 nd edition, Saunders, Edinburgh, London, New<br />

York, Oxford, Philadelphia, St Louis, Sydney, Toronto; chapter 2 Pp<br />

46-84.<br />

Salvolini U., Scarabino T. (2006) : High Field Brain MRI,<br />

Use in Clinical Practice, Springer-Verlag , Berlin, Heidelberg, New<br />

York, chapter 14 Pp 177-185.<br />

Sanghvi D.A., Anand S. and Limaye U. (2007): Hyperacute<br />

Stroke Imaging : How much is enough?. Indian Journal of Radiology<br />

and Imaging ;17(4): 237-241.<br />

Saunders D.E. (2000) : MR spectroscopy in stroke. British<br />

Medical Bulletin ; 56 (No 2) 334-345.<br />

Saur D., Kucinski T., Grzyska U., Eckert B., Eggers C.,<br />

Niesen W., Schoder V., Zeumer H., Weiller C., and Rother<br />

J.(2003): Sensitivity and Interrater Agreement of CT and Diffusion-<br />

Weighted MR Imaging in Hyperacute Stroke. AJNR ;24:878–885.<br />

Schaefer P.W., Grant P.E. and Gonzalez G. (2000): State of<br />

the Art: Diffusion-Weighted MR Imaging of the Brain. Radiology;<br />

217:331-345.<br />

Schaller B. (2007): State Of The Art, Imaging in Stroke, Nova<br />

Science publishers, Inc, New York; vol 1, chapter 4 Pp 89-109.


- 226 -<br />

References<br />

Schellinger P.D., Jansen O., Fiebach J.B., Hacke W., and<br />

Sartor K.(1999) : A Standardized MRI Stroke Protocol Comparison<br />

with CT in Hyperacute Intracerebral Hemorrhage. Stroke;30:765-768.<br />

Schramm P., Schellinger P.D., Fiebach J.B., Heiland S.,<br />

Jansen O., Knauth M., Hacke W. ,and Sartor K. (2002) :<br />

Comparison of CT and CT Angiography Source Images With<br />

Diffusion-Weighted Imaging in Patients With Acute Stroke Within 6<br />

Hours After Onset. Stroke; 33:2426-2432.<br />

Schramm P., Schellinger P.D., Klotz E., Kallenberg K.,<br />

Fiebach J.B., Kulkens S., Heiland S., Knauth M., and Sartor K.<br />

(2004) : Comparison of Perfusion Computed Tomography and<br />

Computed Tomography Angiography Source Images With Perfusion-<br />

Weighted Imaging and Diffusion-Weighted Imaging in Patients With<br />

Acute Stroke of Less Than 6 Hours’ Duration. Stroke; 35:1652-1658.<br />

Seitz R.J., Meisel S., Weller P., Junghans U, Wittsack<br />

H.J.and Siebler M (2005) : Initial Ischemic Events: Perfusion-<br />

Weighted MR Imaging and Apparent Diffusion Coefficient for Stroke<br />

Evolution. Radiology; 237: 1020-1028.<br />

Smith W.S., Roberts H.C., Chuang N.A., Ong K.C., Lee<br />

T.J., Johnston S.C. and Dillon W.P.(2003): Safety and Feasibility of<br />

a CT Protocol for Acute Stroke: Combined CT, CT Angiography and<br />

CT Perfusion Imaging in 53 Consecutive Patients. AJNR; 24:688-690.<br />

Smoller S.W., Anderson G., Psaty B.M., Black H.R.,<br />

Manson J., Wong N., Francis J., Grimm R., Kotchen T., Langer<br />

R.,and Lasser N.(2000): Hypertension and Its Treatment in<br />

Postmenopausal Women Baseline Data from the Women’s Health<br />

Initiative. Hypertension; 36:780-789.


- 227 -<br />

References<br />

Snell R. S. (2007): Clinical Anatomy By Systems, Lippincott<br />

Williams& Wilkins, Philadelphia, Baltimore; Pp 687-689.<br />

Snell R. S. (2008): Clinical Anatomy By Regions, 8 th edition,<br />

Lippincott Williams& Wilkins, Philadelphia, Baltimore ;Pp 687-689.<br />

Somay G., Topaloglu, Somay H., Araal O., Halac G.U. and<br />

Bulkan M.(2006): Cerebrovascular Risk Factors and Stroke Subtypes<br />

in Different Age Groups: A Hospital-Based Study. Turk J Med Sci; 36<br />

:23-29.<br />

Somford D.M., Marks M.P., Thijs V.N. and Tong D.C.<br />

(2004): Association with early CT abnormalities, Infarct Size, and<br />

Apparent Diffusion Coefficient Reduction in Acute Ischemic Stroke.<br />

AJNR;25:933-938.<br />

Sorensen A.G. and Reimer P. (2000): Cerebral MR Perfusion<br />

Imaging: Principles and Current Applications, Georg Thieme Verlag,<br />

Stuttgart, New York ; 3-61.<br />

Srinivasan A, Goyal M, Al Azri F and Lum C.( 2006): State<br />

of Art Imaging of Acute Stroke. RadioGraphics; 26: 75-96.<br />

Sunshine J.L., Bambakidis N., Tarr R.W., Lanzieri C.F.,<br />

Zaidat O.O., Suarez J.I., Landis D.M.D., and Selman W.R. (2001):<br />

Benefits of Perfusion MR Imaging Relative to Diffusion MR Imaging<br />

in the Diagnosis and Treatment of Hyperacute Stroke.AJNR; 22:915-<br />

921.<br />

Sutton D. (2003) : Textbook Of Radiology And Imaging, 7th<br />

edition, Churchill Livingstone, vol II, chapter 6 Pp 1489-1846.<br />

Tank P. W. (1999): Grant's Dissector, fourteenth edition,<br />

Lippincott Williams& Wilkins, Philadelphia, Baltimore; Pp 220&<br />

225.


- 228 -<br />

References<br />

Torbey M.T. and Selim M.H. (2007): The Stroke Book,<br />

Cambridge University Press, New York, Melbourne, Madrid, Cape<br />

Town, Singapore, Sao Paulo; chapter 4 Pp 49-65.<br />

Uflacker R. (2007): Atlas of vascular Anatomy: An<br />

Angiographic Approach , 2 nd edition, Lippincott Williams& Wilkins,<br />

Philadelphia, Baltimore, New York, London, Buenos Aires, Hong<br />

Kong, Sydney, Tokyo; chapter 2 Pp 12-23.<br />

Weir J. and Abrahams P.H. (2000): Imaging atlas of human<br />

anatomy. 2 nd edition. Mosby-Wolf, London:163,181 and 182.<br />

Weissleder R., Wittenberg J., Harisinghani M.G. (2003):<br />

Primer of Diagnostic Imaging,3rd edition, Mosby, Philadelphia,<br />

Pennsylvania, chapter 6 Pp 485-588.<br />

Wessels T, Wessels C, Ellsiepen A, Reuter I, Trittmacher S,<br />

Stolz E and Jauss M. (2005): Contribution of Diffusion-Weighted<br />

Imaging in Determination of Stroke Etiology. AJNR ;27 :35-39.<br />

Westbrook C. (1999): Handbook of MRI Technique, 2 nd<br />

edition, Blackwell Science, Oxford, London, Edinburg, Malden, Paris:<br />

47-132.<br />

Wilcock D.J., Jones D.K., Horsfield M.A. and Cherryman<br />

G.R.(1999): Echoplanar MRI in patients with acute stroke syndrome.<br />

The British Journal of Radiology; 72:914-922.<br />

Wittsack HJ, Ritzl A, Fink GR, Wenserski F, Seitz RJ,<br />

Modder U and Freund HJ. (2002): MR imaging in Acute Stroke:<br />

Diffusion-Weighted and Perfusion Imaging Parameters for Predicting<br />

Infarct Size. Radiology;222:397-403.<br />

Wityk R.J. and Llinas R.H.(2006): Stroke, ACP Press; 5-40.


- 229 -<br />

References<br />

Wu O., Koroshetz W.J., Østergaard L., Buonanno F.S.,<br />

Copen W.A., Gonzalez R.G., Rordorf G., Rosen B.R., Schwamm<br />

L.H., Weisskoff R.M.,and Sorensen A.G.(2001) : Predicting Tissue<br />

Outcome in Acute Human Cerebral Ischemia Using Combined<br />

Diffusion- and Perfusion-Weighted MR Imaging. Stroke; 32:933-942.<br />

Yang J.J., Hill M.D., Morrish W.F., Hudon M.E., Barber<br />

P.A., Demchuk A.M., Sevick R.J., Frayne R.(2001): Post-contrast<br />

3D TOF MRA: Possible Role in Acute Stroke?. Proc. Intl. Soc. Mag.<br />

Reson. Med 9:1390.<br />

Yang Q., Tress B.M., Barber P.A., Desmond P.M., Darby<br />

D.G., Gerraty R.P., Li T.,and Davis S.M.(1999) : Serial Study of<br />

Apparent Diffusion Coefficient and Anisotropy in Patients With Acute<br />

Stroke. Stroke; 30:2382-2390.


ﺔﻗﺎﻋﻹا<br />

ﻰﻟإ<br />

- 1 -<br />

ﻲﺑﺮﻌﻟا ﺺﺨﻠﻤﻟا<br />

يدﺆﺗ ﻰﺘﻟا بﺎﺒﺳﻷا ﻢهأ ﻦﻣ ﺮﺒﺘﻌﺗ ﺎﻤآ تﺎﻴﻓﻮﻟا بﺎﺒﺳأ ﻢهأ ﻦﻣ ﺔﻴﻏﺎﻣﺪﻟا ﺔﺘﻜﺴﻟا ﺮﺒﺘﻌﺗ<br />

راﺮﻘﻟا ﺬﺧأ ﻦﻣ ﻦﻜﻤﺘﻠﻟ تﻻﺎﺤﻟا ﻩﺬﻬﻟ<br />

ﺮﻜﺒﻤﻟا ﺺﻴﺨﺸﺘﻟا ةروﺮﺿ ﺐﺟﻮﺘﺴﻳ ﺎﻤﻣ ﺮﻀﺤﺘﻤﻟا ﻢﻟﺎﻌﻟا ﻲﻓ<br />

ﺦﻤﻟا ﻦﻣ ءﺰﺟ ﻰﻟا ﺔﻳﻮﻣﺪﻟا ةروﺪﻟا ﻲﻓ رﻮﺼﻗ<br />

ﻦﻋ ﺔﺠﺗﺎﻧ تﻻﺎﺣ<br />

ﻰﻟإ<br />

. جﻼﻌﻟا ﺔﻘﻳﺮﻃ ﻦﻋ ﺢﻴﺤﺼﻟا<br />

ﺔﻴﻏﺎﻣﺪﻟا ﺔﺘﻜﺴﻟا تﻻﺎﺣ ﻢﺴﻘﻨﺗ<br />

. ﺦﻤﻟا ﻞﺧاد ﻒﻳﺰﻨﻟا ﻦﻋ ﺔﺠﺗﺎﻧ تﻻﺎﺣ وأ<br />

و ﺎﻬﺘﻟﻮﻬﺴﻟ رﺎﺸﺘﻧﻻا ﻊﺳاو ﺔﻴﻏﺎﻣﺪﻟا ﺔﺘﻜﺴﻟا ﺺﻴﺨﺸﺗ ﻲﻓ ﺔﻐﺒﺻ نوﺪﺑ ﺔﻴﻌﻄﻘﻤﻟا ﺔﻌﺷﻷا ماﺪﺨﺘﺳا نإ<br />

رﻮﺼﻗ ﻦﻋ ﺔﺠﺗﺎﻨﻟا تﻻﺎﺤﻟا ﺾﻌﺑ ﻰﻟإ<br />

ﺔﻓﺎﺿﻻﺎﺑ ﻒﻳﺰﻨﻟا تﻻﺎﺣ ﺺﻴﺨﺸﺗ ﻦﻣ ﺎﻨﻜﻤﺗ ﺚﻴﺣ ﺎﻬﺘﻋﺮﺳ<br />

مﺪﻋو<br />

تﻻﺎﺤﻟا ﻢﻈﻌﻤﻟ ﺮﻜﺒﻤﻟا ﺺﻴﺨﺸﺘﻟا ﻲﻓ ﺎﻬﺘﻴﺳﺎﺴﺣ مﺪﻋ ﺎﻬﺑﻮﻴﻋ<br />

ﻦﻣ ﻦﻜﻟ و ﺔﻳﻮﻣﺪﻟا<br />

ةروﺪﻟا ﻲﻓ<br />

. ﺔﺑﺎﺻﻹا<br />

ﻢﺠﺣ ﺪﻳﺪﺤﺗ ﻲﻓ ﺔﻗﺪﻟا مﺪﻋ و ةﺮﻴﻐﺼﻟا تﺎﺑﺎﺻﻹا<br />

ﺾﻌﺑ رﺎﻬﻇإ<br />

ﻰﻠﻋ ةرﺪﻘﻟا<br />

ﺔﺘﻜﺴﻟا تﻻﺎﺣ ﺺﻴﺨﺸﺗ ﻲﻓ ﺔﻴﻌﻄﻘﻤﻟا ﺔﻌﺷﻷا ﻦﻣ ﺔﻗد ﺮﺜآأ يﺪﻴﻠﻘﺘﻟا ﻲﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟا ﺮﺒﺘﻌﻳ<br />

ﺺﻴﺨﺸﺗ ﻰﻠﻋ ةرﺪﻘﻟا مﺪﻋ ﺎﻤﻬﺑﻮﻴﻋ ﻦﻣ ﻦﻜﻟ و ﺔﻳﻮﻣﺪﻟا<br />

رود ءﺎﺟ ﺎﻨه ﻦﻣ و ﺔﺑﺎﺻﻹا<br />

ﻦﻣ ﻰﻟوﻷا تﺎﻋﺎﺳ ﺖﺴﻟا ﻰﻟإ<br />

ةروﺪﻟا ﻲﻓ رﻮﺼﻗ ﻦﻋ ﺔﺠﺗﺎﻨﻟا ﺔﻴﻏﺎﻣﺪﻟا<br />

ثﻼﺜﻟا لﻼﺧ تﻻﺎﺤﻟا ﻦﻣ ﺮﻴﺜﻜﻟا<br />

ﺎﻬﺗرﺪﻘﻟ ﺔﻴﻏﺎﻣﺪﻟا ﺔﺘﻜﺴﻟا<br />

تﻻﺎﺣ ﺺﻴﺨﺸﺗ ﻰﻓ ﻰﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟا ماﺪﺨﺘﺳﺎﺑ ﺔﻳوﺮﺘﻟا و<br />

ﻲﻠﻋ ﺎﻬﺗرﺪﻗ ﻲﻟا ﻪﻓﺎﺿﻻﺎﺑ ﻪﺑﺎﺻﻹا<br />

ﻦﻣ ﻰﻟوﻷا<br />

تﺎﻋﺎﺳ ﺖﺴﻟا لﻼﺧ تﻻﺎﺤﻟا<br />

رﺎﺸﺘﻧﻻا<br />

ﻢﻈﻌﻣ ﺺﻴﺨﺸﺛ ﻰﻠﻋ<br />

. ﺔﻨﻣﺰﻤﻟاو<br />

ﻩدﺎﺤﻟا ﺔﻴﻏﺎﻣﺪﻟا ﺔﺘﻜﺴﻟا تﻻﺎﺣ ﻦﻴﺑ ﺔﻗﺮﻔﺘﻟاو ﻪﻘﻴﻗﺪﻟا ثﺎﺑﺎﺻﻹا<br />

ﺺﻴﺨﺸﺗ<br />

ﻰﻓ ﻪﻣاﺪﺨﺘﺳا ﻦﻜﻤﻳ ﻚﻟﺬﺑو ﺦﻤﻟا ﺎﻳﻼﺧ لﻼﺧ ءﺎﻤﻟا تﺎﺌﻳﺰﺟ رﺎﺸﺘﻧا ﻪﻧﺄﺑ رﺎﺸﺘﻧﻻا ﻒﻳﺮﻌﺗ ﻦﻜﻤﻳ<br />

ﻲﻓ رﻮﺼﻗ ﻦﻋ ﺔﺠﺗﺎﻨﻟا تﻻﺎﺤﻟا<br />

ﻲﻓ<br />

رﺎﺸﺘﻧﻻا ﻞﻣﺎﻌﻣ ﻞﻘﻳ ﺚﻴﺣ ﺦﻤﻟا ﻲﻓ ﺔﺘﻴﻤﻟا ﻪﺠﺴﻧﻷا ﺪﻳﺪﺤﺗ<br />

ﻢﺛ ﻦﻣو ﺔﺑﺎﺼﻤﻟا ﺎﻳﻼﺨﻟا ﻞﺧاد ءﺎﻤﻟا تﺎﺌﻳﺰﺟ ﺔﺒﺴﻧ دادﺰﺗ ﺚﻴﺣ ﻰﻌﻴﺒﻄﻟا ﻪﻟﺪﻌﻣ ﻦﻋ<br />

ﺔﻘﻳﺮﻄﺑ<br />

ﺎﻬﻓﺎﺸﺘآا ﻦﻜﻤﻳ اﺪﺟ<br />

و ﺔﻐﺒﺻ يأ ﻦﻘﺣ جﺎﺘﺤﺗ<br />

ﻻ<br />

ةﺮﻜﺒﻤﻟا تاﺮﻴﻐﺘﻟا ﻩﺬه<br />

ﺎﻬﻧأ<br />

ﻲﺑﺮﻌﻟا ﺺﺨﻠﻤﻟا<br />

ﺔﻘﻳﺮﻄﻟا ﻩﺬه<br />

ﺔﻴﺴﻴﻃﺎﻨﻐﻣ صاﻮﺧ ﺎﻬﻟ ةدﺎﻣ ﻊﺒﺘﺘﺑ ﻪﺑﺎﺴﺣ ﻢﺘﻳو ﺦﻤﻟا ﺎﻳﻼﺧ ﻰﻟإ<br />

ﺔﻳﻮﻣﺪﻟا ةروﺪﻟا<br />

. ﺎﻳﻼﺨﻟا دوﺪﺣ ﻞﺧاد ﺎهرﺎﺸﺘﻧا ﺪﻳﺪﺤﺗ ﻢﺘﻳ<br />

زﺎﺘﻤﺗو<br />

ﻰﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟا ماﺪﺨﺘﺳﺎﺑ رﺎﺸﺘﻧﻻا<br />

. ﺮﻴﺼﻗ ﺖﻗو قﺮﻐﺘﺴﺗ<br />

مﺪﻟا لﻮﺻو ﺎﻬﺑ ﺪﺼﻘﻴﻓ ﺔﻳوﺮﺘﻟا ﺎﻣأ<br />

ﻲﻧﺎﻌﻳ فﻮﺳ ﺔﻳﻮﻣﺪﻟا ةروﺪﻟا ﻲﻓ رﻮﺼﻗ ﻦﻣ ﻲﻧﺎﻌﻳ يﺬﻟا ءﺰﺠﻟا . ﺦﻤﻠﻟ ﺎﻬﻟﻮﺻو ءﺎﻨﺛأ<br />

. ﻪﻴﻟإ<br />

ﺔﻐﺒﺼﻟا ﻞﺜﻣ<br />

ﺔﻐﺒﺼﻟا لﻮﺻو ﺮﺧﺄﺗ وأ مﺪﻋ ﻦﻣ


- 2 -<br />

ﻲﺑﺮﻌﻟا ﺺﺨﻠﻤﻟا<br />

ﻢﺋاد ﻒﻠﺘﺑ ﺐﻴﺻأ يﺬﻟا ﺦﻤﻟا ﻦﻣ ءﺰﺠﻟا ﺔﻓﺮﻌﻣ ﻦﻜﻤﻳ ﺔﻳوﺮﺘﻟا رﻮﺻ ﻊﻣ رﺎﺸﺘﻧﻻا رﻮﺻ ﺔﻧرﺎﻘﻣ ﺪﻨﻋ<br />

ﺔﻠﺑﺎﻗ ﺖﻟاز ﺎﻣو ﻰﻧﺎﻳﺮﺸﻟا داﺪﺴﻧﻹﺎﺑ<br />

تﺮﺛﺄﺗ ﻰﺘﻟا ﺔﺠﺴﻧﻷا ﺪﻳﺪﺤﺗ ﻦﻜﻤﻳ ﻚﻟﺬآو<br />

جﻼﻌﻠﻟ ﻞﺑﺎﻗ ﺮﻴﻏ و<br />

. جﻼﻌﻠﻟ<br />

ﺔﺘﻜﺴﻟا ﺐﺒﺳ ﺺﻴﺨﺸﺗ ﻲﻓ ﺔﻴﻤهأ وذ ﻰﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟا ﺔﻄﺳاﻮﺑ ﺦﻤﻟا ﻦﻴﻳاﺮﺷ ﺮﻳﻮﺼﺗ ﺮﺒﺘﻌﻳ<br />

ﻲﻓ رﻮﺼﻗ ﻦﻋ ﺔﺠﺗﺎﻨﻟا تﻻﺎﺤﻟا ﻲﻓ ﻲﻧﺎﻳﺮﺸﻟا داﺪﺴﻧﻻا تﻻﺎﺣ ﻞﺜﻣ ﺔﻴﻠﺧﺪﺗ ﺮﻴﻏ ﺔﻘﺑﺮﻄﺑ ﺔﻴﻏﺎﻣﺪﻟا<br />

ﺔﺠﺴﻧأ ﻞﺧاد ﺔﻴﻀﻳﻷا داﻮﻤﻟا<br />

. ﻲﻧﺎﻳﺮﺸﻟا دﺪﻤﺘﻟا رﺎﺠﻔﻧا ﻦﻣ ﺞﺗﺎﻨﻟا ﻒﻳﺰﻨﻟا تﻻﺎﺣ و ﺔﻳﻮﻣﺪﻟا ةروﺪﻟا<br />

ﺔﻴﻤآ سﺎﻴﻗ ﻰﻠﻋ ﺪﻤﺘﻌﻴﻓ ﻲﻔﻴﻄﻟا ﻲﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟﺎﺑ ﺮﻳﻮﺼﺘﻟا<br />

ةروﺪﻟا ﻲﻓ رﻮﺼﻗ ﻦﻋ ﺔﺠﺗﺎﻨﻟا ﺔﻴﻏﺎﻣﺪﻟا ﺔﺘﻜﺴﻟا تﻻﺎﺤﻟ ﺮﻜﺒﻤﻟا ﺺﻴﺨﺸﺘﻟا<br />

ﻦﻣ ﻦﻜﻤﺘﻳ<br />

ﻚﻟﺬﺑ و ﺦﻤﻟا<br />

. ﻦﻴﺘﻋﻮﻤﺠﻣ<br />

ﻰﻟإ<br />

. ﺔﺑﺎﺼﻤﻟا ﺔﺠﺴﻧﻷا ﻲﻓ ﺔﻴﻀﻳﻷا داﻮﻤﻟا ﺔﻴﻤآ سﺎﻴﻗ<br />

ﻢﻬﻤﻴﺴﻘﺗ ﻢﺗ ﺔﻴﻏﺎﻣﺪﻟا ﺔﺘﻜﺴﻟا<br />

ﻦﻣ نﻮﻧﺎﻌﻳ ﺎﻀﻳﺮﻣ نﻮﺴﻤﺧ<br />

ﻲﻓ رﻮﺼﻗ ﻦﻋ ﺔﺠﺗﺎﻨﻟا ﺔﻴﻏﺎﻣﺪﻟا<br />

ﺔﺘﻜﺴﻟا ﻦﻣ نﻮﻧﺎﻌﻳ ﺎﻀﻳﺮﻣ<br />

ﺔﺠﺗﺎﻨﻟا ﺔﻴﻏﺎﻣﺪﻟا ﺔﺘﻜﺴﻟا ﻦﻣ نﻮﻧﺎﻌﻳ ﺎﻀﻳﺮﻣ<br />

11<br />

39<br />

ﺎﻣأ<br />

ﻖﻳﺮﻃ ﻦﻋ ﺔﻳﻮﻣﺪﻟا<br />

ﺔﻟﺎﺳﺮﻟا ﻩﺬه ﺖﻨﻤﻀﺗ<br />

ﻦﻣ نﻮﻜﺘﺗ ﻰﻟوﻷا ﺔﻋﻮﻤﺠﻤﻟا<br />

ﻦﻣ نﻮﻜﺘﺗ ﺔﻴﻧﺎﺜﻟا ﺔﻋﻮﻤﺠﻤﻟا ﺎﻣأ ﺔﻳﻮﻣﺪﻟا ةروﺪﻟا<br />

ﻦﻴﻧﺮﻟاو ﺔﻴﻌﻄﻘﻤﻟا ﺔﻌﺷﻷا ﺔﻴﺳﺎﺴﺣ ﺮﺜآأ ﻰﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟا ماﺪﺨﺘﺳﺎﺑ رﺎﺸﺘﻧﻻا<br />

.( ةﺮﻜﺒﻤﻟا ةدﺎﺤﻟا و ةدﺎﺤﻟا تﻻﺎﺤﻟا)<br />

تﻻﺎﺤﻠﻟ<br />

ﺮﻜﺒﻤﻟا ﺺﻴﺨﺸﺘﻟا<br />

ﻰﻌﻴﺒﻄﻟا ﻪﻟﺪﻌﻣ ﻦﻋ ﻪﻧﺎﺼﻘﻧ ﻢﺗ ﺚﻴﺣ ﺔﻄﻠﺠﻟا<br />

. ﺦﻤﻟﺎﺑ ﻒﻳﺰﻨﻟا ﻦﻋ<br />

نأ ﺎﻧﺪﺟو ﺪﻗو<br />

ﻲﻓ يﺪﻴﻠﻘﺘﻟا ﻰﺴﻴﻃﺎﻨﻐﻤﻟا<br />

ﻞﺣاﺮﻣ فﻼﺘﺧﺎﺑ رﺎﺸﺘﻧﻻا ﻞﻣﺎﻌﻣ ﺮﻴﻐﺗ ﺎﻨﻈﺣﻻ<br />

ﺪﻘﻟو<br />

. ﺔﻨﻣﺰﻤﻟا تﻻﺎﺤﻟا ﻲﻓ ﻪﺗدﺎﻳز ﻢﺗو ةﺮﺧﺄﺘﻤﻟا ةدﺎﺤﻟا و ةدﺎﺤﻟاو ةﺮﻜﺒﻤﻟا ةدﺎﺤﻟا تﻻﺎﺤﻟا ﻲﻓ<br />

ماﺪﺨﺘﺳﺎﺑ ﺔﻳوﺮﺘﻟا ﺺﺤﻔﻟ<br />

7<br />

ﻲﻓ<br />

( ﺾﻳﺮﻣ<br />

: ﻲﺗﻻا ﺪﺟوو<br />

17)<br />

ةﺮﻜﺒﻤﻟا ةدﺎﺤﻟا و ةدﺎﺤﻟا تﻻﺎﺤﻟا<br />

ﻢﻈﻌﻣ ﺖﻌﻀﺧ ﺪﻗو<br />

ﺔﻳوﺮﺘﻟا رﻮﺻ ﻊﻣ رﺎﺸﺘﻧﻻا رﻮﺻ ﺔﻧرﺎﻘﻣ ﻢﺗو ﻰﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟا<br />

رﺎﺸﺘﻧﻻا رﻮﺻ ﻲﻓ ﺎﻬﻤﺠﺣ ﻦﻣ ﺮﺒآأ ﺔﻳوﺮﺘﻟا رﻮﺻ ﻲﻓ ﺔﺑﺎﺻﻹا<br />

ﻢﺠﺣ ﺖﻧﺎآ<br />

ﺔﺠﺴﻧأ دﻮﺟو اﺬه ﻲﻨﻌﻳو ةﺮﻴﺒﻜﻟا ﻦﻴﻳاﺮﺸﻟا داﺪﺴﻧا ﺔﺠﻴﺘﻧ<br />

6<br />

ﺔﺑﺎﺻﻹا<br />

ﻚﻠﺗ ﺖﻧﺎآو تﻻﺎﺣ<br />

. جﻼﻌﻠﻟ ﺔﻠﺑﺎﻗ ﺖﻟاز ﺎﻣو ﻰﻧﺎﻳﺮﺸﻟا داﺪﺴﻧﻹﺎﺑ<br />

تﺮﺛﺄﺗ<br />

ﻲﻓ رﺎﺸﺘﻧﻻا رﻮﺻ ﻲﻓ ﺎﻬﻤﺠﺤﻟ ﺔﻠﺛﺎﻤﻣ ﺔﻳوﺮﺘﻟا رﻮﺻ ﻲﻓ<br />

ﺔﺑﺎﺻﻹا<br />

ﻢﺠﺣ ﺖﻧﺎآ<br />

دﻮﺟو مﺪﻋ اﺬه ﻲﻨﻌﻳو ةﺮﻴﻐﺼﻟا<br />

ﻦﻴﻳاﺮﺸﻟا داﺪﺴﻧا ﺔﺠﻴﺘﻧ ﺔﺑﺎﺻﻻا ﻚﻠﺗ ﺖﻧﺎآو تﻻﺎﺣ<br />

ﻲﻓ رﺎﺸﺘﻧﻻا<br />

رﻮﺻ ﻲﻓ ﺎﻬﻤﺠﺣ ﻦﻣ ﺮﻐﺻأ ﺔﻳوﺮﺘﻟا رﻮﺻ ﻲﻓ<br />

. جﻼﻌﻠﻟ ﺔﻠﺑﺎﻗ ﺔﺠﺴﻧأ<br />

ﺔﺑﺎﺻﻹا<br />

ﻢﺠﺣ ﺖﻧﺎآ<br />

رﻮﺻ ﻲﻓ ةدﻮﺟﻮﻣ ﺎﻬﻨﻜﻟو ﺔﻳوﺮﺘﻟا رﻮﺻ ﻲﻓ ةدﻮﺟﻮﻣ ﺮﻴﻏ ﺔﺑﺎﺻﻻا ﺖﻧﺎآ و ﻦﻴﺘﻟﺎﺣ


ةﺮﻴﻐﺼﻟا<br />

ﻦﻴﻳاﺮﺸﻟا داﺪﺴﻧا ﺔﺠﻴﺘﻧ تﺎﺑﺎﺻﻹا<br />

ﻚﻠﺗ ﺖﻧﺎآ ةﺪﺣاو ﺔﻟﺎﺣ ﻲﻓ رﺎﺸﺘﻧﻻا<br />

ﻦﻴﻧﺮﻟا ﺔﻄﺳاﻮﺑ ﺦﻤﻟا ﻦﻴﻳاﺮﺷ ﺮﻳﻮﺼﺘﻟ ( ﺔﻟﺎﺣ 37)<br />

ﻲﻓ ةدﻮﺟﻮﻣ ﻲﻧﺎﻳﺮﺸﻟا داﺪﺴﻧﻻا تﻻﺎﺣ ﻢﻈﻌﻣ نأ ﺎﻧﺪﺟوو<br />

- 3 -<br />

. ﺔﻳوﺮﺘﻟا ةدﺎﻋإ<br />

ثوﺪﺣو<br />

ﻲﺑﺮﻌﻟا ﺺﺨﻠﻤﻟا<br />

ﻰﻟوﻷا ﺔﻋﻮﻤﺠﻤﻟا تﻻﺎﺣ ﻢﻈﻌﻣ ﺖﻌﻀﺧ ﺪﻗو<br />

ﻦﻴﻳاﺮﺸﻟا داﺪﺴﻧا نﺎﻴﺒﻟ ﻰﺴﻴﻃﺎﻨﻐﻤﻟا<br />

ﺮﺜآأ ةﺮﻴﺒﻜﻟا ﻦﻴﻳاﺮﺸﻟا تﻻﺎﺣ ﻲﻓو ﺔﻨﻣﺰﻤﻟا تﻻﺎﺤﻟا ﻲﻓ ﺎﻬﻨﻣ ﺮﺜآأ ةدﺎﺤﻟاو ةﺮﻜﺒﻤﻟا ةدﺎﺤﻟا تﻻﺎﺤﻟا<br />

و ﻲﻔﻴﻄﻟا ﻲﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟﺎﺑ ﺮﻳﻮﺼﺘﻟا ( ﺔﻟﺎﺣ 34)<br />

و ةﺮﻜﺒﻤﻟا ةدﺎﺤﻟاو ةدﺎﺤﻟا تﻻﺎﺤﻟا ﻞآ<br />

ﻲﻓ ﺔﺑﺎﺻﻻا نﺎﻜﻣ ﻲﻓ<br />

. ةﺮﻴﻐﺼﻟا<br />

ﻦﻴﻳاﺮﺸﻟا تﻻﺎﺣ ﻲﻓ ﺎﻬﻨﻣ<br />

ﻰﻟوﻷا ﺔﻋﻮﻤﺠﻤﻟا تﻻﺎﺣ ﻢﻈﻌﻣ ﺖﻌﻀﺧ و<br />

ﻚﻴﺘآﻼﻟا ﺾﻤﺣ دﻮﺟو<br />

. ﻪﻴﻠﻋ يﻮﺘﺤﺗ ﻻ ﻰﺘﻟا ﺦﻤﻟا ﻦﻣ ﺔﻤﻴﻠﺴﻟا ءاﺰﺟﻷﺎﺑ ﺔﻧرﺎﻘﻣ ﺔﻨﻣﺰﻤﻟاو<br />

ةﺮﺧﺄﺘﻤﻟا<br />

ﺎﻨﻈﺣﻻ ﺪﻗ<br />

ةدﺎﺤﻟا تﻻﺎﺤﻟا ﻢﻈﻌﻣ<br />

ﻦﻴﺗﺎﻳﺮﻜﻟا ﺺﻘﻧ ﺎﻀﻳأ ﺎﻨﻈﺣﻻ و تﻻﺎﺤﻟا<br />

ﻞآ ﻲﻓ ﻪﻧﺎﺼﻘﻧ ﺎﻨﻈﺣﻻ ﺪﻘﻓ ﻚﻴﺗﺮﺒﺳﻷا ﺾﻤﺤﻟ ﺔﺒﺴﻨﻟﺎﺑ ﺎﻣأ<br />

ءاﺰﺟﻷﺎﺑ ﺔﻧرﺎﻘﻣ<br />

ﺔﺑﺎﺻﻹا<br />

نﺎﻜﻣ ﻲﻓ تﻻﺎﺤﻟا ﻦﻴﺑ ﻦﻴﻟﻮﻜﻟا ﺔﻴﻤآ ﻲﻓ فﻼﺘﺧﻻاو تﻻﺎﺤﻟا ﻢﻈﻌﻣ ﻲﻓ<br />

. ﺔﻤﻴﻠﺴﻟا<br />

ﺎﻀﻳﺮﻣ 11 ﻦﻣ نﻮﻜﺘﺗو ﺦﻤﻟﺎﺑ ﻒﻳﺰﻨﻟا ﻦﻋ ﺔﺠﺗﺎﻨﻟا ﺔﻴﻏﺎﻣﺪﻟا ﺔﺘﻜﺴﻟا : ﺔﻴﻧﺎﺜﻟا ﺔﻋﻮﻤﺠﻤﻠﻟ ﺔﺒﺴﻨﻟﺎﺑ ﺎﻣأ<br />

. تﺎﻋﻮﻤﺠﻣ<br />

ةﺪﻋ ﻰﻟإ<br />

ﻒﻳﺰﻨﻟا نﺎﻜﻣ ﺐﺴﺣ ﻢﻬﻤﻴﺴﻘﺗ ﻢﺗ<br />

ﻒﻳﺰﻨﻠﻟ داﺪﺘﻣا ﻪﺑ ﺪﺣاو ﺾﻳﺮﻣ ﻢﻬﻨﻣ ﺎﻀﻳﺮﻣ 8ﻰﻠﻋ<br />

يﻮﺘﺤﺗو ﺦﻤﻟا ﻞﺧاد ﻒﻳﺰﻨﻟا تﻻﺎﺣ<br />

. ﺦﻤﻟا ﻦﻴﻄﺑ ﻞﺧاد<br />

. ﺔﻴﺗﻮﺒﻜﻨﻌﻟا مﻷا ﺖﺤﺗ ﻒﻳﺰﻨﻟا ﻦﻣ نﻮﻧﺎﻌﻳ ﻦﻴﺘﻟﺎﺣ<br />

. ﺦﻤﻟا ﻦﻴﻄﺑ ﻞﺧاد ﻒﻳﺰﻨﻟا ﻦﻣ ﻲﻧﺎﻌﺗ ةﺪﺣاو ﺔﻟﺎﺣ<br />

ضﺮﻤﻟا ﺖﻴﻗﻮﺗ ﺐﺴﺣ ( ﻞﺣاﺮﻣ)<br />

عاﻮﻧأ ةﺪﻋ ﻰﻟإ<br />

ﻢﻬﻤﻴﺴﻘﺗ ﻢﺗ ﺪﻘﻓ ﺦﻤﻟا ﻞﺧاد ﻒﻳﺰﻨﻟا تﻻﺎﺤﻟ<br />

يﻮﺘﺤﺗو ( ﻒﻳﺰﻨﻟا ثوﺪﺣ ﺬﻨﻣ ﻰﻟوﻷا 24 لﻼﺧ)<br />

ﻰﻠﻋ يﻮﺘﺤﺗو<br />

( ﻒﻳﺰﻨﻟا<br />

ثوﺪﺣ ﺬﻨﻣ<br />

مﺎﻳأ<br />

3-2<br />

يﻮﺘﺤﺗو ( ﻒﻳﺰﻨﻟا<br />

ثوﺪﺣ ﺬﻨﻣ مﻮﻳ 14-3<br />

ﻦﻣ)<br />

ةﺮﻜﺒﻤﻟا ةدﺎﺤﻟا ﺦﻤﻟا ﻞﺧاد ﻒﻳﺰﻨﻟا تﻻﺎﺣ<br />

ﻦﻣ)<br />

. ﻦﻴﻀﻳﺮﻣ<br />

ﻰﻠﻋ<br />

ةدﺎﺤﻟا ﺦﻤﻟا ﻞﺧاد ﻒﻳﺰﻨﻟا تﻻﺎﺣ<br />

. ﻦﻴﻀﻳﺮﻣ<br />

ةﺮﺧﺄﺘﻤﻟا ةدﺎﺤﻟا ﺦﻤﻟا ﻞﺧاد ﻒﻳﺰﻨﻟا تﻻﺎﺣ<br />

.<br />

ﻰﺿﺮﻣ<br />

3 ﻰﻠﻋ<br />

<br />

<br />

<br />

ﺔﺒﺴﻨﻟﺎﺑ<br />

: ﻞﻤﺸﺗو


ﻰﻠﻋ يﻮﺘﺤﺗو<br />

( ﻒﻳﺰﻨﻟا<br />

ثوﺪﺣ ﺬﻨﻣ ﻦﻴﻋﻮﺒﺳا ﻦﻣ ﺮﺜآأ)<br />

- 4 -<br />

ﺔﻨﻣﺰﻤﻟا ﺦﻤﻟا ﻞﺧاد ﻒﻳﺰﻨﻟا<br />

. ﺪﺣاو ﺾﻳﺮﻣ<br />

ﻲﺑﺮﻌﻟا ﺺﺨﻠﻤﻟا<br />

ﺐﻴآﺮﺗو ﻒﻳﺰﻨﻟا ﺔﻠﺣﺮﻣ فﻼﺘﺧﺎﺑ يﺪﻴﻠﻘﺘﻟا ﻲﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟا رﻮﺻ ﻲﻓ ﻒﻳﺰﻨﻟا ﺮﻬﻈﻣ ﻒﻠﺘﺨﻳو<br />

ةﺮﻜﺒﻤﻟا ةدﺎﺤﻟا<br />

تﻻﺎﺤﻟا<br />

ةدﺎﺤﻟا تﻻﺎﺤﻟا ﺾﻌﺑو<br />

ﻲﻓ ﻞﻘﻳ<br />

ﺔﻨﻣﺰﻤﻟا تﻻﺎﺤﻟا<br />

ﻪﻧأ ﺎﻧﺪﺟو ﺚﻴﺣ رﺎﺸﺘﻧﻻا ﻞﻣﺎﻌﻣ ﺎﻀﻳأ ﻒﻠﺘﺨﻳو<br />

ﻲﻓ<br />

ﺪﻳﺰﻳو<br />

ﺎﻧﺪﺟوو ﻰﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟا ﺔﻄﺳاﻮﺑ ﺦﻤﻟا ﻦﻴﻳاﺮﺷ ﺮﻳﻮﺼﺘﻟ<br />

<br />

ﻦﻴﺑﻮﻠﺟﻮﻤﻴﻬﻟا<br />

ةﺮﺧﺄﺘﻤﻟا ةدﺎﺤﻟا تﻻﺎﺤﻟا ﺾﻌﺑو ةدﺎﺤﻟاو<br />

ﻒﻳﺰﻨﻟا<br />

. ةﺮﺧﺄﺘﻤﻟا<br />

تﻻﺎﺣ ﻞآ ﺖﻌﻀﺧ ﺪﻗو<br />

ﻦﻴﺘﻟﺎﺣ ﻲﻓ ﻦﻴﻳاﺮﺸﻟا<br />

ﻲﻓ ﻖﻴﺿو ﺦﻤﻟا ﻞﺧاد ﻒﻳﺰﻨﻟا تﻻﺎﺣ ﻦﻣ ةﺪﺣاو ﺔﻟﺎﺣ ﻲﻓ ﻦﻴﻳاﺮﺸﻟا ﻲﻓ داﺪﺴﻧا<br />

دﺪﻤﺗو<br />

ﺔﻴﺗﻮﺒﻜﻨﻌﻟا مﻷا ﺖﺤﺗ<br />

ﻒﻳﺰﻨﻟا تﻻﺎﺣ ﻦﻣ ةﺪﺣاو ﺔﻟﺎﺣ و<br />

. ﺔﻴﺗﻮﺒﻜﻨﻌﻟا مﻷا ﺖﺤﺗ<br />

ﺦﻤﻟا ﻞﺧاد ﻒﻳﺰﻨﻟا تﻻﺎﺣ ﻦﻣ<br />

ﻒﻳﺰﻨﻟا تﻻﺎﺣ ﻦﻣ ةﺪﺣاو ﺔﻟﺎﺣ ﻲﻓ ﻲﻧﺎﻳﺮﺷ<br />

: ﻲﺗﻻا ﺎﻨﺼﻠﺨﺘﺳا<br />

لﻼﺧ<br />

ﺔﻴﻏﺎﻣﺪﻟا ﺔﺘﻜﺴﻟا تﻻﺎﺣ ﺺﻴﺨﺸﺗ ﻲﻓ ﺮﻴﺒآ رود ﻪﻟ ﻰﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟا ماﺪﺨﺘﺳﺎﺑ رﺎﺸﺘﻧﻻا نإ<br />

ﺔﻗﺮﻔﺘﻟاو ﻪﻘﻴﻗﺪﻟا ثﺎﺑﺎﺻﻹا<br />

ﺺﻴﺨﺸﺗ ﻲﻠﻋ ﻪﺗرﺪﻗ ﻲﻟا ﻪﻓﺎﺿﻻﺎﺑ ﻪﺑﺎﺻﻹا<br />

ﻦﻣ ﻰﻟوﻷا تﺎﻋﺎﺳ ﺖﺴﻟا<br />

ﻦﻣ ﺎﻨﻜﻤﺘﻓ ﻰﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟا ماﺪﺨﺘﺳﺎﺑ ﺔﻳوﺮﺘﻟا ﺎﻣأ ﺔﻨﻣﺰﻤﻟاو ﻩدﺎﺤﻟا ﺔﻴﻏﺎﻣﺪﻟا ﺔﺘﻜﺴﻟا تﻻﺎﺣ ﻦﻴﺑ<br />

ﻦﻴﻳاﺮﺷ ﺮﻳﻮﺼﺗ<br />

مﺪﺨﺘﺴﻳو جﻼﻌﻠﻟ ﺔﻠﺑﺎﻗ ﺖﻟاز ﺎﻣو ﻰﻧﺎﻳﺮﺸﻟا داﺪﺴﻧﻹﺎﺑ<br />

تﺮﺛﺄﺗ ﻰﺘﻟا ﺔﺠﺴﻧﻷا ﺔﻓﺮﻌﻣ<br />

ﻲﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟﺎﺑ ﺮﻳﻮﺼﺘﻟا ﺎﻣأ<br />

ﺪﻗو<br />

ﻦﻴﻳاﺮﺸﻟا داﺪﺴﻧا نﺎﻴﺑ ﻲﻓ ﻰﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟا ﺔﻄﺳاﻮﺑ ﺦﻤﻟا<br />

ﻲﻓ رﻮﺼﻗ ﻦﻋ ﺔﺠﺗﺎﻨﻟا ﺔﻴﻏﺎﻣﺪﻟا ﺔﺘﻜﺴﻟا تﻻﺎﺣ ﻲﻓ ﺔﻴﻀﻳﻻا داﻮﻤﻟا ﻲﻓ تﺎﻓﻼﺘﺧﻻا ﺮﻬﻈﻴﻓ ﻲﻔﻴﻄﻟا<br />

. ﺔﻳﻮﻣﺪﻟا ةروﺪﻟا<br />

ﺔﻠﺣﺮﻣ فﻼﺘﺧﺎﺑ رﺎﺸﺘﻧﻻا ﻞﻣﺎﻌﻣو<br />

يﺪﻴﻠﻘﺘﻟا ﻲﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟا رﻮﺻ ﻲﻓ ﻒﻳﺰﻨﻟا ﺮﻬﻈﻣ ﻒﻠﺘﺨﻳو<br />

.<br />

ﻦﻴﺑﻮﻠﺟﻮﻤﻴﻬﻟا ﺐﻴآﺮﺗو ﻒﻳﺰﻨﻟا


- 5 -<br />

ﻲﺑﺮﻌﻟا ﺺﺨﻠﻤﻟا


ﻲﺴﻴﻃﺎﻨﻐﻤﻟا ﻦﻴﻧﺮﻟا ﻦﻣ ﺔﻔﻠﺘﺨﻣ<br />

ﻖﻳزﺎﻗﺰﻟا ﺔﻌﻣﺎﺟ<br />

ﺐﻄﻟا ﺔﻴﻠآ<br />

ﺔﻴﺼﻴﺨﺸﺘﻟا ﺔﻌﺷﻷا ﻢﺴﻗ<br />

تﺎﻴﻨﻘﺘﺑ<br />

ﺔﻴﻏﺎﻣﺪﻟا ﺔﺘﻜﺴﻟا ﺮﻳﻮﺼﺗ<br />

ﻦﻣ ﺔﻣﺪﻘﻣ ﺔﻟﺎﺳر<br />

ﺔﺒﻴﺒﻄﻟا<br />

يﺮﻳﺮﺤﻟا ﺪﻤﺤﻣ ﺪﻤﺤﻣ رﺎﻔﻐﻟا ﺪﺒﻋ ﻰﻨﻣ<br />

ﺔﻴﺼﻴﺨﺸﺘﻟا ﺔﻌﺷﻷا ﺪﻋﺎﺴﻣ سرﺪﻣ<br />

ﻖﻳزﺎﻗﺰﻟا ﺔﻌﻣﺎﺟ-ﺐﻄﻟا<br />

ﺔﻴﻠآ<br />

ﺔﻴﺼﻴﺨﺸﺘﻟا ﺔﻌﺷﻷا ﻲﻓ ﻩارﻮﺘآﺪﻟا ﺔﺟرد ﻰﻠﻋ لﻮﺼﺤﻠﻟ ﺔﺌﻃﻮﺗ<br />

يوﺎﻄﻨــﻃ<br />

فاﺮﺷإ ﺖﺤﺗ<br />

ﺪــﻤــﺣأ<br />

ﻲــﺤــﺘـــﻓ<br />

ﺔﻴﺼﻴﺨﺸﺘﻟا ﺔﻌﺷﻷا ذﺎﺘﺳأ<br />

ﺐﻄﻟا ﺔﻴﻠآ<br />

ﻖﻳزﺎﻗﺰﻟا ﺔﻌﻣﺎﺟ<br />

ﺔـﻔـﻴـﻠـﺧ<br />

ﻞـــﻴــﺒــﻧ<br />

ﺎـــﻴــــﻟاد<br />

ﺔﻴﺼﻴﺨﺸﺘﻟا ﺔﻌﺷﻷا ذﺎﺘﺳأ<br />

ﺐﻄﻟا ﺔﻴﻠآ<br />

ﻖﻳزﺎﻗﺰﻟا ﺔﻌﻣﺎﺟ<br />

سوراﺪﻴﻋ ﺪﻴﻤﺤﻟاﺪﺒﻋ يﺪﺠﻣ<br />

بﺎﺼﻋﻷا و ﺦﻤﻟا ضاﺮﻣأ ﺪﻋﺎﺴﻣ ذﺎﺘﺳأ<br />

ﺐﻄﻟا ﺔﻴﻠآ<br />

ﻖﻳزﺎﻗﺰﻟا ﺔﻌﻣﺎﺟ<br />

ﻖﻳزﺎﻗﺰﻟا ﺔﻌﻣﺎﺟ – ﺐﻄﻟا ﺔﻴﻠآ<br />

2009<br />

/<br />

/<br />

/<br />

د<br />

د.<br />

أ<br />

. أ<br />

د.<br />

أ

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

Saved successfully!

Ooh no, something went wrong!