10.07.2015 Views

FNPS Japan 2007 - Fetal and Neonatal Physiological Society

FNPS Japan 2007 - Fetal and Neonatal Physiological Society

FNPS Japan 2007 - Fetal and Neonatal Physiological Society

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.

35th Annual Meeting ofThe <strong>Fetal</strong> <strong>and</strong> <strong>Neonatal</strong> <strong>Physiological</strong> <strong>Society</strong> (<strong>FNPS</strong>)Hotel Van der Valk, Maastricht, the Netherl<strong>and</strong>s22 – 25 June 2008Maastricht, the Netherl<strong>and</strong>s, awaits you in 2008!During this <strong>FNPS</strong> meeting we will celebrate the careers ofJelte de Haan <strong>and</strong> Carlos Blanco,both of whom will have retired by 2008MaastrichtIn June the weather will be mild <strong>and</strong> sunny in Maastricht. The oldest <strong>and</strong> most Burgundian city in theNetherl<strong>and</strong>s offers many ways to relax (terraces, pubs <strong>and</strong> restaurants). Located in the hilliest part ofthe Netherl<strong>and</strong>s, its hills <strong>and</strong> marlstone caves offer great views <strong>and</strong> numerous possibilities for nicewalks, biking, horse-riding, etc. Situated in the center of Europe, Maastricht is a one-hour drive fromBrussels or Düsseldorf, a two-hour drive from Amsterdam. The city also has its own airport withconnections to London, Amsterdam, Pisa, etc.Hotel Van der ValkKnown for its spacious rooms <strong>and</strong> reasonable prices, the Hotel van der Valk in Maastricht is a luxury5-star hotel <strong>and</strong> only a 10-minute walk from the centre of town. The <strong>2007</strong> price for a st<strong>and</strong>ard room is €85 <strong>and</strong> a business suite is € 132,50. When sharing a room, this conference will certainly offer “valuefor money.”<strong>Society</strong> Sport EventThe classic <strong>FNPS</strong> Sport Event will not be forgotten. With the Dutch organizing things, you can be surethat we will have a real competition!Organizing CommitteeThe familiar faces from the Netherl<strong>and</strong>s (i.e. Bilardo, Blanco, De Haan, Mulder, De Vries, Erwich,Hasaart, Nijhuis, Oei, Van den Berg, Visser, etc.) will participate in the Organizing Committee to createa meeting that is successful - both scientifically <strong>and</strong> socially.Contact for the Dutch 2008 <strong>FNPS</strong> MeetingJan G. NijhuisConference Agency Maastrichte-mail: jnij@sgyn.azm.nlinfo@conferenceagency.comT +31 43 387 47 64 T +31 43 361 91 92P.O. Box 5800 P.O. Box 1402NL-6202 AZ MaastrichtNL-6201 BK MaastrichtPage 7


Previous meetings of the <strong>FNPS</strong>1974 Oxford, United Kingdom1975 Oxford, United Kingdom1976 Malmö, Sweden1977 Oxford, United Kingdom1978 Nijmegen, The Netherl<strong>and</strong>s1979 Paris, France1980 Oxford, United Kingdom1981 Maastricht, The Netherl<strong>and</strong>s1982 London, Canada1983 Malmö, Sweden1984 Oxford, United Kingdom1985 Haifa, Israel1986 Banff, Canada1987 Groningen, The Netherl<strong>and</strong>s1988 Cairns, Australia1989 Reading, United Kingdom1990 Pacific Grove, USA1991 De Eemhof, The Netherl<strong>and</strong>s1992 Niagara-the-Lake, Canada1993 Plymouth, United Kingdom1994 Palm Cove, Australia1995 Malmö, Sweden1996 Arica, Chile1997 S.Margherita Ligure, Italy1998 Lake Arrowhead, USA1999 Vliel<strong>and</strong>, The Netherl<strong>and</strong>s2000 Southampton, United Kingdom2001 Auckl<strong>and</strong>, New Zeal<strong>and</strong>2002 Prague, Czech Republic2003 Banff, Canada2004 Tuscany, Italy2005 Glenelg, South Australia2006 Cambridge, UKPage 8


Social EventsMonday, August 27Sporting Event16:00-Activity Room 3F 「Matsushima Minami」, Hotel Sendai PlazaDress code: CasualNintendo Wii WILL OFFER UNPRECEDENTED VIDEO GAME EXPERIENCES FOR EVERYONEAn afternoon of fun, the highlight of which will be the fun sporting game.2006 - For more than 20 years, video game players have used body language to "help" them play.With Nintendo's upcoming Wii? console, those movements become a real part of the play. Aftergrabbing the Wii Remote for the first time, hesitation gives way to concentration. Confidence builds.Excitement morphs into pure delight. And everyone watching says the same thing: "Hey - let metry!"What drives this phenomenon? A remarkable controller <strong>and</strong> games that enhance the experience …realizing that the swing of your arm - not the movement of your thumb - causes a baseball to leavethe park or a sword to find its mark. The Wii console introduces the next leap in gaming, one whereplayers not only control their characters on the screen, but they also become them.Why not test your ability <strong>and</strong> take part? Participants will be divided into groups. The winning teamwill receive a prize.Monday, August 27Dinner Sendai Night19:30-Typical <strong>Japan</strong>ese restaurant in Sendai cityDress code: CasualEnjoy a typical <strong>Japan</strong>ese restaurant, which is called “Izakaya” in <strong>Japan</strong>ese.Izakaya are drinking places that offer a variety of small dishes, such as robata (grilled food), salads<strong>and</strong> finger food. It is probably the most popular restaurant type among the <strong>Japan</strong>ese people.Izakaya tend to be informal, <strong>and</strong> the people at one table usually share all dishes, rather thanordering <strong>and</strong> eating individually.Page 9


Social Events continuedTuesday 28 th AugustExcursion14:00-20.30Matsushima sightseeingZuiganji was founded in 828 as a temple of the Tendai sect. It is now one of the Tohoku's mostfamous Zen temples, well known for its beautifully painted sliding doors (fusuma). Zuiganji Templewas built by Lord Date Masamune. The main hall was built in the traditional study room style ofarchitecture known as shoinzukuri. The roof of its magnificent, single-story main structure is tiled inthe hongawarabuki style, in which round <strong>and</strong> square tiles are laid down alternately. Many parts ofthe temple have been designated as national treasures or important cultural properties. The mainhall, priests' quarters <strong>and</strong> corridors are designated national treasures.Matsushima Bay CruiseDress code: very casualThe fun excursion cruise will be held in the afternoon. You can enjoy talking with other friends <strong>and</strong>colleagues with some foods <strong>and</strong> drinks on a ship for 2 hours.Matsushima, a small bay dotted with more than 260 isl<strong>and</strong>s beautifully covered with evergreens,is one of <strong>Japan</strong>'s celebrated Three Views. Poems have been composed on Matsushima since oldendays. "Matsushima is the most scenic place in <strong>Japan</strong>," wrote Matsuo Basho, the master haiku poet."… Some isl<strong>and</strong>s soar, pointing to the heavens, while others crouch, creeping on the waves….Thepines are deep green, <strong>and</strong> their branches <strong>and</strong> needles have been bent by exposure to the seabreeze….The l<strong>and</strong>scape is enchanting, like an embellished beauty's face."Wednesday 29 th AugustGala dinner19.30-21.30Main Hall 3FDress code: Lounge suitsThis promises to be the social highlight of the meeting <strong>and</strong> is definitely an evening not to be missed.Entertainment will be supplied by Show Asano who is a brilliant shamisen player.shamisen is a stringed instrument shaped like a banjo, but its outst<strong>and</strong>ing feature is that it has nofrets. It is played by striking its three strings with a plectrum <strong>and</strong> is used as accompaniment forKabuki <strong>and</strong> bunraku plays <strong>and</strong> for folk songs. It came from China through Okinawa <strong>and</strong> spreadthroughout <strong>Japan</strong>, in which it developed independently from the original style. It became therepresentative instrument of <strong>Japan</strong> since the Edo Period(1603-1867).Page 10


Notes for speakersOral presentationOral presentations will take place in the Main Hall at the Hotel SendaiPlaza.All oral presentations will be given in Microsoft Powerpoint Office 2000/2002/2003 on a PC.Speakers are asked to submit their presentations on CD or a memory stick to the technician on thePC preview desk at least one hour before their presentation.If you wish to use Macintosh computer or Windows VISTA or moving images, please bring your owncomputer.Oral presentations will be 10 minutes in length followed by a 5 minute discussion. Due to the timeschedule, speakers must ensure their presentation does not exceed this time limit.There will be a prize for the best oral presentation, which will be presented at the Gala dinner onAugust 29th (Wed).Poster presentationPosters can be displayed throughout the meeting. However, there will be a 1.5 hour formal postersession(Wednesday 29th August 11:00 - 12:30).These will be held in Poster Presentation Room(Matsushima Higashi)..There will be a prize for the best poster presentation, which will be presented at the Gala dinner onAugust 29th (Wed).Set-up <strong>and</strong> Removal Hours:Set-up: Sunday, August 26, 10:00-13:00Removal: Wednesday, August 29, 16:45-17:00Poster Guidelines:Poster presenters are expected to be present at their display during thefollowing hours:Wednesday, August 29, 11:00-12:30 (Poster Session)Please Note:*Posterboard size: 180*180cm*A presentation number to be placed at the top left of the poster will beprovided by the secretariat. Each author is requested to indicate "the title,""the authors' names"<strong>and</strong> "the authors' affiliations" at the top right of the panel witin anarea measuring 160cm wide by 20cm high.*Posters are attached to the boards with thumbtacks, which will beprovided by the Secretariat. No paste, glue, staples or nails are permitted.*Presenters are asked to remain with their posters during the aboveassigned hours to answer questions <strong>and</strong> exp<strong>and</strong> on their material for visitors.Page 11


General InformationRegistrationRegistration Desk will be located on the 3 rd floor at Hotel Sendai Plaza.Opening hours:Satuday 25 th August 17:00-19:00Sunday 26 th August 9:00-17:00Monday 27 th August 7:30-19:30Tuesday 28 th August 8:00-14:00Wednesday 29 th August 8:00-19:30It will be manned throughout the meeting <strong>and</strong> should be the first port of call for delegates <strong>and</strong>accompanying persons requiring any assistance.Internet accessInternet access will be available, free of charge on the 3 rd floor at Hotel Sendai Plaza. Out ofconsideration to others, delegates are kindly requested to limit their access time to 10 minuteseach.Trade ExhibitorsTrade Exhibitors will be located on the 3 rd floor at Hotel Sendai Plaza.Please take the time to visit them as they have been generous sponsors of this meeting.RefreshmentsAfternoon tea will be served in the poster presentation room(Matsushima Higashi) on theLunch will be served in the activity room(Matsushima Minami).Page 12


ACCESS TO SENDAIPage 13


SENDAI HOTEL MAPHOTEL SENDAI PLAZA2-20-1 HON-CHOU AOBA-KU, SENDAI-CITY, MIYAGI, 980-0014 JAPANTelephone: 022-262-7111 Facsimile: 022-262-8169Page 14


HOTEL SENDAI PLAZA 3FPage 15


AT A GLANCEPage 16


Page 17


Page 18


Page 20


ProgramPresenting authors are underlinedSATURDAY, AUGUST 25, <strong>2007</strong>17:00-19:00 Registration19:00 You can freely participate in the banquet of JSMES* meetingSUNDAY, AUGUST 26, <strong>2007</strong>9:00- Registration12.00 – 12.45 LunchPlenary lecture 1 :ChairpersonTsuyomu Ikenoue13:30-14:00 <strong>Fetal</strong> Heart Rate Monitoring: Is it Time to Establish International Consensus onIntrapartum Management?Julian T. Parer14:00-14:05 DiscussionPlenary lecture 2 :ChairpersonsShiro Kozuma14:05-14:35 Developmental Origins of Disease - a <strong>Japan</strong>ese problem <strong>and</strong> a <strong>Japan</strong>eseanswer?Mark Hanson14:35-14:40 DiscussionPlenary lecture 3 :ChairpersonYoshio Matsuda14:40-15:10 An excess of circulating antiangiogenic factors: the cause of the maternalhypertensive syndrome of preeclampsia Richard Levine15:10-15:15 Discussion15.15 – 15.45 Afternoon teaPlenary lecture 4 :ChairpersonHiroshi Chisaka15:45-16:15 The affect of maternal hyperglycemia on fetal outcomeHuixia Yang16:15-16:20 DiscussionPage 21


SUNDAY, AUGUST 26, <strong>2007</strong> (continued)Plenary lecture 5 :ChairpersonTakashi Okai16:20-16:50 The pathogenesis of preterm brain injuryLaura Bennet16:50-16:55 DiscussionDINNER(We offer a meal coupon. You can choose one of the restaurant in Hotel Sendai Plaza.)MONDAY, AUGUST 27, <strong>2007</strong>7.30 – RegistrationSESSION 1 : <strong>Fetal</strong> GrowthChairpersonsGraham JenkinAlison Forhead8:00-8:30 Keynote speakerAbstract1: Impact of low birthweight on developing organs: separating the effectsof IUGR <strong>and</strong> preterm birth.(including Discussion)Richard Harding8:30-8:40 Abstract2: Postnatal high-fat feeding impacts on metabolic profiles <strong>and</strong>gluconeogenic capacity during fasting <strong>and</strong> this is exaggerated by prenatalundernutrition in adolescent lambsAnna Hauntoft Kongsted <strong>and</strong> Mette Olaf Nielsen8:40-8:45 Discussion8:45-8:55 Abstract3: Maternal Isocaloric High-protein Diet Ameliorates the Elevation in bothBlood Pressure <strong>and</strong> Cardiac Remodeling in the Adult Mice Offspring withMaternal Calorie Restriction Possible Involvement of Branched-Chain AminoAcids-Hiroaki Itoh, Makoto Kawamura, Shigeo Yura, Haruta Mogami,Fujii Tsuyoshi <strong>and</strong> Norimasa Sagawa8:55-9:00 Discussion9:00-9:10 Abstract4: Maternal ovine cortisol concentrations are important for key endocrinefactors involved in fetal growthEllen Jensen, Bennet L, Wood CE, Gunn AJ <strong>and</strong> Keller-Wood MPage 22


MONDAY, AUGUST 27, <strong>2007</strong> (continued)9:10-9:15 Discussion9:15-9:25 Abstract5: Diet-Induced Obesity <strong>and</strong> Prenatal Undernutrition Lead to CentralLeptin Resistance by Different MechanismsMhoyra Fraser, Charisma K Dhaliwal, Stefan Krechowec,Mark Vickers <strong>and</strong> Bernhard H Breier9:25-9:30 Discussion9:30-9:40 Abstract6: Impact of maternal exposure to constant light during late gestation onpostnatal adrenal function in the capuchin monkey: long terms effects on DHAS<strong>and</strong> transient effects on cortisol.Claudia Torres-Farfan, N Mendez, H Richter, Gj Valenzuela <strong>and</strong> M Seron-Ferre9:40-9:45 Discussion9:45-9:55 Abstract7: Maternal signals control the fetal biological clockHidenobu Ohta, Xu Shanhai, Takahiro Moriya, Masayuki Iigo, Tatsuya Watanabe,Norimichi Nakahata, Hiroshi Chisaka, Tadashi Matsuda, Toshihiro Ohura,Yoshitaka Kimura, Nobuo Yaegashi, Shigeru Tsuchiya, Hajime Tei <strong>and</strong> Kunihiro Okamura9:55-10:00 Discussion10:00-10:10 Abstract8: Dietry protein restriction of pregnant mouse induces alteredmethylation of Oct-4 <strong>and</strong> Sphk-1 gene promoters in the liver of offspring.Jun Murotsuki, Kunihiro, Okamura, Hiroi Kaku, Yoshihiko Araki,Hiroshi Yoshitake, Mark A. Hanson <strong>and</strong> Felino R. Cagampang10:10-10:15 Discussion10:15-10:25 Abstract9: Transcriptional suppression of IGF-2 mediated by altered histonemodifications in a maternal protein restricted IUGR mouse modelMichiyo Nakamura, Sharif J, Ito T, Chisaka H, Kimura Y, Mitsuya K <strong>and</strong> Okamura K10:25-10:30 Discussion10:30-11:00 Morning tea (poster viewing)SESSION 2 : NeonateChairpersonsR Usha RajIan Wright11:00-11:30 Keynote speakerAbstract10: The state of the art in head cooling for neonatal encephalopathyAlistair J Gunn(including Discussion)Page 23


MONDAY, AUGUST 27, <strong>2007</strong> (continued)11:30-11:40 Abstract11: Cerebral oxygenation measured by spatially resolved spectroscopy innewborn lambsFlora Wong, Theodora Alexiou, Brodecky V, Samarasinghe T,Wilkinson M <strong>and</strong> Walker AM11:40-11:45 Discussion11:45-11:55 Abstract12: Treatment of fetal lung hypoplasia with antenatal corticosteroids:effects on pulmonary circulation <strong>and</strong> respiration in the newborn sheepKeiji Suzuki, Hooper SB <strong>and</strong> Harding R11:55-12:00 Discussion12:00-12:10 Abstract13: Clinical presentation, causes <strong>and</strong> outcome of Persistent PulmonaryHypertension of Newborns (PPHN) in a tertiary care hospital of Karachi.Samana Ali, Parkash Jai, Feroze Asher, Atiq Mehnaz <strong>and</strong> Khan Iqtidar12:10-12:15 Discussion12:15-12:25 Abstract14: Disease spectrum term neonates admitting in <strong>Neonatal</strong> ICUSamana Ali <strong>and</strong> Salat Sohail12:25-12:30 Discussion12:30-12:40 Abstract15: Prenatal Diagnosis of Schizencephaly with Septo-optic Dysplasia byUltrasound <strong>and</strong> Magnetic Resonance ImageJeng-Hsiu Hung, Shu-Huei Shen, Wan-You Guo, Chih-Yao Chen,Kuan-Chong Chao, Ming-Jie Yang <strong>and</strong> Chia-Yi Selena Hung12:40-12:45 Discussion12:45-13:45 LunchSESSION 3 : Perinatal medicineChairpersonsJulian T. ParerTomoaki Ikeda13:45-13:55 Abstract16: A possibility of applying system-engineering methods to amniotic fluidvolume changes.Yoshiyasu Hombo <strong>and</strong> Michio Ooshita13:55-14:00 DiscussionPage 24


MONDAY, AUGUST 27, <strong>2007</strong> (continued)14:00-14:10 Abstract17: Pregnancy after Stillbirth <strong>and</strong> Home <strong>Fetal</strong> Heart Rate Monitoringvia the InternetJason H Collins14:10-14:15 Discussion14:15-14:25 Abstract18: Prediction of fetal coarctation of the aorta from the three vessel <strong>and</strong>tracheal viewHelena Gardiner, Hikoro Matsui , Lucia Pasquini, Anna Seale,Mats Mell<strong>and</strong>er, Michael Roughton <strong>and</strong> Siew Yen Ho14:25-14:30 Discussion14:30-14:40 Abstract19: Development of new digital fetal monitor "Behaviogram" for earlytomid pregnancyNorio Shinozuka14:40-14:45 Discussion14:45-14:55 Abstract20: Comparison of long axis cardiac function assessed by Vector VelocityImaging <strong>and</strong> conventional tissue Doppler <strong>and</strong> M-mode methods in the humanfetusHelena Gardiner, Hikoro Matsui <strong>and</strong> Ioannis Germanakis14:55-15:00 Discussion15:00-15:10 Abstract21: The genetic analysis of deep vein thrombosis during pregnancy <strong>and</strong>perinatal associated disorders in japaneseReiko Neki, N. Yamada, Y. Tokito, N. Iwanaga, K. Ueda, K. Yamanaka,M. Nozawa, T. Ikeda, T. Fujita, J. Ishikawa, Y. Sato, T. Miyata, O. Fukui <strong>and</strong> N,Suehara15:10-15:15 Discussion15:15-15:25 Abstract22: A new monitoring method for FGR fetusesKazunao Suzuki, Motoi Sugimura <strong>and</strong> Naohiro Kanayama15:25-15:30 Discussion16:00- SPORTING EVENT19:30- DINNER SENDAI NIGHTPage 25


TUESDAY, AUGUST 28, <strong>2007</strong>8.00 – RegistrationSESSION 4 : BrainChairpersonsLaura BennetMhoyra Fraser8:30-9:00 Keynote speakerAbstract23: Growth hormone: A neurotrophic factor during chick embryogenesis.Steve Harvey, Marie-Laure Baudet <strong>and</strong> Esmond J S<strong>and</strong>ers(including Discussion)9:00-9:10 Abstract24: Maternal administration of dexamethasone causes significantalterations in brain activity in the preterm fetal sheep.Joanne Davidson, Lindsea Booth, Josine Quaedackers,Alistair J Gunn <strong>and</strong> Laura Bennet9:10-9:15 Discussion9:15-9:25 Abstract25: Therapeutic Hypothermia Changes the Prognostic Value of ClinicalEvaluation of <strong>Neonatal</strong> Encephalopathy.Alistair Jan Gunn, Andrew Whitelaw, John Barks, John S. Wyatt,Denis Azzopardi, Charlene M. Robertson <strong>and</strong> Marianne Thoresen9:25-9:30 Discussion930-9:40 Abstract26: Can erythropoietin be used to protect the fetal sheep brain fromrepeated exposure to endotoxin?Robert De Matteo, L Cardamone, V Stacy, N Blasch, M Probyn,N Hale, S Rees <strong>and</strong> R Harding9:40-9:45 Discussion9:45-9:55 Abstract27: Anti-inflammatory effects of Sulfasalazine in an ovine model of inutero infection: a prospect for neuroprotection?Burcu Saglam, Graham Jenkin, Suzanne L. Miller <strong>and</strong> Euan M. Wallace9:55-10:00 Discussion10:00-10:10 Abstract28: <strong>Fetal</strong> brain injury in single umiblical artery ligation (SUAL) inducedIUGRSuzanne L. Miller, Low HM, Supramaniam VG,Castillo-Meléndez M, Jenkin G <strong>and</strong> Wallace EM10:10-10:15 DiscussionPage 26


TUESDAY, AUGUST 28, <strong>2007</strong> (continued)10:15-10:25 Abstract29: Perinatal DiI tracing of developing hypothalamic connectionsIrina G. Makarenko <strong>and</strong> Alpeeva E.V10:25-10:30 Discussion10:30-11:00 Morning tea (poster viewing)SESSION 5 : HypoxiaChairpersonsBrian KoosAlistair Gunn11:00-11:30 Keynote speakerAbstract30: Chronic hyoxia : Consquence <strong>and</strong> Treatment.Suzanne L. Miller , Veena G. Supramaniam, Euan M. Wallace<strong>and</strong> Graham Jenkin(including Discussion)11:30-11:40 Abstract31: Proliferation in the subventricular zone (SVZ) after severe hypoxia<strong>and</strong> induced hypothermia in preterm fetal sheepSherly George, Robert Barrett, Laura Bennet,Justin Dean <strong>and</strong> Alistair Jan Gunn11:40-11:45 Discussion11:45-11:55 Abstract32: The ontogeny of chemoreflex <strong>and</strong> hemodynamic responses toprolonged umbilical cord occlusion in fetal sheepGuido Wassink, Laura Bennet, Lindsea C. Booth, Ellen C. Jensen,Bert Wibbens, Justin M Dean, <strong>and</strong> Alistair J Gunn11:55-12:00 Discussion12:00-12:10 Abstract33: Effect of acute <strong>and</strong> chronic hypoxia on amniotic fluid gases in chickembryogenesisM. V. Nechaeva, H. Toenhardt <strong>and</strong> D. Marquardt12:10-12:15 Discussion12:15-12:25 Abstract34: Maternal treatment with allopurinol diminishes fetal cardiac oxidativestress following repeated episodes of ischaemia-reperfusion in sheepJan B Derks, H Torrance , MA Oudijk, AS Thakor,T Cindrova-Davies, F van Bel, GHA Visser, GJ Burton <strong>and</strong> DA GiussaniPage 27


TUESDAY, AUGUST 28, <strong>2007</strong> (continued)12:25-12:30 Discussion12:30-12:40 Abstract35: Pre-existing hypoxia is associated with a delayed but more sustainedrise in T/QRS ratio during prolonged umbilical cord occlusion in near-term fetalsheepLaura Bennet, Bert Wibbens, Jenny A. Westgate,Harmen H. De Haan, Guido Wassink, <strong>and</strong> Alistair J. Gunn12:40-12:45 Discussion12:45- Lunch14:00- Free or excursion (including cruising dinner)WEDNESDAY, AUGUST 29, <strong>2007</strong>8.00 – RegistrationSESSION 6 : PlacentaChairpersonsEllen JensenAlan Bocking8:30-9:15 Invited lectureRole of PEG10 in mammalian placenta development <strong>and</strong> evolution.Fumitoshi Ishino(including Discussion)9:15-9:25 Abstract36: High incidence of ketosis <strong>and</strong> hyper-homocysteinemia in the lasttrimester of <strong>Japan</strong>ese mothers.without any complicationsHideoki Fukuoka, Hiroko Watanabe, Yasushi Nagai,Chiyoko Ogasawara <strong>and</strong> Shigetaka Asano9:25-9:30 Discussion9:30-9:40 Abstract37: Genomic imprinting in the placenta involves histone tail modificationsindependent of DNA methylationKohzoh Mitsuya <strong>and</strong> Kunihiro Okamura9:40-9:45 DiscussionPage 28


WEDNESDAY, AUGUST 29, <strong>2007</strong> (continued)9:45-9:55 Abstract38: Lactobacilli supernatant inhibits TNF-α production <strong>and</strong> COX2expression in LPS-activated placental trophoblastsAlan Bocking, M Yeganegi, C Watson, 2 S Kim, G Reid <strong>and</strong> J Challis9:55-10:00 Discussion10:00-10:10 Abstract39: Interaction between Lipoxygenase Pathways <strong>and</strong> Progesterone onthe Regulation of 11 -Hydroxysteroid Dehydrogenase type 2 in Cultured HumanTerm Placental TrophoblastsKazuyo Sato, Hiroshi Chisaka, John R.G.Challis <strong>and</strong> Kunihiro Okamura10:10-10:15 Discussion10:15-10:25 Abstract40: Distribution of the thyroid hormone transporter, MCT8, in fetal <strong>and</strong>placental tissues of murine, human <strong>and</strong> ovine speciesAlison J Forhead, Jenny L Macrae, Theo J Visser,Graham J Burton, FB Peter Wooding <strong>and</strong> Abigail L Fowden10:25-10:30 Discussion10:30-11:00 Morning tea (with poster session)11:00-12:30 Poster sessionFor a list of poster presentation, please refer to posters12:30-13:45 LunchSESSION 7 : Circulation, CardiovascularChairpersonsJun MurotsukiJan Derks13:45-14:30 Invited lectureCell sheet engineering for tissue <strong>and</strong> organ regenerationTeruo Okano(including Discussion)14:30-14:40 Abstract41:Determinants of the number of cardiomyocytes in sheep: theinfluence of fetal <strong>and</strong> postnatal growthRichard Harding, Victoria Stacy, Megan Probyn,Robert DeMatteo, Nigel Wreford <strong>and</strong> M. Jane Black14:40-14:45 DiscussionPage 29


WEDNESDAY, AUGUST 29, <strong>2007</strong> (continued)14:45-14:55 Abstract42: Modulation of Pulmonary Vascular Smooth Muscle Cell (PVSMC)Phenotype in Hypoxia: Role of cGMP-Dependent Protein Kinase (PKG)J Usha Raj, Weilin Zhou, Chiranjib Dasgupta <strong>and</strong> Sewite Negash14:55-15:00 Discussion15:00-15:10 Abstract43: Human fetal cardiovascular baro-reflex responses in maternal steroidadministration analyzed by pulsed Doppler minute tissue displacementmeasurementNorio Shinozuka15:10-15:15 Discussion15:15-15:25 Abstract44: Effects of maternal betamethasone administration on regional bloodflow in the intrauterine growth restricted (IUGR) ovine fetusVeena Supramaniam, Jenkin G, Wallace EM <strong>and</strong> Miller SL15:25-15:30 Discussion15:30-15:40 Abstract45: A Novel extraction method of fetal electrocardiogram from thecomposite abdominal signal.Yoshitaka Kimura,Takuya Ito, Michiyo Nakamura, Kaori Uchida, Hiroshi Chisaka,Norihiro Katayama, Mitsuyuki Nakao <strong>and</strong> Kunihiro Okamura15:40-15:45 Discussion15:45-15:55 Abstract46: The effects of the tocolytics atosiban <strong>and</strong> nifedipine on fetalmovements, heart rate, <strong>and</strong> blood flow.Jan B Derks, Roel de Heus, Eduard J.H. Mulder <strong>and</strong> Gerard H.A. Visser15:55-16:00 Discussion16:00-16:10 Abstract47:The effect of Sulfasalazine on LPS-induced changes in fetaloxygenation <strong>and</strong> carotid blood flowBurcu Saglam, Graham Jenkin, Suzanne Miller <strong>and</strong> Euan Wallace16:10-16:15 Discussion16:15-16:45 Afternoon teaGeoffrey Dawes Lecture16:45-17:45 Avian influenza <strong>and</strong> p<strong>and</strong>emic threat(including Discussion)ChairpersonKunihiro OkamuraHitoshi OshitaniPage 30


WEDNESDAY, AUGUST 29, <strong>2007</strong> (continued)17:45-18:45 Business Meeting/AGM19:30- GALA DINNERPage 31


Page 32


Page 34


LIST OF POSTER PRESENTATIONP1: The role of the neural sympathetic <strong>and</strong> parasympathetic systems in diurnal <strong>and</strong> sleep statecardiovascular rhythms in the late gestation ovine fetus.Ellen Jensen, Laura Bennet, Sarah-Jane Guild,Lindsea C. Booth, Jenny A. Westgate <strong>and</strong> Alistair J. GunnP2: Np95, a master regulator of the epigenome, is essentially required for embryonic developmentSharif Jafar, Muto M., Nakamura M., Takebayashi S.,Okano M., Koseki H., Nagamune T., Mitsuya K. <strong>and</strong> Okamura KP3: Evaluation of the placental vascularity using fractal parameters: could they help in earlydiagnosis of fetal growth restriction?Caterina Guiot, E. Piccoli, F. Saccom<strong>and</strong>i <strong>and</strong> T. TodrosP4: <strong>Fetal</strong> <strong>and</strong> perinatal growth curves: what can we learn from them?Caterina Guiot, Tullia Todros, Antonio Gliozzi <strong>and</strong> Pier Paolo DelsantoP5: Nitric Oxide <strong>and</strong> CD4+25+Regulatory T cell production from iNOS knockout mice <strong>and</strong> their F1mice treated with LPS.Hidenori Takahashi, Toshiaki Okawa, Keiya Fujimori <strong>and</strong> Akira SatoP6: Nitric Oxide production in placental tissue from iNOS knockout mice with LPS <strong>and</strong> theiroffsprings.Hidenori Takahashi, Toshiaki Okawa, Keiya Fujimori <strong>and</strong> Akira SatoP7: Maternal severe undernutrition during both late gestation <strong>and</strong> lactation period inducehypertension in male rat offspringHidenori Takahashi, Toshiaki Okawa, Keiya Fujimori <strong>and</strong> Akira SatoP8: Regulation of maternal feeding during lactation period may control adulthood hypertension.Hidenori Takahashi, Toshiaki Okawa, Keiya Fujimori <strong>and</strong> Akira SatoP9: Maternal regulation of high fat nourishment during lactation period reduce a hypertension ofmale offspringHidenori Takahashi, Toshiaki Okawa, Keiya Fujimori <strong>and</strong> Akira SatoP10: Epigenetic stability <strong>and</strong> developmental plasticity in cloned placentae during somatic cellnuclear tranfer.Naomi Matsuda, Jafar Sharif, Kimiko Inoue, Narumi Ogonuki,Hiromi Miki, Hiroshi Chisaka, Kunihiro Okamura, Atsuo Ogura <strong>and</strong> Kohzoh MitsuyaP11: Male disadvantage? Gender <strong>and</strong> fetal cardiovascular responses to asphyxia in preterm fetalsheepLaura Bennet, Bert Wibbens, Jenny A. Westgate,Harmen H. De Haan, Guido Wassink <strong>and</strong> Alistair J. GunnPage 35


P12: <strong>Fetal</strong> brain, liver <strong>and</strong> renal blood flow assessment using 3D power Doppler ultrasoundFong –Ming Chang, Chiung-Hsin Chang, Chen-Hsiang Yu,Lin Kang, Pei-Ying Tsai <strong>and</strong> Huei-Chen KoP13: Molecular Genetic Study on Angiotensin-Converting Enzyme Gene <strong>and</strong> Preeclampsia inTaiwanese: Two polymorphisms Tested <strong>and</strong> as HaplotypesFong –Ming Chang, Ming-Hui Chen, Chiung-Hsin Chang,Chia-Fu Li , Chen-Hsiang Yu <strong>and</strong> Huei-Chen KoP14: Clinical application of electromechanical delay time for indirect evaluation of fetal bloodpressure during labor <strong>and</strong> delivery. A preliminary study of 18 full-term infants.Yasuyuki Kawagoe, Hiroshi Sameshima <strong>and</strong> Tsuyomu IkenoueP15: Temporal Changes in Mediators of Microvascular Tone are Influenced by Sex <strong>and</strong>Glucocorticoid Exposure in Preterm InfantsIan Wright, Stark MJ <strong>and</strong> Clifton VLP16: The effect of carperitide, human atrial natriuretic peptide, on acute heart failure caused byendocardial cushion defect with atrioventricular block in a premature infant: a case reportYuichiro Miura, Tadashi Matsuda, Masaki Sato, Shizuko Akiyama,Ryuta Kitanishi, Takushi Hanita, Tatsuya Watanabe <strong>and</strong> Hidenobu OhtaP17: Effect of Maternal Stress on <strong>Fetal</strong> Heart Rate Assessed by Vibroacoustic StimulationIkuko Makino, Yoshio Matsuda, Marie Yoneyama , Kyoko Hirasawa ,Koichiro Takagi, Hiroaki Ohta <strong>and</strong> Yukuo KonishiP18: Non-Invasive long-term fetal ECG monitoringJan B. Derks, Graatsma E.M., Mulder E.J.H.<strong>and</strong> Visser G.H.AP19: The Relationship of Microchimerism to AbortionTomoko Sato, Keiya Fujimori, Akira Sato, Hitoshi Ohto, Kenich Hata,Hiroko Sasaki, Mihoko Yazawa, Shizuka Honda <strong>and</strong> Mayumi NoguchiP20: The effect of cerebral hypothermia on cortisol <strong>and</strong> ACTH responses after umbilical cordocclusion in preterm fetal sheep.Joanne Davidson, Mhoyra Fraser, Andrew S. Naylor,Vincent Roelfsema, Alistair J. Gunn <strong>and</strong> Laura BennetP21: The prevention of fetal brain <strong>and</strong> lung injuries based on interleukin-6 levels in amniotic fluidbefore birthTakushi Hanita, Tadashi Matsuda, Masaki Sato, Shizuko Akiyama,Ryuta Kitanishi, Yuichiro Miura, Tatsuya Watanabe, <strong>and</strong> Hidenobu OhtaP22: Effect of intrauterine inflammation on induction of antenatal periventricular leukomalacia inchronically instrumented fetal sheepMasatoshi Saito , Tadashi MatsudaPage 36


P23: Maternal Low Protein Diet Aggravates <strong>Fetal</strong> White Matter Damage Caused by InfectionTakuya Ito, Kaori Uchida, Michiyo Nakamura, HiroshiChisaka, Yoshitaka Kimura <strong>and</strong> Kunihiro OkamuraP24: Prenatal diagnosis of bradycardia using fetal electrocardiogram via maternal abdomenYoshitaka Kimura, Michiyo Nakamura, Kaori Uchida, Takuya Ito,Hiroshi Chisaka <strong>and</strong> Kunihiro OkamuraP25: Endothelin-1 inhibits both L-type Ca2+ current <strong>and</strong> ATP-sensitive K+ current in neonatal ratventricular myocytes.Yasuhiro Katsube, Nobuko Suzuki, Makoto Watanabe, Masanori Abe,Miharu Hajikano, Mitsuhiro Kamisago, Ryuji Fukazawa <strong>and</strong> Shunichi OgawaP26: Placenta accreta/increta in unscarred uterusKozo Akagi, Shima T, Ishigaki N, Oota S, Hayasaka A, Fujita N, Asano K <strong>and</strong> Wada YP27: QT interval during pregnancy in Long QT syndromeYuri Tokitou, Akiko Omoto, Naoshi Yamada, Naoko Iwanaga, Kaoru Yamanaka,Keiko Ueda, Masayo Nozawa, Reiko Neki, Wataru Shimizu <strong>and</strong> Tomoaki IkedaP28: High altitude chronic hypoxia modifies cardiovascular responses in newborn sheep dependingon the time <strong>and</strong> duration of the hypoxic insult during gestation.EA Herrera, G Ebensperger,RA Riquelme, C Torres-Farfan,VR Reyes, JT Parer, DA Giussani <strong>and</strong> AJ Llanos.Page 37


Page 38


Abstract Plenary lecture 1<strong>Fetal</strong> heart rate monitoring: Is it time to establish internationalconsensus on Intrapartum management?J T ParerDepartment of Obstetrics, Gynecology & Reproductive SciencesUniversity of California San FranciscoDespite numerous attempts in the past several yearsthere is no broad agreement on FHR pattern management.In order to develop a framework we have classified fetalheart rate monitor patterns according to risk of fetalacidemia, <strong>and</strong> risk of evolution to a more serious pattern,<strong>and</strong> we have used this information to construct ast<strong>and</strong>ardized process for FHR pattern management, withthe ultimate aim of minimizing newborn acidemia withoutexcessive obstetric intervention.Each FHR pattern has been color-coded, from no threat offetal acidemia (green, no intervention required) to severethreat of acidemia (red, rapid delivery recommended).Three intermediate categories (blue, yellow <strong>and</strong> orange)require escalated informing of appropriate individuals forintervention <strong>and</strong> resuscitation (obstetrician, anesthesiologist,<strong>and</strong> neonatal resuscitator) <strong>and</strong> preparation for urgentdelivery, e.g. staff <strong>and</strong> surgical suite availability, <strong>and</strong>conservative techniques to ameliorate the FHR patterns.We have identified 134 FHR patterns classified bybaseline rate, baseline variability, <strong>and</strong> type ofdeceleration. Based on the best available evidence wehave assigned a risk of newborn acidemia or low 5 minApgar score to these patterns. We have also evaluatedeach pattern for the risk that the pattern would furtherevolve into a pattern with a higher risk of acidemia.This framework is potentially applicable to the institutionswhere it was developed, <strong>and</strong> will need to be modified forother situations depending on the logistics, facilities <strong>and</strong>personnel available. This may provide a framework fordeveloping algorithms for st<strong>and</strong>ardized management ofFHR patterns during labor, which can be tested for validity.NotesPage 40


Plenary lecture 2Developmental Origins of Disease – a <strong>Japan</strong>ese problem <strong>and</strong> a<strong>Japan</strong>ese answer?AbstractMark HansonInstitute Of Developmental Sciences, University of Southampton, UKIt is now well-established that low birthweight is linked toan increased risk of many chronic, non-communicableadult diseases. But size at birth is a poor proxy measureof the prenatal environment, so that smallness does notnecessarily mean that pathological processes havestarted even before birth. Moreover, animal studies showthat stimuli such as unbalanced nutrition in pregnancy canproduce effects on the offspring which resemble manyaspects of the human metabolic syndrome, but withoutinvariably reducing size at birth. Changes in the trajectoryof development of a range of tissues, organs <strong>and</strong> controlsystems is part of the ‘strategy’ which the developingembryo, <strong>and</strong> then the fetus, adopt to optimise thephenotype developed to match the environment predictedfor postnatal life. The processes involved in thisdevelopmental matching (1) have presumably beenselected through evolutionary pressure in many species,including our own, to maximise survival to reproduce.Negative consequences of this strategy may emerge inlater life, <strong>and</strong> increasing longevity in many populations willincrease their incidence. When mismatch occurs, the riskof disease is increased. This may be due to inaccuratecues about the environment, transmitted from the mother,or because the environment has changed within ageneration.Historically, <strong>Japan</strong>ese people were very long-lived. This ischanging as diets <strong>and</strong> physical activity patterns change,increasing potential mismatch. Other factors in <strong>Japan</strong> alsoaffect development. Falling family size increasesprimiparous pregnancy <strong>and</strong> childbearing age is increasing.Both of these increase maternal constraint of fetaldevelopment. Whilst <strong>Japan</strong> was highly effective inreducing the incidence of low birthweight <strong>and</strong> pretermbirth in the 1970s <strong>and</strong> 80s, mean birthweight has nowfallen over about the last 20yr, especially in cities. Thisappears to be due to dieting in young women to loseweight, perhaps to smoking, <strong>and</strong> advice from clinicians torestrict weight gain in pregnancy to low levels.Unbalanced diet <strong>and</strong> low BMI send inappropriate cues tothe developing fetus, increasing risk of mismatch.Coupled with a tendency to deposit abdominal fat whichmay be partly genetic, there are a range of factors whichmust raise great concern about the future incidence ofchronic disease in <strong>Japan</strong>. But perhaps <strong>Japan</strong> can teachother countries lessons about large scale educationalinterventions too – the Basic Law on Shokuiku enacted inJune 2005 promotes knowledge about food <strong>and</strong> the abilityto make food choices, at multiple levels in the population.Will this <strong>Japan</strong>ese problem have a <strong>Japan</strong>ese solution?MAH is supported by the British Heart Foundation1. Gluckman PD <strong>and</strong> Hanson MA. Mismatch – why ourworld no longer fits our bodies. OUP 2006.NotesPage 41


Abstract Plenary lecture 3An Excess of Circulating Anti-Angiogenic Factors: the Cause ofthe Hypertensive Syndrome of PreeclampsiaRichard J. Levine, MDDivision of Epidemiology, Statistics, <strong>and</strong> Prevention Research, National Institute of Child Health<strong>and</strong> Human Development, Department of Health <strong>and</strong> Human Services, Bethesda, MD, USAThe maternal syndrome of preeclampsia may be causedby release of excessive quantities of two anti-angiogenicproteins from the placenta into maternal blood: solublefms-like tyrosine kinase 1 (sFlt1) <strong>and</strong> soluble endoglin(sEng). High circulating concentrations of both moleculesare usually required to cause preeclampsia. Exogenousgene transfer of sFlt1 <strong>and</strong> sEng into rats using anadenoviral vector produced hypertension, proteinuria,glomerular endotheliosis, <strong>and</strong> a HELLP-like syndromewith hemolysis, elevated circulating levels of liverenzymes, low platelet counts, <strong>and</strong> severe restriction offetal growth. sFlt1 acts through binding <strong>and</strong> neutralizingvascular endothelial growth factor (VEGF) <strong>and</strong> placentalgrowth factor (PlGF), preventing them from attaching tocell-surface receptors. sEng is thought to interfere withbinding of TGF-β to endoglin cell-surface co-receptors.Circulating concentrations of sFlt1 <strong>and</strong> sEng are elevatedweeks, even months, before onset of preeclampsia; <strong>and</strong>they increase considerably during the last two months ofnormal pregnancy. High levels are associated with earlypreeclampsia onset <strong>and</strong> the delivery of a small-forgestational-ageinfant. Risk factors for preeclampsia –such as multiple gestation (increases risk) <strong>and</strong> cigarettesmoking (decreases risk) – appear to operate by alteringangiogenic factor levels. I hypothesize that 1) an excessof circulating anti-angiogenic factors is both necessary<strong>and</strong> sufficient to cause clinical preeclampsia <strong>and</strong> that 2)rising levels of circulating anti-angiogenic factors play animportant role in concluding pregnancy <strong>and</strong> triggering theonset of labor. In the future I believe that successfultreatment will become available for the prevention ortherapy of preeclampsia by antagonizing or removingsFlt1 <strong>and</strong>/or sEng or by administering VEGF or PlGF.NotesPage 42


Plenary lecture 4Effects of maternal hyperglycemia on both mothers <strong>and</strong> theirinfantsAbstractHuixia YangDepartment of Obstetrics <strong>and</strong> Gynecology, Peking University First Hospital, Beijing, ChinaWith the improved nutrition <strong>and</strong> continued industrializationover the past decade, both of obesity <strong>and</strong> diabetes inChina are increasing quickly. Gestational DiabetesMellitus (GDM) is a growing problem in China. Aprospective population-based study of 16,286 pregnantwomen has been conducted in 18 cities of China. Theoverall prevalence of abnormal carbohydrate metabolismwas 10.4% when using ADA criteria, the prevalence inSouth of China was higher than that of in the North.Many studies have showed that GDM is linked to a varietyof separate conditions, including: preterm labor,preeclampsia, increased cesarean section rates,macrosomia, birth injury, stillbirth, <strong>and</strong> neonatalhypoglycemia <strong>and</strong> hyperbilirubinemia. With tight maternalblood glucose controlled, pregnancy outcomes in womenwith GDM have been improved.Glucose intolerance diagnosed during pregnancy,identifies women at high risk for Type 2 diabetes later inlife. Recent studies have also shown GDM to beassociated with increased risk of obesity <strong>and</strong> metabolicsyndrome later in life for both mothers <strong>and</strong> their babies.So, it will be very important to early diagnose <strong>and</strong> managethese women with GDM in order to improve both mothers<strong>and</strong> their infants’ outcomes.NotesPage 43


Abstract Plenary lecture 5The pathogenesis of preterm brain injury: underst<strong>and</strong>ing timingLaura Bennet<strong>Fetal</strong> Physiology <strong>and</strong> Neuroscience Group, Dept PhysiologyThe University of Auckl<strong>and</strong>, Auckl<strong>and</strong>, New Zeal<strong>and</strong>A viable neuroprotection treatment will soon be availablefor the term newborn infant suffering fromhypoxic-ischaemic encephalopathy: that of cerebralhypothermia. Currently, however, we do not know whetherthis treatment will be effective for the more vulnerablepreterm infant. Application of hypothermia, or any othertreatment, requires that we underst<strong>and</strong> the mechanismsmediating neural injury <strong>and</strong> when injury occurs.While it is likely that many of the mechanisms mediatinginjury at term are the same for the preterm, aspects of thepatterns <strong>and</strong> evolution of injury are quite different. Preterminfants have a high rate of neurodevelopmental h<strong>and</strong>icap.Recent imaging studies have revealed that adverseoutcomes are strongly associated with reduced braingrowth <strong>and</strong> neural complexity later in life. Increasing datasuggest that these chronic deficits primarily reflect acuteneuronal <strong>and</strong> glial injury sustained during adverse eventsin utero, such as exposure to severe hypoxia-ischaemia<strong>and</strong> inflammation. This chronic impairment appears to bepartly due to upregulation of physiological apoptosis,related to input deprivation <strong>and</strong> output isolation secondaryto acute white <strong>and</strong> grey matter damage <strong>and</strong> axonal injury.However, progenitor cells in the subventricular zone arealso vulnerable to injury, <strong>and</strong> loss of this critical populationlikely further compromises brain development. Acutehypoxic brain damage in premature infants has beenprincipally related to a supposed unique cellularvulnerability to glutamate <strong>and</strong> free radicals. However, it isunlikely that in vivo these damaging processes occur inisolation, <strong>and</strong> that the brain is simply a passive ‘immature’target for toxic factors.In this talk I will examine recent evidence that survival ofvulnerable, injured neurons <strong>and</strong> immatureoligodendrocytes after acute injury is determined by adynamic balance between damaging events such asincreased glutamate receptor activity, <strong>and</strong> active neuralprotection by endogenous neural inhibitors, which occursduring the first few hours after the insult (the ‘latent’phase). I will briefly discuss whether treatments likehypothermia can reduce acute neuronal <strong>and</strong> glial injury inthe preterm, <strong>and</strong> thus potentially improve the chronicdevelopment of the brain.NotesPage 44


Geoffrey Dawes LectureAvian influenza <strong>and</strong> p<strong>and</strong>emic threatHitoshi OshitaniDepartment of Virology, Tohoku University, Graduate School of MedicineSince December 2003, avian influenza A(H5N1) has swept through poultry populationsacross Asia. It has now spread to Europe, Middle-East <strong>and</strong> Africa. The outbreaks are historicallyunprecedented in scale <strong>and</strong> geographical spread. Their economic impact on the agricultural sectorof the affected countries has been enormous. Evidence shows that the H5N1 virus is most likelyendemic in many parts of Asia. It has established an ecological niche in poultry, making it extremelydifficult to prevent outbreaks. Outbreaks have recurred despite aggressive control measures,including the culling of more than 160 million poultry. During this period, more than 300 humancases with an overall fatality rate around 60%, have been confirmed in 12 countries. Most of humaninfections can be linked to contact with infected poultry, but isolated instances of inefficienthuman-to-human transmission may have occurred.When the virus acquires capability of efficient human-to-human transmission, globalinfluenza p<strong>and</strong>emic in human population can occur. It is expected that the next p<strong>and</strong>emic wouldresult in huge impact on human health, economy <strong>and</strong> whole society of the world. Influenza virusesare genetically unstable <strong>and</strong> their behavior cannot be predicted. For these reasons, the currentoutbreaks of influenza A(H5N1) in poultry <strong>and</strong> humans has moved the world closer to a p<strong>and</strong>emicthan any time since 1968, when the last such event occurred.Immediate actions should be taken by all relevant partners to minimize the risk of nextp<strong>and</strong>emic by implementing various control measures in poultry <strong>and</strong> human. It is necessary to beprepared for emergence of a virus with p<strong>and</strong>emic potential. If early signs of potential p<strong>and</strong>emic aredetected rapidly, early containment of the virus may be possible by implementing aggressivemeasures including mass drug administration in affected areas. All countries should also beprepared for the worst case scenario, i.e. influenza p<strong>and</strong>emic by improving their p<strong>and</strong>emicpreparedness. By implementing various measures including the use of vaccines <strong>and</strong> antivirals <strong>and</strong>other public health interventions, the impact of a next p<strong>and</strong>emic can be minimized.Page 46


Abstractinvited lectureRole of PEG10 in mammalian placenta development <strong>and</strong>evolutionFumitoshi Ishino 1 , Ryuichi Ono 1 , Sunsuke Suzuki 1 <strong>and</strong> TomokoKaneko-Ishino 21 Department of Epigenetics, Medical Research Institute, Tokyo Medical <strong>and</strong> Dental University.2 Tokai University, School of Health Sciences.Eutherian chorioallantoic placenta is an evolutionary neworgan that enables fetal growth <strong>and</strong> development inmother for a relatively long pregnancy period. Manygenes are known to be involved in placenta formation,however, it is still unknown how this new organ evolved inthe eutherians. PEG10 (paternally expressed 10) is animprinted gene that is derived from a sushi-ichi relatedretrotransposon that is highly conserved in eutherianmammals but not in birds <strong>and</strong> fish. We have recentlydemonstrated that lack of Peg10 causes early embryoniclethality in mice due to sever placental defects lackingspongiotrophoblast <strong>and</strong> labyrinth layers. Thus, Peg10plays an essential role in mouse development in spite ofits exogenous origin.Comparative genome study among three mammaliangroups, monotremes, marsupials <strong>and</strong> eutherians, hasdemonstrated that PEG10 also exists in wallaby (amarsupial species), but not in platypus (a monotremespecies). Interestingly, marsupial PEG10 locates exactlythe same position to that of eutherian PEG10, betweenSGCE <strong>and</strong> PPP1R9A genes, <strong>and</strong> its amino acid sequenceis conserved. These results indicate that integration ofthe original PEG10 retrotransposon occurred in one of thecommon ancestors of marsupials <strong>and</strong> eutherians, <strong>and</strong>transformed to the present PEG10 form presumably bymultiple mutations before divergence of these two groups.Conservation of PEG10 means that it has high selectiveadvantage in mammalian development under naturalselection, therefore, the event of PEG10 acquisition maybe one of the driving forces in mammalian placentalevolution.NotesPage 48


invited lectureAbstractCell Sheet Engineering for Tissue <strong>and</strong> Organ RegenerationTeruo OkanoInstitute of Advanced Biomedical Engineering <strong>and</strong> Science Tokyo Women’s Medical UniversityIdeal tissue construction in vitro <strong>and</strong> in vivo can beachieved by cell sheet engineering that has beenestablished with three core technologies: First,noninvasive harvest of cultured cells as cell sheets can beachieved using previously developed temperatureresponsiveculture dishes. The surfaces are hydrophobicat 37°C, but become hydrophilic below 32°C. Therefore,various cell types adhere, spread, <strong>and</strong> proliferate on thesurfaces similarly to on commercial tissue culture dishes.By temperature reduction, cells spontaneously detachfrom the surfaces <strong>and</strong> confluent cells are recovered as asingle contiguous monolayer sheet with intact cell-celljunctions <strong>and</strong> deposited extracellular matrix. Second,the harvested viable cell sheets can be transferred toother surfaces, such as culture dishes or devices (2D cellsheet manipulation) because of the adhesive extracellularmatrix proteins associated with the basal side of cellsheets. Thus, tissue regeneration with cell sheetengineering can be accomplished by transplantation ofsingle cell sheets, as with skin, cornea <strong>and</strong> periodontalligaments. Finally, recovered cell sheets can be layered toreconstruct complex stratified tissue architectures such asliver lobules, kidney glomeruli, <strong>and</strong> cardiac patches (3Dcell sheet manipulation). For example, layeredcardiomyocyte sheets harvested from temperatureresponsive dishes show synchronous pulsations <strong>and</strong>diffuse gap junction formation. When transplanted into thesubcutaneous tissues of nude rats, spontaneous beatingscould be macroscopically observed <strong>and</strong> maintained forover 1 year. We believe that these 2D <strong>and</strong> 3D cellmanipulations using cell sheet tissue engineering, willbecome new <strong>and</strong> revolutionary tools for tissueengineering.NotesPage 49


NotesPage 50


Abstract1Keynote speakerImpact of low birthweight on developing organs: separating theeffects of IUGR <strong>and</strong> preterm birth.Richard Harding PhD, DSc.,Department of Anatomy <strong>and</strong> Cell Biology, Monash University, Melbourne 3800, AustraliaNumerous epidemiological studies have shownassociations between low birthweight (LBW) <strong>and</strong> adversehealth outcomes. However most such studies do notdistinguish between the effects of IUGR <strong>and</strong> preterm birth,both of which can cause LBW <strong>and</strong> alter org<strong>and</strong>evelopment. The brain, lungs, heart <strong>and</strong> kidneys, forexample, develop both before <strong>and</strong> after birth, <strong>and</strong>therefore could be affected by IUGR <strong>and</strong> preterm birth.The sheep is a good model as birth occurs at a similarstage of organ development as in humans.the effects of preterm birth on the development of theseorgans are relatively unexplored.As 8-10% of births are affected by IUGR, <strong>and</strong> similarnumbers occur prematurely, it is important that theseparate effects of IUGR <strong>and</strong> preterm birth on criticalorgan development are defined; appropriate animalmodels are essential. It is also important thatepidemiological studies distinguish between LBW due toIUGR <strong>and</strong> preterm birth.Using sheep, we have investigated the separate effects ofIUGR <strong>and</strong> preterm birth on lung development <strong>and</strong> lungstructure through to maturity. LBW induced by IUGRduring late gestation impairs alveolar formation, resultingin reduced alveolar number, thicker alveolar walls <strong>and</strong>reduced airway support in adult animals 1,2 ; airway wallstructure was not affected 3 . In contrast, LBW due tomoderately preterm birth has no persistent effects onalveoli 4 , but adult airways were more reactive 5 . Our datasuggest that IUGR predominantly affects lungparenchyma whereas preterm birth primarily affects theairways. In recent studies we have also shown how IUGRaffects kidney 6 , brain 7,8 <strong>and</strong> heart 9 development; however1. Maritz et al. Pediatric Pulmonology 32: 201-210; 2001.2. Maritz et al. Pediatric Research 55: 287-295; 2004.3. Wignarajah et al. Pediatric Research 51: 681-688;2002.4. Cock et al. Pediatric Pulmonology 40: 336-348; 2005.5. Snibson et al. Experimental Lung Research, 32:215-228; 20066. Mitchell et al. Pediatric Research 55: 769-773; 2004.7. Rees & Harding. Neuroscience Letters 361: 111-114;2004.8. Duncan et al. J Neuropath & Exp Neurol 63, 1131-43;2004.9. Bubb et al. J Physiology 578: 871-881; <strong>2007</strong>.NotesPage 52


Postnatal high-fat feeding impacts on metabolic profiles <strong>and</strong>gluconeogenic capacity during fasting <strong>and</strong> this is exaggeratedby prenatal undernutrition in adolescent lambs.Mette Olaf Nielsen & Anna Hauntoft KongstedDepartment of Basic Animal <strong>and</strong> Veterinary Sciences, The Faculty of Life Sciences,Copenhagen University, Grønnegårdsvej 7, DK-1870 Frederiksberg C, Denmark.Abstract2Liver function <strong>and</strong> metabolic profiles during fasting <strong>and</strong> agluconeogenic (propionate) challenge was studied inlambs exposed to different prenatal <strong>and</strong> postnatalnutritional levels <strong>and</strong> diets. For the last 6 weeks prepartum20 twin-pregnant Shropshire ewes were assigned to eithera High (ME <strong>and</strong> protein according to requirements) orLow (50% of High) diet. Three days post-partum the twinlambs were assigned to each their feeding: CONV (milkreplacer <strong>and</strong> dried hay to achieve 250g weight gain/d) orHCHF (High-Fat-High-Carbohydrate: 38% cream + milkreplacer + maize ad libitum). When approx. 6 months oldthe lambs were fasted for 47 hours, subjected to apropionate challenge between hours 45-46 <strong>and</strong> refed athour 47.From hours 0-44 plasma glucose levels were similar in allgroups. During the propionate challenge HCHF lambs hadslightly slower glucose production <strong>and</strong> cleared glucoseslower than CONV. HCHF lambs also had fatty livers <strong>and</strong>higher plasma gamma glutamyl transferase (GGT) levels.For the prenatal Low diet the same trend was seen as forHCHF, but less pronounced. Low/HCHF lambs thustended to have the slowest, while High/CONV had thefastest glucose clearance after the propionate challenge.Plasma triglyceride levels declined markedly in HCHFlambs over the first 16 hours of fasting upon cessation ofcream feeding, but triglyceride as well as NEFA levelsremained consistently higher in HCHF compared toCONV lambs throughout fasting <strong>and</strong> propionate challenge.NEFA gradually increased in all groups during fasting,rapidly declined after the propionate-injection, <strong>and</strong>increased again 30-60 minutes after, ie. the reversepattern of that observed for glucose.Plasma BOHB levels were initially similar in all groups.After 16 hours of fasting, a marked <strong>and</strong> steady increasewas observed in HCHF lambs, in comparison with adelayed <strong>and</strong> very modest increase in CONV lambs. TheBOHB increases during fasting were further exaggeratedin Low lambs.In conclusion, HCHF lambs showed indications ofreduced liver (gluconeogenic) function, impairedglucose-clearance <strong>and</strong> a more ketogenic metabolismduring fasting. This appeared to be exaggerated in lambsexposed to prenatal undernutrition (Low), ie. as aconsequence of a pre- <strong>and</strong> postnatal nutrition mismatch.NotesPage 53


Abstract3Maternal Isocaloric High-protein Diet Ameliorates the Elevation inboth Blood Pressure <strong>and</strong> Cardiac Remodeling in the Adult MiceOffspring with Maternal Calorie Restriction Possible Involvement ofBranched-Chain Amino Acids–Hiroaki Itoh 1 , Makoto Kawamura 2 , Shigeo Yura 2 , Haruta Mogami 2 , FujiiTsuyoshi 2 , Norimasa Sagawa 31 Osaka National Hospital, Osaka; 2 Departmemt of Gynecology <strong>and</strong> Obstetrics Kyoto UniversityGraduate School of Medicine, Kyoto, 3 Department of Obstetrics <strong>and</strong> Gynecology Mie UniversitySchool of Medicine, Tsu, <strong>Japan</strong>Background;Epidemiological evidences have elucidated thatundernutrition in utero is a risk factor for cardiovasculardisorders (CVD) in adulthood. However, the mechanismis yet be fully understood. Recently, we demonstrated amouse animal model of intra uterine growth restriction(18% decrease in pup weight) with maternal calorierestriction (30%) using regular chow diet (RCD) (CellMetab 2005, 1;371). After birth, the undernourishedoffspring (UN offspring) showed a significant increase insystolic blood pressure <strong>and</strong> cardiac remodeling at 16 wks,compared to normally nourished offspring (NN offspring)(Endocrinology <strong>2007</strong>,148:1218).Aim;The aim of the present study was to investigate the effectof maternal high protein intake, isocaloric base, on thedevelopment of risk factors of CVD in adult offspring.Methods;The maternal food restriction was applied to pregnant micewith either RCD or isocaloric high-protein diet (HPD).Amino analysis was carried out in the fetal plasma, whichwas obtained by uterine section at 18.5 dpc. Systolic bloodpressure <strong>and</strong> cardiac remodeling-associated morphologicalparameters, i.e. perivascular fibrosis of the coronary artery,cardiomegaly <strong>and</strong> cardiomyocyte enlargement, weremeasured in the adult offspring (P-UN offspring) incomparison with UN <strong>and</strong> NN offspring.Results;Maternal isocaloric HPD augmented fetal plasmaconcentrations of branched-chain amino acids (BCAA:valine, leucine <strong>and</strong> isoleucine). At 8 <strong>and</strong> 16 wks,significant systolic blood pressure increase was observedin UN offspring, but not in P-UN offspring, as comparedwith NN offspring. In the similar manner, all of cardiacremodeling-associated parameters measured weresignificantly elevated in UN offspring, but not in P-UNoffspring, as compared with NN offspring at 16 wks.Conclusion;It is suggested that maternal isocaloric HPD may alleviatethe deterioration in systolic blood pressure as well as incardiac remodeling in the adult offspring with maternalcalorie restriction, probably at least partly via exposure toBCAA in utero.NotesPage 54


AbstractMaternal ovine cortisol concentrations are important for keyendocrine factors involved in fetal growth4Jensen EC 1 , Bennet L 1 , Wood CE 2 , Gunn AJ 1 <strong>and</strong> Keller-Wood M 3 .1 Department of Physiology, Faculty of Medical <strong>and</strong> Health Sciences, The University ofAuckl<strong>and</strong>, Auckl<strong>and</strong>, New Zeal<strong>and</strong>, 2 The Department of Physiology <strong>and</strong> Functional Genomics<strong>and</strong> 3 The Department of Pharmacodynamics, College of Medicine, The University of Florida,Gainesville, Florida, USAThe normal elevation of maternal plasma corticosteroidconcentrations during pregnancy is important for thesupport of fetal growth <strong>and</strong> development. Both elevatedmaternal cortisol levels <strong>and</strong> reduced maternal cortisolbelow normal levels reduce fetal growth, <strong>and</strong> elevatedmaternal cortisol levels cause fetal hypertension. Themechanisms mediating these changes in fetal growthremain unknown. We examined the hypothesis thatchronic alterations in maternal plasma cortisolconcentrations reduce insulin-like growth factor (IGF)mRNAs <strong>and</strong> alter binding protein (BP) mRNA, <strong>and</strong> soreduce fetal growth.Ewes of mixed breeds carrying singleton pregnancies wereoperated on between 112 <strong>and</strong> 116 days of gestation (115 ±0.4, term=147 days). Animals were r<strong>and</strong>omly assigned intothree groups before surgery. The first group consisted of sixcontrol animals, the second group consisted of five ewesthat were administered cortisol by continuous intravenousinfusion (1 mg/kg/day) (high cortisol), <strong>and</strong> the thirdconsisted of five ewes that were adrenalectomized <strong>and</strong>replaced with 0.5-0.6 mg cortisol/kg/day <strong>and</strong> 3 µgaldosterone/kg/day in order to produce cortisolconcentrations similar to endogenous concentrations inintact nonpregnant animals (low cortisol). Fetuses werestudied from the day after surgery until 130 (± 0.2) days ofgestation. <strong>Fetal</strong> liver was snap frozen in liquid nitrogen<strong>and</strong> stored at -80 o C for further mRNA analysis.<strong>Fetal</strong> liver IGF-II <strong>and</strong> IGFBP-3 mRNA were decreased inthe low cortisol group compared to controls (p


Abstract5Diet-Induced Obesity <strong>and</strong> Prenatal Undernutrition Lead to CentralLeptin Resistance by Different MechanismsCharisma K Dhaliwal, Stefan Krechowec, Mark Vickers, Bernhard H Breier,Mhoyra FraserNational Research Centre for Growth <strong>and</strong> Development <strong>and</strong> Liggins Institute, Faculty of Medical <strong>and</strong>Health Sciences, The University of Auckl<strong>and</strong>, Auckl<strong>and</strong>, New Zeal<strong>and</strong>Background:We have previously reported that prenatal undernutritionleads to a dysregulation of appetite suppressive effectsthrough alterations in neuropeptide gene expression ofwhole hypothalami, which may be associated with central<strong>and</strong> peripheral leptin resistance [1]. In the current study,we exp<strong>and</strong> our initial observations <strong>and</strong> investigate theneuroendocrine transcriptional response <strong>and</strong> leptinsensitivity in the arcuate nucleus (ARC) of rats exposed toprenatal undernutrition (UN) or a postnatal high-fat diet(DIO).Methods:Pregnant Wistar rats were fed a st<strong>and</strong>ard chow diet eitherad libitum (AD) or at 30% of AD intake throughoutgestation (UN). At weaning, female offspring were fedeither a chow [C], high fat [HF] (30% fat wt/wt) or calorierestricted [CR] (70% of st<strong>and</strong>ard chow intake) diet adlibitum for the remainder of the study. At 142 ± 5 days, AD<strong>and</strong> UN offspring received either recombinant rat leptin(2.5µg/g/day) or saline for 14 days, subcutaneously [2].Results:Using quantitative reverse transcription-polymerase chainreaction (qRT-PCR), we found that both NPY <strong>and</strong> ObRbmRNA expression in the ARC was significantly increasedin UN animals (P


Impact of maternal exposure to constant light during lategestation on postnatal adrenal function in the capuchinmonkey: long terms effects on DHAS <strong>and</strong> transient effects oncortisol.Abstract61 Torres-Farfan C, 1 Mendéz N, 2 Richter H, 3 Valenzuela G <strong>and</strong> 1,4 Seron-Ferre M.ICBM, Fac Medicina, Universidad de Chile; 2 Fac Medicina, Universidad Austral de Chile, 3 Dpt ofWomen’s Health, Arrowhead Regional Medical Center, Colton, CA. 4 Universidad de Tarapaca<strong>and</strong> CIHDE, Arica, Chile.Several adult diseases are linked with alterations of thehypothalamus-pituitary-adrenal axis in utero. Maternalmelatonin suppression (by exposure to constant lightduring last third of gestation) induced precociousmaturation of the fetal adrenal (JPineal Res 41:58-66,2006), increased plasma cortisol concentrations in thenewborn soon after birth (JPhysiol 554:841-856, 2004);<strong>and</strong> resulted in an increased cortisol response to ACTH atfirst month of postnatal age (2006 <strong>FNPS</strong> P:4). Incontrast, at one month of age, basal DHAS <strong>and</strong> the DHASresponse to ACTH were decreased. To assess whetherthe effects of maternal exposure to constant light on thenewborn adrenal were permanent, we measuredresponsiveness to ACTH at 8 months of ageProtocols:Four pregnant capuchin monkeys were maintained inconstant light (LL) from 63% gestation <strong>and</strong> other fourremaining in light:dark cycle (14:10; Control). Afterdelivery the LL newborn <strong>and</strong> its mother returned to 14:10photoperiod. At 8 month postpartum the newbornsreceived dexamethasone (2.5mg/kg, im) <strong>and</strong> twelve hourslater, blood samples were drawn at 0-30-60 <strong>and</strong> 120 minpost ACTH 1-24 (125 μg/kg, iv). Cortisol <strong>and</strong> DHAS wasmeasured by RIA.Results <strong>and</strong> conclusion:Maternal exposure to constant light during gestation hadlong lasting effects on DHAS <strong>and</strong> transient effects oncortisol. At 8 months of age, the decrease in DHAS <strong>and</strong> inthe DHAS response to ACTH was maintained, whereascortisol response to ACTH returned to normal. Thepresent data suggest that exposure to constant lightduring pregnancy selectively impairs postnatal DHASproduction, probably by decreasing the number of DHASproducing cells. These results are consistent with thepreviously suggested role of maternal melatonin duringgestation as trophic factor for the DHAS producing cells ofthe fetal adrenal gl<strong>and</strong>.Supported-Fondecyt1050833-SBMC.NotesPage 57


Abstract7 Maternal feeding controls fetal biological clockXu Shanhai 2 , Hidenobu Ohta 1,2,3 , Takahiro Moriya 4 , Masayuki Iigo 5 , TatsuyaWatanabe 1,3 , Norimichi Nakahata 4 , Hiroshi Chisaka 1,2 , Tadashi Matsuda 1,3 ,Toshihiro Ohura 3 , Yoshitaka Kimura 6 , Nobuo Yaegashi 2 , ShigeruTsuchiya 3 , Hajime Tei 7 , <strong>and</strong> Kunihiro Okamura 1,21 Center for Perinatal Medicine, Tohoku University Hospital, Sendai, <strong>Japan</strong>, 2 Department ofObstetrics <strong>and</strong> Gynecology, Tohoku University Hospital, Sendai, <strong>Japan</strong>, 3 Department ofPediatrics, Tohoku University Hospital, Sendai, <strong>Japan</strong>, 4 Department of Cellular Signaling,Tohoku University, Sendai, <strong>Japan</strong>, 5 Department of Applied Biochemistry, Faculty of Agriculture,Utsunomiya University, Utsunomiya, <strong>Japan</strong>, 6 Tohoku University Biomedical EngineeringResearch Organization, Sendai, 980-8574, <strong>Japan</strong> , 7 Research Group of Chronogenomics,Mitsubishi Kagaku Institute of Life Sciences, Tokyo, <strong>Japan</strong>Circadian physiological rhythms of the fetus are affectedby oscillators in the maternal brain that are coupled to theenvironmental light cycle. Circadian gene expressionsoccur in the fetal brain <strong>and</strong> organs during pregnancyalthough it is unclear how the fetal biological clock iscoupled to the maternal clock. To study the link betweenfetal <strong>and</strong> maternal biological clocks, we investigated theeffects of cycles of food availability (which exert powerfulentraining effects on maternal behavior) on the rhythms ofgene expression in the fetal suprachiasmatic nucleus(SCN). We exposed pregnant rats to a restricted-feeding(RF) regimen, in which food was available only for 4 hoursduring the light portion of a 12-hour:12-hour LD cycle.With a transgenic rat model in which the mouse Period1(a circadian clock gene) promoter has been linked to aluciferase reporter, we continuously monitored therhythmic expression of this "clock gene" by recording lightemission from tissues in vitro. While rhythmicity in thematernal SCN remained phase-locked to the light-darkcycle, restricted feeding entrained the fetal SCN, shiftingits rhythm by 5 hours within the 22-day pregnancy (ad libfed group, n=5; RF fed group, n=5). Our resultsdemonstrate that feeding cycles can entrain the fetal SCNindependently of both maternal SCN <strong>and</strong> light cycle, <strong>and</strong>the results also suggest the need to reexamine thematernal-fetal communication mechanism..NotesPage 58


AbstractDietary protein restriction of pregnant mouse induces alteredmethylation of Oct-4 <strong>and</strong> Sphk-1 gene promoters in the liver ofoffspring8Jun Murotsuki 1 , Kunihiro Okamura 1 , Hiroi Kaku 2 , Yoshihiko Araki 3 , HiroshiYoshitake 3 , Mark A. Hanson 4 , Felino R. Cagampang 4Department of Obstetrics <strong>and</strong> Gynecology, Tohoku University School of Medicine 1 , <strong>and</strong>Department of Obstetrics <strong>and</strong> Gynecology, Iwate Medical University School of Medicine 2 ,Institute for Environmental <strong>and</strong> Gender-Specific Medicine, Juntendo University GraduateSchool of Medicine 3 , Centre for Developmental Origins of Health <strong>and</strong> Disease, University ofSouthampton 4The developmental origins hypothesis postulates thatduring critical ontogenetic periods, transientenvironmental stimuli perturb developmental pathways<strong>and</strong> induce permanent changes in gene expression,metabolism, <strong>and</strong> chronic disease susceptibility. Onelikely mechanism is via early nutritional influences onepigenetic gene modification consisting of the presence ofa methyl group on the carbon 5 of a cytosine residue.This modification is responsible for an important form ofgene regulation in eukaryotes. In the present study, wehave tested the hypothesis that maternal low-protein dietaltered epigenetic regulation of specific gene of theoffspring. C57BL/6 female mice were mated <strong>and</strong> on theday the plug was detected, these females were thenr<strong>and</strong>omly allocated to be fed isocaloric diets consisting18% protein or 9% protein. At delivery, offspring werekilled <strong>and</strong> the livers were removed immediately, frozen inliquid nitrogen <strong>and</strong> stored at -80C. Genomic sequencingafter bisulfite modification is used to study site-specificDNA methylation. DNA methylation status of Oct-4 <strong>and</strong>Sphk-1 gene upstream regions in the mouse liver wasanalyzed. Hepatic Oct-4 or Sphk-1 promoter methylationwas not significantly different between both groups.However, DNA methylation pattern of the genomic DNA isspecific in low-protein diet group. Aberrant Oct-4 <strong>and</strong>Sphk-1 gene expression may cause perturbations in celldifferentiation. We suggest that the epigeneticmechanism consisting of DNA methylation underlies thefetal programming theory.NotesPage 59


Abstract9Transcriptional suppression of Igf2 mediated by altered histonemodifications in a maternal protein restricted IUGR mouse modelNakamura M 1 , Sharif J 1,2 , Ito T 3 , Chisaka H 1 , Kimura Y 3 , Mitsuya K 3 , <strong>and</strong>Okamura K 11 Department of Obstetrics <strong>and</strong> Gynecology, Tohoku University Graduate School of Medicine;2 Department of Chemistry <strong>and</strong> Biotechnology, School of Engineering, The University of Tokyo;3 Tohoku University Biomedical Engineering Research OrganizationAIMSDietary restriction during pregnancy may provide cues formetabolic disorders such as hypertension <strong>and</strong> glucoseintolerance. Although prenatal programming can beexplained by certain epigenetic modifications, the precisemechanisms remain obscure. We have investigated theeffect of maternal nutrition on the fetus.MethodsMice were fed a control (C) diet or a protein restricted(PR) diet from 7 days prior to conception to throughoutpregnancy. Another two groups of mice were prepared byreversing the diet of the C <strong>and</strong> PR at mid-gestation (d10)to measure the effects of nutrition at early <strong>and</strong> lategestational stages. Dams were sacrificed at d17 <strong>and</strong> fetal<strong>and</strong> placental weight was measured. Expression ofpromoter specific Igf2 mRNA in fetal liver was analysed byquantitative real-time RT-PCR. Histone modifications inrespective promoter regions of Igf2 (P1-P3) was analysedto elucidate the epigenetic mechanism behind theobserved phenotypes.RESULTSProtein restricted (PR) diet throughout pregnancy did notaffect placental <strong>and</strong> fetal weight. Interestingly,administration of PR diet during late gestation (d10-d17)caused significant reduction of fetal weight, a hallmark ofintrauterine growth restriction (IUGR). In this group, theexpression of P1 promoter specific Igf2 expression wasalso reduced by approximately 50%. However, the P2 <strong>and</strong>P3 transcripts did not exhibit any notable change in Igf2expression. ChIP analyses revealed that the P1 promoterregion was depleted of H3K4me3 <strong>and</strong> H4K20me3permissive modifications <strong>and</strong> enriched in H3K27me3repressive modification. These histone modifications wereunaltered in the P2 <strong>and</strong> P3 promoter regions.CONCLUSIONSWe report a novel epigenetic regulatory mechanism thatrepresses promoter specific Igf2 expression in fetal liverduring maternal protein restriction. Our results suggestthat the fetus is capable of adapting to an environmentalcue if it is provided from the onset of gestation. But if theexternal insult is applied during a critical period ofdevelopment, a mismatch between the expected <strong>and</strong>experienced environment occurs which affects the fetalability to maintain its natural developmental program. Ourworks will provide new insights into the epigenetic <strong>and</strong>molecular basis of fetal programming <strong>and</strong> the response ofthe fetus to the early nutritional environment.NotesPage 60


AbstractKeynote speaker10 The state of the art in head cooling for neonatal encephalopathyAlistair Jan Gunn<strong>Fetal</strong> Physiology <strong>and</strong> Neuroscience Group, Dept of Physiology, The University of Auckl<strong>and</strong>,Auckl<strong>and</strong>, New Zeal<strong>and</strong>.The possibility that hypothermia started during or afterresuscitation at birth might reduce brain damage <strong>and</strong>cerebral palsy has tantalized clinicians for a long time.The key insight was that transient severehypoxia-ischemia can precipitate a complex biochemicalcascade leading to delayed neuronal loss. There is nowstrong experimental <strong>and</strong> clinical evidence that mild tomoderate cooling can interrupt this cascade, <strong>and</strong> improvenumbers of infants surviving without disability in themedium-term. The key remaining issues are to find betterways of identifying babies who are most likely to benefit,to define the optimal mode <strong>and</strong> conditions of hypothermia<strong>and</strong> to further improve the effectiveness of treatment.Existing trial data suggests a number of possible avenues.These trials have confirmed that outcome is stronglyinfluenced by severity of neonatal encephalopathy. In theCoolCap study, infants with the most severeelectrographic changes did not seem to respond tohypothermia. In contrast, there was a similar relativebenefit for hypothermia in infants with clinically definedmoderate <strong>and</strong> severe encephalopathy, raising thepossibility that aEEG monitoring may be more effective inidentifying infants at a treatable time after birth.Intriguingly four days after rewarming infants withcontinuing moderate encephalopathy who were treatedwith hypothermia had a significantly higher rate offavorable outcome than controls, suggesting thathypothermia may change the prognostic value of clinicalassessment. Further, the CoolCap trial has suggested forthe first time that greater birth weight in control infants washighly associated with adverse outcome, even afteradjustment for pyrexia <strong>and</strong> severity of encephalopathy,however, larger infants also showed greater benefit fromcooling. Thus, potentially the biology of neonatal injurymay differ between larger <strong>and</strong> smaller infants.NotesPage 62


AbstractCerebral oxygenation measured by spatially resolved spectroscopyin newborn lambs11Wong F 1,2 , Theodora Alexiou 1 , Brodecky V 1 , Samarasinghe T 1 , WilkinsonM 1 , Walker AM 11Ritchie Centre for Baby Health Research, Monash University, Clayton, Victoria, Australia;2Newborn Services, Monash Medical Centre, Clayton, Victoria, AustraliaBackground:Loss of cerebral autoregulation in preterm infants hasbeen associated with adverse outcome 1 . Howeverassessment of cerebral autoregulation is problematic dueto difficulty with continuous measurement of cerebralblood flow (CBF). Spatially Resolved Spectroscopy (SRS)enables continuous cotside measurement of meancerebral oxygen saturation, expressed as a tissueoxygenation index (TOI). Theoretically, if cerebral oxygenconsumption is constant, changes in TOI reflect changesin CBF. As yet, the CBF-TOI relationship has not beenestablished experimentally. We aimed to evaluate therelationship between simultaneous measurements of CBF<strong>and</strong> TOI in the newborn lamb brain, as a suitable animalmodel for the preterm human brain.Method:In four newborn lambs (2-6 d of age), TOI was measuredcontinuously with SRS (Hamamatsu NIRO-200). CBF wasmeasured with an ultrasonic flow probe (TransonicTM)implanted around the superior sagittal sinus. Mean arterialpressure (MAP) was recorded via an axillary intra-arterialcatheter <strong>and</strong> pressure transducer. An inflatable siliconrubber cuff was positioned around the commonbrachiocephalic artery; inflation of this cuff reduced arterialpressure distal to the cuff <strong>and</strong> hence cerebral perfusionpressure. Relationships between changes in CBF <strong>and</strong> TOIwere evaluated in the time <strong>and</strong> frequency domain.Coherence analysis was performed to quantify correlation ofCBF <strong>and</strong> TOI in a frequency-specific manner; a coherencescore ≥ 0.5 indicates a significant relationship between thewaveforms of CBF <strong>and</strong> TOI in a given frequency rangeResults:CBF <strong>and</strong> TOI showed concordant changes during MAPfluctuations (Fig.1). In time domain analysis, there issignificant correlation between ∆TOI(%) <strong>and</strong> ∆CBF(%)TOI(%)CBF(ml/min)MAP(mmHg)6050400 2 4 6 8 1010500 2 4 6 8 10806040200 2 4 6 8 10Time (min)Figure 1. TOI <strong>and</strong> CBF during MAP fluctuations(R 2 =0.53, p


Abstract12Treatment of fetal lung hypoplasia with antenatal corticosteroids:effects on pulmonary circulation <strong>and</strong> respiration in the newbornsheep*Suzuki K, Hooper SB, Harding R*Center for Maternal, <strong>Fetal</strong>, <strong>and</strong> <strong>Neonatal</strong> Medicine, Saitama Medical Center, Saitama MecicalUniversity, Kawagoe, Saitama, <strong>Japan</strong>Department of Physiology, Monash University, Melbourne, Victoria, AustraliaAims:We have previously shown that fetal lung hypoplasia (LH)has more influence on the pulmonary circulation than onventilation in newborn lambs (Suzuki K et al; Pediatr Res57:530-6, 2005). The objective of this study was todetermine the effects of antenatal maternal corticosteroidadministration on the pulmonary circulation <strong>and</strong>respiration in a sheep model of LH.Methods:LH was induced in 12 ovine fetuses by tracheo-amnioticshunt <strong>and</strong> amniotic fluid drainage beginning at ~105 daysof gestation (term ~147days). In 6 of these fetuses(LH+S group), one dose of betamethasone (11.4 mg im)was given to the mother 24 hours before elective delivery.At ~140 days, after vascular catheterizations <strong>and</strong>ultrasonic flow probe implantation, lambs were delivered<strong>and</strong> ventilated for 2 hours under sedation, during whichwe recorded systemic <strong>and</strong> pulmonary artery pressures,left pulmonary artery blood flow, airway pressure <strong>and</strong>ventilatory air-flow.Results:Control LH LH+S(n=6) (n=6) (n=6)Lung weight (g/kg BW) 29.8+/-2.7 *21.7+/-1.3 **19.6+/-2.1Lung DNA (mg/kg BW) 186+/-13 *145+/-7 **132+/-11FiO 2 at 2h 0.38+/-0.03 0.35+/-0.02 0.33+/-0.01Crs at 2h0.57+/-0.04 *0.40+/-0.03 **0.41+/-0.03(mL/cmH 2 O/kg BW)PVR at 2h(mmHg/mL/min*kg BW) 0.32+/-0.04 **0.76+/-0.06 ## 0.44+/-0.04Crs: total respiratory system compliancePVR: pulmonary vascular resistancedifference between control <strong>and</strong> LH (*p


Clinical presentation, causes <strong>and</strong> outcome of PersistentPulmonary Hypertension of Newborns (PPHN) in a tertiary carehospital of Karachi.Abstract13Ali Samana, Parkash Jai, Feroze Asher, Atiq Mehnaz, Khan IqtidarDepartment of Pediatrics. Aga Khan University Karachi, PakistanIntroduction:PPHN is a cardiopulmonary disorder characterized bysystemic arterial hypoxemia secondary to elevatedpulmonary vascular resistance with resultant shunting ofpulmonary blood flow to the systemic circulation.Objectives:All studies related to PPHN available in literature are ofdeveloped world where advanced treatment is easilyavailable. There is no data found to see the clinical course<strong>and</strong> outcome of PPHN in developing world. Our objectivewas to see clinical presentation, common causes <strong>and</strong>outcome of PPHN in a tertiary care hospital of Pakistan.Study Design:A retrospective descriptive study is conducted in <strong>Neonatal</strong>intensive care unit of Aga Khan Hospital from July 2000 toJuly 2005.All babies < 30 days of life with respiratory distress wereincluded who were diagnosed as PPHN by either one ofthe following laboratory results:• Echocardiography showing Pulmonary ArterialHypertension.• Failed Hyperoxia test.• Significant difference of pre <strong>and</strong> Post ductal bloodgases (> 20 mmHg).All neonates with cyanotic heart disease or in the absenceof above mentioned inclusion criteria were excluded.Results:Total 8o charts were pulled through ICD system of medicalrecord. Out of which 16 cases were excluded due topresence of cyanotic structural heart diseases <strong>and</strong>missing charts. 64 cases were included in our study. 67%patients were presented before 24 hours of life. 72%patients were male. 57% patients were AGA. 90% patientsrequired mechanical ventilation. 70% patients requiredventilation for 6 days. Pneumonia was the most commoncause (42%) found. Sepsis <strong>and</strong> Meconium aspirationsyndrome were the second <strong>and</strong> third common causes ofPPHN. Hypothermia found in 25% cases. Hypoglycemiafound in 14% cases. Blood Culture was +ve in 26%cases.25% developed Pneumothorax. 50% patientsexpired while 45% survived <strong>and</strong> remaining were shifted toother hospital. Patients presenting


Abstract14 Disease spectrum of term neonates admitting in <strong>Neonatal</strong> ICUAli Samana, Salat SohailDepartment of Pediatrics, Aga Khan University, Karachi, PakistanIntroduction:Newborn babies born after 36 weeks of gestation areconsidered as term neonates. In comparison to pretermbabies, term babies have a smooth course <strong>and</strong> do notrequire intensive care admission. Some of them still needintensive care admission.In developed countries the common problems of newbornbabies admitted in NICU are infections, prematurity, birthasphyxia, <strong>and</strong> congenital anomalies. We don’t know thedisease spectrum of neonates admitting in NICU ofdeveloping country. Multiple studies have beenpublished to know the disease spectrum <strong>and</strong> outcome ofpreterm babies but no significant data found for termbabies both with in <strong>and</strong> out side our country.ObjectivesOur objectives were:• To know the type of diseases acquired by term babiesrequiring intensive care hospitalization.• To assess the outcome of full term babies in terms ofrecovery/discharges, expiries or shifts to otherhospitals.MethodologySetting:• This study is done in neonatal intensive care unit(NICU) of the Aga Khan University Hospital Karachi(AKUH) from Jan 2005 to Dec 2005.Sampling Technique:• It was purposive sampling. All term neonates admittedin NICU during study period will be included.Inclusion Criteria:• All term (>36 weeks) neonates (0 to 28th day of life)born inside or outside AKUH born admitted in NICUwere included in study.Exclusion Criteria:• All preterm neonates


AbstractPrenatal Diagnosis of Schizencephaly with Septo-opticDysplasia by Ultrasound <strong>and</strong> Magnetic Resonance Image15Jeng Hsiu HungDepartment of Obstetrics <strong>and</strong> Gynecology, Taipei Veterans General Hospital, 201, Section 2,Shih-Pai Rd, Taipei 112, TaiwanAbstractsA 28-year-old gravida, G1 A0, menarche at 12 years old,was referred to our fetal diagnosis unit at 28 +2 gestationalweeks because of suspected fetal cerebral anomalies. Ontwo-dimensional ultrasonography, the cephalic axial viewshowed multiple hypoechoic spaces in the fetal brain,both cerebral cortex <strong>and</strong> occipital lobe showed bilateraldefects, <strong>and</strong> the septum pellucidum was absent. Multipleirregularly shaped cystic lesions connected withsubarachnoid spaces were observed by three-dimensionalultrasonography in surface rendering mode. Septo-opticdysplasia with dysgenesis of corpus callosum was furtherconfirmed by prenatal MRI. Since schizencephaly withbilateral clefts involve bilateral frontal, parietal <strong>and</strong>occipital lobes, learning disability, epilepsy <strong>and</strong> cerebralpalsy are often present after birth. The flaccid mobility ofall four extremities of the fetus, demonstrated prenatallyby real-time ultrasound <strong>and</strong> functional MRI, can forecastthe risk of postnatal spastic quadriplegia.NotesPage 67


NotesPage 68


Abstract16A possibility of applying system-engineering methods toamniotic fluid volume changes.Yoshiyasu Hombo, Michio OoshitaKanazawa, <strong>Japan</strong>Amniotic fluid volume (AFV) during pregnancy is usuallysurveyed using amniotic fluid index (AFI). However, thisone-dimensional index is unable to predict the actual AFV.We then assumed each pocket’s shape as a half-ellipsoidor as a crescentric-cylinder <strong>and</strong> calculated its volume (ml)using distances measured along three axes. We reportedelsewhere that the sum of all volumes of the pockets wasa good predictor of AFV (pAFV) in near-tem pregnancy.We have extended the use of the pAFV to the secondtrimester <strong>and</strong> obtained 15005 measurements of pAFVfrom 12 to 42 weeks in 1352 pregnancies. The mean ofpAFV increased at the rate of 40-55ml/week from 12 to 22weeks, reached a peak of 590ml in 27 weeks <strong>and</strong>decreased at the rate of 20-30ml/week after 31 weeks.The regression equation of pAFV by weeks was:pAFV=0.0033W 4 -0.3633W 3 +12.702W 2 -137.71W +437.68(W; gestational weeks, R 2 =0.994).Other investigators have also presented a regressionequation for estimation of fetal body weight (EFBW) byweek. We added the pAFV to EFBW <strong>and</strong> obtained volumeof amniotic cavity (VOAC) for each week. The regressionequation was; VOAC=-0.0032W 4 +0.2177W 3 -1.8459W 2+22.675W-263.2(R 2 =0.9997), which showed that theamniotic cavity continued to exp<strong>and</strong> in a smooth manner.Therefore, the inner pressure of the amniotic fluid shouldpush outwards <strong>and</strong> stretch amniotic membrane.Investigators have shown that major sources of amnioticfluid are fetal urine <strong>and</strong> lung exudates <strong>and</strong> major removalroutes are fetal swallowing <strong>and</strong> the intra-membranouspathway.Taking these into consideration, we have derived twodifferential equations describing AFV <strong>and</strong> its osmolarity,which correspond to the state <strong>and</strong> output ones in field ofsystem engineering, respectively. This suggests thatAFV-changes during pregnancy can be analyzed usingsystem-engineering means.NotesPage 70


AbstractPregnancy after Stillbirth <strong>and</strong> Home <strong>Fetal</strong> Heart Rate Monitoringvia the Internet.17Jason H Collins MDObstetrics & Gynecology, Collins ClinicSlidell, LA, U.S.AStillbirth affects 30,000 pregnancies each year in theUnited States. Umbilical Cord Accident (UCA) accountsfor 20%-25% of all stillbirths or 2-4 deaths/1000 live births.Recent studies have shown pregnancy after stillbirth hasa recurrent stillbirth risk of 5-10 fold. The specific risk ofUCA recurrence is unknown. It is hypothesized that theuse of Home <strong>Fetal</strong> Heart Rate Monitoring can identify atrisk fetuses for UCA. Those fetuses with umbilical cordcompression patterns can be evaluated <strong>and</strong> impendingrecurrent stillbirth prevented.Aims:To prospectively follow 40 patients with a history of UCAstillbirth <strong>and</strong> their subsequent pregnancy by using homefetal heart rate monitoring daily via the internet. Todetermine the presence of umbilical cord compressionpatterns <strong>and</strong> evaluate the delivery record for recurrentUCA.Methods:Patients with a UCA stillbirth history contacted PregnancyInstitute (PI) through the internet web site(www.preginst.com). Patients were interviewed about theirpregnancy loss <strong>and</strong> apparent cause. Patients who decideto repeat pregnancy were offered an evaluation at 28-30weeks at PI. This visit consisted of a 30 min <strong>Fetal</strong> HeartRate Recording <strong>and</strong> an Ultrasound for UCA recurrence.Patients were instructed on home use of a hospital gradeFDA approved fetal monitor (Analogic:Boston, MA).Patients monitored for 30 minutes every night. Recordingswere sent via the internet to PI .The software wasdeveloped by Remote Care <strong>and</strong> is FDA approved <strong>and</strong>HIPPA compliant. Delivery findings were noted throughpatient interview.Results:29 out of 40 patients (study cases) had UCA recurrence at28-30weeks. 20 patients delivered with a recurrent UCAwith 1of these a recurrent UCA Stillbirth. All patientsdelivered by 37 weeks. 4 patients had expeditedC/Sections for FHR decelerations during labor.Conclusion: UCA can repeat in a subsequent pregnancyafter UCA related stillbirth. Home <strong>Fetal</strong> Heart RateMonitoring can detect umbilical cord compression <strong>and</strong>possibly offer an effective method of identifying the fetusat risk of repeat UCA stillbirth.NotesPage 71


Abstract18Prediction of fetal coarctation of the aorta from the three vessel<strong>and</strong> tracheal viewHikoro Matsui 1 , Lucia Pasquini 1 , Anna Seale 2 , Mats Mell<strong>and</strong>er 1,2 , MichaelRoughton 2 , Siew Yen Ho 2 , Helena Gardiner 1,21 Faculty of Medicine, Imperial College at Queen Charlotte's & Chelsea Hospital,2 Royal Brompton Hospital, London, UKBackground: Prenatal diagnosis of isolated coarctationof the aorta (CoA) suffers from high false positive <strong>and</strong>false negative rates. It is a duct-dependent lesion that isfatal if not treated early. Suspicion of isolated CoA atscreening relies on sonographic disproportion at fourchamber <strong>and</strong>/or great arterial views. We aimed to developZ-scores for the aortic isthmus <strong>and</strong> measure theisthmal:duct ratio (I:D) in normal fetuses <strong>and</strong> test thesescores in fetuses referred with suspected CoA. IsolatedCoA was defined as hearts with normal situs <strong>and</strong>connections, with or without a VSD <strong>and</strong> a narrowtransverse aortic arch or isthmus. Cases with bicuspidaortic valve were included.Methods: The aortic isthmal diameter, immediatelyproximal to the insertion of the arterial duct, wasmeasured prospectively in the transverse plane (threevessel <strong>and</strong> tracheal view) in 221 normal fetuses at 18 to37 weeks’ gestation. The ductal diameter (D) wasmeasured immediately before it entered the descendingaorta (DAo) in the same view. Four examiners contributedto the data collection. All measurements were repeatedthree times by a single investigator <strong>and</strong> averaged.Z-scores were created relating isthmal <strong>and</strong> ductaldiameters to femoral length <strong>and</strong> gestational age <strong>and</strong> theI:D ratio recorded.These scores were tested retrospectively on archiveddigital data in 200 fetuses we knew to have normaloutcomes <strong>and</strong> in 31 with suspected CoA. All hadpostnatal follow-up for at least one year. Thesemeasurements were examined by a single observer,blinded to outcome <strong>and</strong> ROC curves created.Results: Inter-observer mean difference of isthmalZ-scores was -0.04mm (95% confidence interval -0.8,0.71). The I:D ratio remained constant mean (SD) 0.99(0.13) 95% CI normal values (0.74, 1.23). ROC curvesshowed AUC Isthmus of 0.98 <strong>and</strong> I:D ratio 0.985. Serialisthmal Z-scores improved to > -2 in most normalneonates by term. (Figure)ConclusionsThe isthmal Z scores <strong>and</strong> I:D ratio distinguishes thosewith coarctation from fetuses with a normal aortic arch atfirst scan. In those with arch hypoplasia I:D ratio was abetter indicator of need for surgery.NotesPage 72


AbstractDevelopment of new digital fetal monitor “Behaviogram” forearly to mid pregnancy19Norio ShinozukaDepartment of Obstetrics <strong>and</strong> Gynecology, <strong>Japan</strong> Red Cross Medical CenterObjectivesChange in fetal behavior <strong>and</strong> heat rate reflects fetalcondition in utero. However , at early to mid trimesterpregnancy there has been a methodological limitation toobtain these information in quantitative terms. Tomonitor heart rate at beat to beat quality with fetalbehavioral parameter such as FBM, we devised newsystem named “Behaviogram”Method / HardwareBased the previous studies (Ref), the multi-transducer /multi-channel (7 Tr x 18ch ) Doppler system wasdeveloped. The whole transducer size is same as that ofan ordinal cardiotocogram. With this system, multi-pointtissue Doppler signals from fetus are measured. FHR iscalculated from Doppler digital phase signal using twodimensional correlation analysis. FBM is recorded fromminute tissue displacement signals. <strong>Fetal</strong> gross bodymovements (FGM) are expressed as the power ofDoppler shift signals.To examine the clinical feasibility <strong>and</strong> to optimize dataprocessing protpcol of devised system, the clinical data offetuses from pregnancy 15 to 37 weeks (15-19w:n=18,20-24w:n=16, 25-29w; n=23,35w-:n=13) were analyzed.Results Example of the display indicates FHR. FBM, FGMwas shown in Fig 1. FHR was measured with nearly beatto beat quality from fetus at 15w of gestation. From 20wonward, simultaneous FHR with behavior (FBM. FGM)were successfully recorded (Fg1).ConclusionProposed system showed a potential to be a nextgeneration fetal monitoring device for early to midtrimester.Ref:Shinozuka N et al. J Ultrasound Med,1994:131:19-25Yamakoshi Y et al. Ultrasonics,1996 34:769-775.Fig1 Example display of “Behaviogram”NotesPage 73


Abstract20Comparison of long axis cardiac function assessed by VectorVelocity Imaging <strong>and</strong> conventional Tissue Doppler <strong>and</strong> M-modemethods in the human fetusHikoro Matsui 1,2 Ioannis Germankis 1,2 , Helena Gardiner 1,21 Faculty of Medicine, Imperial College at Queen Charlotte's & Chelsea Hospital;2 Royal Brompton Hospital, London, UKBackground:We present preliminary data on the feasibility ofmeasuring long axis fetal cardiac function using a newspeckle-tracking algorithm. This allows evaluation ofmyocardial motion from two dimensional echocardiographicdigital images. We aimed to compare long axis cardiacfunction measurements using vector velocity imaging(VVI) with measurements of tissue velocities (TDI) <strong>and</strong>maximal displacement of the atrioventricular ring recordedusing the established methods of pulsed Doppler <strong>and</strong>M-mode in human fetuses.Methods:Conventional <strong>and</strong> VVI measurements were recorded inthe four chamber view in 12 fetuses aged 17 to 34 weeks’gestation. Seven had normal hearts, 3 had structuralmalformations: hypoplastic left heart syndrome,atrioventricular septal defect <strong>and</strong> pulmonary stenosis.One pair had twin-twin transfusion syndrome. All valuesrepresent the mean of 3 measurements at the left freewall, septal <strong>and</strong> right free wall of each atrioventricular ring.Off-line analysis was performed prospectively usingSyngo-VVI software (Siemens) creating a tracking movieautomatically. The average frame rate of the images was89fps (52-146fps). We allowed only 10 minutes fortracking <strong>and</strong> considered recordings that lay outside thistime limit unsuitable for analysis. We created atime-velocity curve of the atrioventricular ring <strong>and</strong>measured myocardial long axis shortening (Sm-VVI) <strong>and</strong>lengthening (Em-VVI <strong>and</strong> Am-VVI). We calculated themaximal amplitude of atrioventricular ring from thetracking coordinates (Amp-VVI). Conventional recordingsof right <strong>and</strong> left ventricular myocardial long axis shortening(Sm-TDI) <strong>and</strong> lengthening (Em-TDI, Am-TDI) were madeusing pulsed Doppler <strong>and</strong> maximum amplitude of theatrioventricular ring displacement from M moderecordings as described previously (Amp-M).Results:Automatic tracking was not feasible in 4 ventricles (16%)but was possible using conventional methods in all but theleft ventricle in the case of hypoplastic left heart syndrome.In line with conventional methods, VVI myocardialvelocities increased with gestational age <strong>and</strong> were higherin RV free wall than in LV free wall. However, while therewas good correlation using both methods, particularlyAmp-M <strong>and</strong> Amp-VVI (R=0.91, p


THE GENETIC ANALYSIS OF DEEP VEIN THROMBOSISDURING PREGNANCY AND PERINATAL ASSOCIATEDDISORDERS IN JAPANESER. Neki 1 , N. Yamada 1 , Y. Tokito 1 , N. Iwanaga 1 , K. Ueda 1 , K. Yamanaka 1 , M.Nozawa 1 , T. Ikeda 1 , T. Fujita 2 , J. Ishikawa 3 , Y. Sato 3 , T. Miyata 3 , O. Fukui 4 ,N,Suehara 41 Department of Perinatology, National Cardiovascular Center, Suita, <strong>Japan</strong> 2 Department ofMaternal Medicine, Osaka Medical Center <strong>and</strong> Research Institute for Maternal <strong>and</strong> ChildHealth, Izumi, <strong>Japan</strong> 3 Research Institute, National Cardiovascular Center, Suita, <strong>Japan</strong>4 Department of Obstetrics, Osaka Medical Center <strong>and</strong> Research Institute for Maternal <strong>and</strong> ChildAbstract21Health, Izumi, <strong>Japan</strong>Introduction:Deep vein thrombosis (DVT) <strong>and</strong> pulmonarythromboembolism (PTE) are easily occurred duringpregnancy because of its hypercoagulation status. It isreported that DVT or PTE is closely related with geneticfactors. Furthermore in Europe <strong>and</strong> the United States,thrombophilia-associated pregnancy wastage is reported.Our purpose is to examine the relation of genetic factorsto DVT/PTE during pregnancy <strong>and</strong> pregnancy wastage in<strong>Japan</strong>ese.Methods:We sequenced the entire coding regions of 3anticoagulant genes, PROS1 (protein S), PROC (proteinC), <strong>and</strong> SERPINC1 (antithrombin) in 178 complicatedpregnant cases: 12 cases of DVT/PTE, 124 cases ofinfertility including habitual abortion, 42 cases of othercomplications including intrauterine growth restriction,intrauterine fetal death, pregnancy induced hypertension,abruptio placenta.Results:Three cases with DVT/PTE had genetic mutations inPROS1,one case in PROC that could affect the activity.The genetic mutations in PROS1, PROC<strong>and</strong> SERPINC1that could affect the activity have been identified in each2cases with infertility. The genetic mutations in PROS1have been identified in 2 cases with other complications.Thus, genetic mutations of 3 genes have been observedin 33.3% in DVT/PTE cases, 4.8% in infertility cases, <strong>and</strong>4.8% in other complicated cases.Conclusions:Our intensive sequence analysis of 3 anticoagulant genesshowed that 12out of 178 cases (6.7%) carried geneticmutations that may influence the functional activity. Weconcluded that we <strong>Japan</strong>ese also have the genetic factorsto DVT/PTE during pregnancy <strong>and</strong> pregnancy wastage.NotesPage 75


Abstract22 A NEW MONITARING METHOD OF FGR FETUSKazunao Suzuki, Motoi Sugimura, Naohiro KanayamaDepartment of Obstetrics & Gynecology, Hamamatsu University School of Medicine,Hamamatsu, Shizuoka, <strong>Japan</strong>The management of FGR is becomoing one of the majorproblems in medicine from the aspect of fetal origin ofadult disease. Near-infrared spectroscopy (NIRS) hasbeen used as a noninvasive method for monitoring thereal -time oxygenation status in area such as the brain<strong>and</strong> striated muscle. Because the oxygenation status ofthe placenta is closely related to the fetal condition, theevaluation of placental oxygen condition would contributefetal management. We developed transabdominalmonitoring of the oxygenation of the placenta by NIRS in15 normal fetus <strong>and</strong> 15 FGR fetus. We succeeded inobtaining oxyhemoglobin <strong>and</strong> deoxyhemoglobin datathrough the maternal abdomen. We calculated tissueoxygenation index (TOI) from these data . The TOI levelsof normal pregnancy were not changed during 2nd <strong>and</strong>3rd trimester. The TOI value of normal fetus was70.1+/-0.6. Interestingly the TOI value of FGR was79.1+/-2.8. The TOI value of FGR was significantly higherthan that of normal pregnancy. This results could beexplained that impairment of trophoblastic functiondecreases the absorption of oxygen of maternaloxyhemoglobin from intervillous space to fetal circulationresulting in high level of TOI in FGR placenta. Thisphenomenon seems to be similar to TOI changes in lowtemperature therapy for the brain. Measurement ofPlacental TOI by transabdominal NIRS could be a new<strong>and</strong> useful indicator for management of FGR.NotesPage 76


Abstract23Keynote speakerGROWTH HORMONE: A NEUROTROPHIC FACTOR DURINGCHICK EMBRYOGENESISSteve Harvey, Marie-Laure Baudet, Esmond J. S<strong>and</strong>ersDepartment of Physiology, University of Alberta, Edmonton, Alberta, T6G 2H7, CanadaEmbryogenesis <strong>and</strong> fetal growth largely occur in theabsence of the pituitary gl<strong>and</strong> <strong>and</strong> are traditionallyconsidered to be growth-without-growth hormonesyndromes. However, while pituitary somatotrophs donot appear ontogenetically until at least ED (embryonicday) 14 of the 21 day incubation period of chickembryogenesis, extrapituitary tissues express the GHgene much earlier in development. Neural tissues, forinstance, (particularly projecting neurons of thefacial/acoustic, glossopharangeal, trigeminal <strong>and</strong> vagalnerves <strong>and</strong> from retinofugal <strong>and</strong> tectotubular tracts)contain abundant GH immunoreactivity <strong>and</strong> GH mRNA byED3 <strong>and</strong> the overlapping distribution of GH receptor(GHR) immunoreactivity <strong>and</strong> GHR mRNA suggests GHinvolvement in early neurogenesis. This possibility issupported by antiapoptotic actions of retinal GH thatpromote the survival of retinal ganglion cells (RGCs),since the immunoneutralization of endogenous GH inRGCs or the siRNA knockdown of endogenous GH mRNAresults in an increase in RGC apoptosis. Retinal GHmay also act as a growth factor during neurogenesis,since GH is found in the retinofugal tract (optic fibre layer,optic nerve head, optic chiasm, optic tract) prior to thesynapsing of these neurons with their target sites in thevisual centre of the brain. The growth of these neuronsmay therefore be due to the anterograde translocation ofthe GH along RGC axons. This possibility is supportedby a loss of GH immunoreactivity in the optic fibre layerafter ED 12 of development, following the synapsing of theretinofugal nerves within the optic tectum of the brain.This possibility is also supported by a trophic effect ofexogenous chicken GH on RGC neurite outgrowth.Neurotrophic actions of GH within the visual systemduring embryogenesis are also indicated by its inductionof insulin-like growth factor (IGF)-1 in the neural retina<strong>and</strong> by the expression of a GH-response gene (GHRG)-1(a specific marker of GH action in birds) in the optictectum <strong>and</strong> other visual centres of the brain. Chickembryogenesis is therefore not a growth-without-GHsyndrome <strong>and</strong> neurogenesis within the visual system, inparticular, appears to be dependent upon local autocrineor paracrine actions of extrapituitary GH that promoteneural development.Supported by NSERC of CanadaNotesPage 78


Maternal administration of dexamethasone causes significantalterations in the electrocortical activity of the preterm fetalsheepJoanne Davidson, Lindsea Booth, Josine Quaedackers, Alistair J Gunn<strong>and</strong> Laura Bennet.<strong>Fetal</strong> Physiology <strong>and</strong> Neuroscience Group, Dept of Physiology, The University of Auckl<strong>and</strong>,Auckl<strong>and</strong>, New Zeal<strong>and</strong>.Abstract24Antenatal corticosteroid therapy enhances fetal lungmaturation in pregnancies at risk for preterm delivery, butmay lead to injury or impaired development of otherorgans such as the brain. This study examined the effectsof a single course of maternally administereddexamethasone (DEX) on fetal brain activity. Pregnantewes at 103 days gestation, received two intramuscularinjections of either DEX (n = 8) or vehicle (n = 7) 24 hoursapart. <strong>Fetal</strong> electrocorticogram (ECoG) activity wasmonitored continuously from 24h before until 120h afterthe first injection. DEX injection one, led to a significantrise in ECoG amplitude, peaking around 12h. A second,smaller rise occurred after injection two with a return tonormal seen after 30 hours. Further analysis of the ECoGsignal showed that the rise in amplitude was mediated byvery high amplitude electrocortical discharges (up to400uV compared to normal background of 50uV) whichwere epileptiform in nature (Figure). ECoG frequency(spectral edge) was not significantly different during thistime, but was significantly elevated compared to controlfrom 17 hours after injection 2. In conclusion,dexamethasone appears to induce short-term epileptiformactivity, <strong>and</strong> long-term alterations in ECoG frequency inthe preterm fetal sheep, at an age which is equivalent tothe 28-30 week human in terms of neural maturation.Studies show that glucocorticoids can facilitate asustained calcium influx through glutamate receptorswhich may mediate the epileptiform activity we observed.The increased spectral edge may reflect dexamethasoneinduced advanced cortical maturation. Further study isrequired to determine whether dexamethasone causedneural injury.ABCDFigure 1. <strong>Fetal</strong> ECoG activity after maternaldexamethasone treatment (A, B, C) vs vehicle treatment (D)NotesPage 79


Abstract25Therapeutic Hypothermia Changes the Prognostic Value ofClinical Evaluation of <strong>Neonatal</strong> Encephalopathy.Alistair J. Gunn, Andrew Whitelaw, John Barks, John S. Wyatt, DenisAzzopardi, Charlene M. Robertson, Marianne Thoresen on behalf of theCoolCap Study GroupObjectives:to evaluate whether therapeutic hypothermia alters theprognostic value of clinical grading of neonatalencephalopathy.Study Design:Secondary analysis of a multicenter study of 234 terminfants with neonatal encephalopathy r<strong>and</strong>omized to headcooling for 72 h starting within 6 h of birth, with rectaltemperature maintained at 34.5±0.5°C, followed byrewarming over 4 h, or st<strong>and</strong>ard care at 37.0±0.5°C.Severity of encephalopathy was measured prer<strong>and</strong>omization<strong>and</strong> on day 4, after rewarming, in 177infants; 31 infants died before day 4 <strong>and</strong> data weremissing for 10 infants. The primary outcome was death orsevere disability at 18 months of age.Results:Milder pre-r<strong>and</strong>omization encephalopathy, greaterimprovement in encephalopathy from r<strong>and</strong>omization today 4, <strong>and</strong> cooling were associated with favorableoutcome in multivariate binary logistic regression.Hypothermia did not affect severity of encephalopathy atday 4, however, among infants with moderateencephalopathy at day 4, those treated with hypothermiahad a significantly higher rate of favorable outcome (31/45infants, 69%, p=0.006) compared with st<strong>and</strong>ard care(12/33, 36%).Conclusions:Infants with moderate encephalopathy on day 4 may havea more favorable prognosis after hypothermia treatmentthan expected after st<strong>and</strong>ard care.NotesPage 80


AbstractCan erythropoietin be used to protect the fetal sheep brain fromrepeated exposure to endotoxin?261 R De Matteo, 1 L Cardamone, 1 V Stacy, 1 N Blasch, 2 M Probyn, 3 N Hale,3 S Rees, 1 R Harding1 Department of Anatomy <strong>and</strong> Cell Biology, Monash University, Melbourne, Australia2 School of Biomedical Sciences, University of Queensl<strong>and</strong>, Brisbane, Australia3 Department of Anatomy <strong>and</strong> Cell Biology, University of Melbourne, Melbourne, AustraliaBackground <strong>and</strong> aim:Intrauterine inflammation <strong>and</strong> infection have been linkedto preterm birth <strong>and</strong> fetal brain injury. In human infants,the brain injury is particularly seen in white matter.Exposure of immature fetal sheep to the bacterialendotoxin lipopolysaccharide (LPS) causes fetalhypoxemia, hypotension <strong>and</strong> white matter injury (Duncanet al 2002). Erythropoietin (EPO) has neuroprotectiveproperties <strong>and</strong> could alleviate the brain injury induced byintra-uterine infection. In the present study our aim wasto determine the physiological effects of EPO in the ovinefetus exposed to LPS.Methods:At 0.7 of gestation (term ~147 days) ewes <strong>and</strong> fetuseswere anesthetized <strong>and</strong> fetuses chronically catheterized.Following 5±1 days of recovery fetuses received either: i)an i.v. bolus dose of LPS (~0.9µg/kg) on 3 consecutivedays (n=9); ii) i.v. bolus of LPS followed 1 hour later by5000IU of recombinant human Epo (rhEpo; n=15); iii)rhEpo alone (n=3); iv) saline alone (n=3). <strong>Fetal</strong> arterialblood gases <strong>and</strong> mean arterial pressure were monitored.Six days after the study period ewes <strong>and</strong> fetuses wereeuthanased <strong>and</strong> the fetal brains examined histologically.Results:<strong>Fetal</strong> exposure to saline alone or rhEpo alone did not alterfetal arterial blood gases or blood pressure. Three fetuses(33%) did not survive treatment with LPS alone, <strong>and</strong> sevenfetuses (47%) died after receiving LPS plus rhEpo. Insurviving fetuses, repeated administration of LPS resultedin hypoxemia <strong>and</strong> an increase in hematocrit (Hct).Fetuses treated with LPS plus rhEpo become significantlymore hypoxemic, displayed higher plasma lactate levels,became more hypotensive <strong>and</strong> tended to be tachycardicthan fetuses treated with LPS alone (p


Abstract27ANTI-INFLAMMATORY EFFECTS OF SULFASALAZINE IN ANOVINE MODEL OF IN UTERO INFECTION: A PROSPECT FORNEUROPROTECTION?Burcu Saglam 1 , Graham Jenkin 2 , Suzanne L. Miller 3 , <strong>and</strong> Euan M. Wallace 11 Department of Obstetrics <strong>and</strong> Gynaecology, 2 Monash Immunology <strong>and</strong> Stem Cell Laboratories<strong>and</strong> 3 Deparment of Physiology, Monash University, Clayton, Victoria.Background:There is a causal relationship between inflammation,arising from intrauterine infection, <strong>and</strong> injury to the fetalbrain. Preterm ovine fetal exposure to lipopolysaccharide(LPS) is a commonly used model for exploring themechanisms underlying inflammation- induced fetal braininjury. Inflammation caused by LPS is mediated by thetranscription factor nuclear kappa-B (NFκB). The aim ofthis study was to investigate the effects of oraladministration of Sulfasalazine (SSZ), an NFκB inhibitor,on circulating tumour necrosis factor-α (TNF-α)concentrations <strong>and</strong> fetal brain injury.Methods:6 twin-pregnant ewes were r<strong>and</strong>omly assigned to receiveSSZ tablets (n=2) or placebo sugar tablets (n=4). Surgerywas performed at 104-106 days gestation for catheterplacement in the femoral artery <strong>and</strong> vein of both of twinfetuses. Each ewe received SSZ or placebo, orally. Onefetus received three daily i.v. infusions of LPS (100ng/kg,placebo-LPS or SSZ-LPS); the control co-twin(placebo-Saline or SSZ-Saline) received saline injectionsof identical volume. <strong>Fetal</strong> plasma samples were taken tomeasure TNF-α <strong>and</strong> fetal brains were collected at postmortem for immunohistochemical evaluation of apoptoticcells by terminal deoxynucleotidyl transferase-mediateddUTP-nick end labelling (TUNEL).Results:There was an increase in plasma TNF-α concentrations(from undetectable levels to 85.77±15.84ng/ml) in theplacebo-LPS fetuses following the first LPS administration,which returned to baseline within 6 hours post-LPSexposure. There was an attenuation of the TNF-αresponse with subsequent LPS exposure. TNF-α wassignificantly increased in the SSZ-LPS fetuses (fromundetectable levels to 42.16±3.86ng/ml) but to a lesserextent than in the Placebo-LPS group. TNF-α was notdetectable in either of the Saline treated groups.TUNEL-positive staining cells were present in the brainsections of Placebo-LPS fetuses <strong>and</strong> there appeared tobe a decrease in TUNEL-positive staining in the SSZ-LPSbrains.Conclusion: Sulfasalazine reduces concentrations ofTNF-α in the fetal circulation <strong>and</strong> there was a decrease inthe presence of TUNEL-positive cells within these fetalbrains. We propose that SSZ may be neuroprotectivefollowing inflammation.NotesPage 82


AbstractFETAL BRAIN INJURY IN SINGLE UMBILICAL ARTERYLIGATION (SUAL) INDUCED IUGR28Low HM 1 , Supramaniam VG 1 , Castillo-Meléndez M 2 , Jenkin G 1 , Miller SL 1 ,Wallace EM 3 .Monash Immunology <strong>and</strong> Stem Cell Laboratories 1 , Departments of Physiology 2 <strong>and</strong> Obstetrics<strong>and</strong> Gynaecology 3 , Monash University, Victoria, 3800, AustraliaIntrauterine growth restriction (IUGR) is associated withincreased risk of premature delivery, risks of death <strong>and</strong>morbidity <strong>and</strong> increased risks of neurological <strong>and</strong>cognitive impairments in IUGR infants. We haveestablished a chronic fetal sheep hypoxia model of singleumbilical artery ligation (SUAL) with brain sparing <strong>and</strong>significantly elevated brain to body weight ratio. Weexamined the morphological changes in the subcorticalwhite matter, striatum, thalamus <strong>and</strong> hippocampusregions of the brain following SUAL.Methods:Pregnant singleton ewes underwent surgery at 105-110days gestation (dGA). Fetuses were instrumented withfemoral artery, jugular vein <strong>and</strong> amniotic fluid catheters.Single umbilical artery ligation (SUAL) was performed toinduce IUGR (n=5) while the cord remained intact incontrols (n=4). Electromyography (EMG) leads weresutured to the external wall of the uterus. SUAL animalswere sacrificed when an increased EMG activity,indicative of labour onset, was observed. Controls weresacrificed at 135 dGA. At post mortem, fetal brains wereperfused with 0.9% saline followed by 4%PFA. The fixedbrains were sectioned into blocks <strong>and</strong> paraffin embedded.White matter injury was assessed using cresyl violet <strong>and</strong>acid fuchsin (CV-AF; neuronal injury), lectin (activatedmicroglia/macrophages), Tau (neurofibrillary tangles) <strong>and</strong>albumin (blood brain barrier; BBB) immunohistochemistry.Results:<strong>Fetal</strong> hypoxaemia <strong>and</strong> IUGR was established via SUAL.IUGR fetuses showed significant brain sparing comparedto controls with brain to body weight ratios of 18.09±0.49<strong>and</strong> 13.45±0.13 respectively. Albumin <strong>and</strong> Tau positivecells were present in the subcallosal bundle, cingulategyrus, thalamic nuclei, periventricular thalamic nuclei <strong>and</strong>internal capsule of SUAL fetuses but not in controls.Lectin positive cell numbers were elevated in the mostseverely affected SUAL fetus (11.34±2.489 cells per fieldof view) <strong>and</strong> minimal in other SUAL fetuses (0.96±0.32)<strong>and</strong> control fetuses (0.56±0.23). Histochemical stainingusing CV-AF identified a slight increase in pyknotic cells inSUAL fetuses (20.87±1.45) compared to controls(18.65±1.77) in the cingulate gyrus, ventral striatum <strong>and</strong>septal nucleus.Conclusion:The brains of SUAL induced IUGR fetuses displayedmarked neuronal damage, activation of microglia/macrophages, presence of neurofibrillary tangles <strong>and</strong> BBBcompromise, which may lead to significant morbidity afterbirth.NotesPage 83


Abstract29 Perinatal DiI tracing of developing hypothalamic connections.Makarenko I.G., Alpeeva E.V.N.,K. Koltzov Institute of Developmental Biology RAS, Moscow, Russian FederationOBJECTIVE:Hypothalamus plays an important role in regulation ofbasic visceral, somatic <strong>and</strong> emotional functions that canbe realized by its connections with numerous brainstructures. There is a notion that disruption ofhypothalamic development may be unsuspected cause ofdisorders such as obesity <strong>and</strong> high blood pressure,emotional or neurological disturbances. Unfortunatelyformation of axonal connections is one of the mostunknown events of brain development. The aim of ourstudy was to describe the time schedule <strong>and</strong>consequence of the development of the hypothalamicprojection systems.METHODS:Carbocyanine dye tracing method was used on the datedrats from E14 to P10. Crystals of DiI were inserted into thedifferent parts of hypothalamus, septum, midbrain <strong>and</strong>pituitary lobes on the prefixed with 4% paraformaldehydebrains. After the storage in the same fixative for 4-16month labeled nerve cells <strong>and</strong> fibers were revealed onthick vibratome sections using fluorescent <strong>and</strong> confocalmicroscopy.RESULTS:Retrogradely labeled neurons were visualized in the brain<strong>and</strong> carefully described after all kinds of the DiIapplications <strong>and</strong> thus revealed the sources of distinctprojection systems. Early prenatal development beginningfrom E14-E15 was specific for hypothalamo-posteriorpituitary, mammillo-tegmental <strong>and</strong> septohypothalamicconnections. It was shown that septohypothalamicconnections are reciprocal <strong>and</strong> more numerous with thepreoptic region <strong>and</strong> anterior hypothalamus than withmedial <strong>and</strong> caudal hypothalamic regions. All these fibersystems are well developed before the birth. Hypothalamicprojections to the intermediate pituitary lobe were revealedfirst close to the end of intrauterine life <strong>and</strong> differentiatedprogressively from P5 to P20. Mammillothalamic tracteven organized by the collateral branches of themammillotegmental tract grows from E18, reach unilateralanteromedial thalamic nucleus on E20-E21 <strong>and</strong> innervatesall anterior thalamic nuclei on postnatal stages (from P2 toP10). Functional activity of the thalamic synapses wasconfirmed by the immunocytochemical visualization ofsynapsin on P4-P5.CONCLUSIONS:Thus we have described specific time schedule of thenormal development of several hypothalamic projectionsystems. This work was supported by RFBR grant07-04-00798.NotesPage 84


AbstractKeynote speaker30 Chronic <strong>Fetal</strong> Hypoxia : Consequences <strong>and</strong> TreatmentSuzanne L. Miller 1,2 , Veena G. Supramaniam 2 , Euan M. Wallace 3 , GrahamJenkin 2Department of 1 Physiology, 2 Monash Immunology <strong>and</strong> Stem Cell Laboratories, <strong>and</strong> 3 Obstetrics<strong>and</strong> Gynaecology, Monash University, Victoria, AustraliaChronic fetal hypoxia, together with intrauterine growthrestriction (IUGR), continue to present serious challengesin modern obstetrics, both in terms of diagnosis <strong>and</strong>treatment options. IUGR is strongly associated withincreased risks of perinatal mortality <strong>and</strong> significant short<strong>and</strong> long term morbidity. To explore possible treatmentstrategies, we use the technique of single umbilical arteryligation (SUAL) in ovine fetuses. This model inducesplacental insufficiency thereby producing chronic hypoxia,cardiovascular redistribution <strong>and</strong> asymmetric fetal growthrestriction, as occurs in human IUGR fetuses. A notablecomplication of IUGR pregnancies is preterm birth <strong>and</strong>therefore glucocorticoids (including betamethasone; BM)are often administered to mature the fetal lungs. However,in our SUAL model, BM is potently vasoactive causingsignificant cerebral hypoperfusion, followed by a largereperfusion in SUAL fetuses which is not observed inhealthy fetuses. The degree of rebound perfusion iscorrelated with fetal brain cellular lipid peroxidation <strong>and</strong>apoptosis, suggesting that BM administration could havesignificant adverse effects in the IUGR fetus. IUGR is alsoassociated with significant fetal oxidative stress <strong>and</strong>inflammation. Mitigation of these factors may lead toimproved perinatal <strong>and</strong> long term outcomes. Using ourSUAL model, we are currently exploring a number ofselect treatment regimens, including phosphodiesteraseinhibitors to improve uterine blood flow, anti-inflammatoryagents <strong>and</strong> antioxidants to reduce adverse effects ofIUGR, thereby improving neonatal wellbeing.Unfortunately, administration of the type 5phosphodiesterase inhibitor sildenafil citrate (Viagra),does not improve uteroplacental blood flow in IUGRfetuses. Outcomes from the maternal administration ofanti-inflammatory or antioxidant agents in IUGR look morepromising as possible future therapies.DR SUZANNE L MILLERQualifications:BSc (Hons), PhD, Department of Physiology, Faculty of Medicine, Nursing <strong>and</strong> Health Sciences Monash University.Current Appointment:Senior Research Officer, Department of Physiology <strong>and</strong> Monash Immunology <strong>and</strong> Stem Cell Laboratories, MonashUniversity. Research Fellow, Royal Australian <strong>and</strong> New Zeal<strong>and</strong> College of Obstetricians <strong>and</strong> Gynaecologists(RANZCOG).PI on NHMRC Program Grant: “Control of Reproductive Processes”CI on NHMRC Project Grant “The effects of maternal glucocorticoid administration in growth restricted fetuses”Research Summary:After completing my PhD at Monash University in 1999, in which I studied the regulation of uterine blood flow duringpregnancy, I gained a Postdoctoral Fellowship at University College London, where I worked with Professors MarkHanson <strong>and</strong> Donald Peebles in the highly regarded <strong>Fetal</strong> <strong>and</strong> <strong>Neonatal</strong> Research Group. My postdoctoral researchaddressed brain growth, development <strong>and</strong> the use of potential neuroprotective strategies to ameliorate the effects ofreduced oxygen availability <strong>and</strong> infection in a fetal sheep model. I returned from the UK in 2001 <strong>and</strong>, since then, havebeen working with a team that is exploring fetal growth, development <strong>and</strong> adaptation in response to chronic fetal hypoxiaor infection in pregnancy. Presently, I am funded by the NHMRC <strong>and</strong> the RANZCOG on projects that investigate the fetaladaptations observed in response to acute hypoxia, chronic hypoxia, reduced blood flow, infection <strong>and</strong> glucocorticoidexposure from the cellular level through to gross developmental <strong>and</strong> cognitive effects, with a view to implementingclinically useful neuroprotective strategies for the at risk fetus <strong>and</strong> neonate.NotesPage 86


AbstractProliferation in the subventricular zone (svz) after severehypoxia <strong>and</strong> induced hypothermia in preterm fetal sheep31Sherly George, Robert Barrett, Laura Bennet, Justin Dean, Alistair JanGunn.<strong>Fetal</strong> Physiology <strong>and</strong> Neuroscience Group, Dept Physiology, The University of Auckl<strong>and</strong>,Auckl<strong>and</strong>, New Zeal<strong>and</strong>Prolonged, moderate cerebral hypothermia is the firstclinically proven therapy to improve recovery fromhypoxia-ischaemia at term. Recent experimental datasuggest that cooling is also neuroprotective after severehypoxia in preterm animals, but that it may reduceproliferation of brain cells in the periventricular whitematter, at a critical period in the development of the brain.Although there is significant ongoing proliferation in theperiventricular regions, new cells are primarily producedin the subventricular zone (SVZ). In a chronicallyinstrumented sheep model of preterm asphyxia, weinvestigated the effects of cerebral hypothermia onproliferation in the SVZ. Preterm (0.7 gestation) fetalsheep received complete umbilical cord occlusion for 25min followed by cerebral hypothermia starting 90 min afterreperfusion <strong>and</strong> continuing until 72 h after occlusion.There was a significant increase in numbers of cellslabeled with proliferating cell nuclear antigen (PCNA), butnot of cells labeled with KI-67, in the hypothermiaocclusiongroup compared with normothermia-occlusionanimals <strong>and</strong> sham controls. In conclusion, despiteprofound suppression of proliferation in the vulnerableperiventricular region after severe hypoxia <strong>and</strong> treatmentwith hypothermia in the developing brain, proliferation inthe SVZ was not suppressed, either by hypoxia or byprolonged hypothermia. The apparent differences in effectof hypothermia suggested by the two markers forproliferation likely reflect the more restricted binding ofPCNA in the cell cycle. Further studies are needed toevaluate the long-term impact of hypoxia <strong>and</strong> hypothermiaon cell division <strong>and</strong> differentiation in the developing brain.NotesPage 87


Abstract32The ontogeny of chemoreflex <strong>and</strong> haemodynamic responses toprolonged umbilical cord occlusion in fetal sheepGuido Wassink, Laura Bennet, Lindsea C. Booth, Ellen C. Jensen, BertWibbens, Justin M Dean, <strong>and</strong> Alistair J Gunn.<strong>Fetal</strong> Physiology <strong>and</strong> Neuroscience Group, Dept Physiology, The University of Auckl<strong>and</strong>,Auckl<strong>and</strong>, New Zeal<strong>and</strong>It has been hypothesized that blunted chemoreflexmediated responses to hypoxia may compromise the abilityof the preterm fetus to adapt to hypoxic or asphyxial stress.However, there are only limited quantitative data on theontogeny of chemoreflex <strong>and</strong> hemodynamic responses tosevere asphyxia. Chronically instrumented fetal sheep at0.6 (n=12), 0.7 (n=12) <strong>and</strong> 0.85 (n=8) of gestational age(ga; term = 147 days) were exposed to 30, 25 or 15minutes of complete umbilical cord occlusion, respectively.At all ages occlusion was associated with early onset ofbradycardia, profoundly reduced femoral blood flow <strong>and</strong>conductance, <strong>and</strong> hypertension. The 0.6ga fetuses showeda significantly slower <strong>and</strong> lesser fall in femoral blood flow<strong>and</strong> conductance compared with the 0.85ga group, with acorrespondingly reduced relative rise in mean arterial bloodpressure. As occlusion continued, the initial adaptation wasfollowed by loss of peripheral vasoconstriction <strong>and</strong>progressive development of hypotension in all groups. The0.85ga fetuses showed significantly more sustainedreduction in femoral conductance but also more rapid onsetof hypotension than either of the younger groups.Electroencephalographic (EEG) activity was suppressedduring occlusion in all groups but the rate <strong>and</strong> degree ofsuppression were least at 0.6ga. In conclusion, thenear-midgestation fetus shows attenuated initial(chemoreflex) responses to severe asphyxia comparedwith more mature fetuses, but more sustainedhemodynamic adaptation <strong>and</strong> reduced suppression ofEEG activity during continued occlusion of the umbilicalcord. These results are consistent with greater cardiac<strong>and</strong> cerebral tolerance to profound hypoxianear-midgestation.NotesPage 88


AbstractEffect of acute <strong>and</strong> chronic hypoxia on amniotic fluid gases inchick embryogenesis.33M.V. Nechaeva 1 , H. Toenhardt 2 , D. Marquardt 2 .1 Institute of Developmental Biology RAS, Moscow, Russia, Mnechaeva2003@yahoo.com.2 Free University, Institute of Veterinary Physiology, Berlin, Germany.Objective:The prenatal hypoxia induces a complex response, <strong>and</strong>numerous functional systems are involved to prevent thedamage of embryo. The mechanism of the developmentalresponse to hypoxia is not entirely clear. We studied thechanges of oxygen tension in amniotic fluid (AF) duringdevelopment of chick embryo <strong>and</strong> under the influence ofacute <strong>and</strong> chronic hypoxia.Methods:Chick eggs were incubated at 37.5°C. Po 2 , Pco 2 , <strong>and</strong> pHwere measured in samples of AF using blood gasanalyzer Radiometer Copenhagen ABL 605 on incubationdays 10 (D10), 12 <strong>and</strong> 14 in control <strong>and</strong> after the eggexposure to acute hypoxia (10%O 2 for 10min) or chronichypoxia (15%O 2 since D6).Results:In control, the Po 2 in AF was about 55 mmHg <strong>and</strong>remained relatively constant during this period. The Pco 2significantly increased from 19.85 ± 0.52 to 30.22 ± 1.87mmHg, while pH significantly decreased from 6.91 ± 0.03to 6.58 ± 0.05 since D10 to D14.Acute hypoxia reduced the Po 2 in AF by 25; 23 <strong>and</strong> 16%from the control values on D10, 12 <strong>and</strong> 14, respectively.The Pco 2 increased, but the effect was significant only onD12. The pH decreased <strong>and</strong> this effect was morepronounced on D14.Chronic hypoxia increased the Po 2 in AF by 20% from thecontrol value on D10, <strong>and</strong> did not change it significantly onD12 <strong>and</strong> D14. The Pco 2 decreased on D10, D12, butdidn’t change on D14. The pH decreased on D10 <strong>and</strong> D12,but rose significantly on D14.Conclusion:The effects of acute <strong>and</strong> chronic hypoxia on gases valuesin AF were different. These results are discussed inregards to the role of oxygen in AF for the embryometabolism during hypoxia <strong>and</strong> for the function ofamniotic membrane, which is not vascularised <strong>and</strong> mayuse oxygen from amniotic fluid. This work was supportedby RFBR grant 05-04-49434.NotesPage 89


Abstract34Maternal treatment with allopurinol diminishes fetal cardiacoxidative stress following repeated episodes of ischaemiareperfusionin sheepH Torrance 1 , JB Derks 1 , MA Oudijk 1 , AS Thakor 2 , T Cindrova-Davies 2 ,F van Bel 1 , GHA Visser 1 , GJ Burton 2 & DA Giussani 21 Department of Perinatology, University Medical Centre Utrecht, The Netherl<strong>and</strong>s <strong>and</strong>2 Department of Physiology, Development & Neuroscience, University of Cambridge, UK.Introduction The prevention <strong>and</strong> managementof perinatal asphyxia remain major concerns inobstetric practice today. Umbilical cordcompressions (UCC) not only induce fetalasphyxia but also episodes of ischaemiareperfusion(I/R). I/R promotes the productionof reactive oxygen species (ROS), for instancevia activation of the xanthine oxidase (XO)pathway, which may lead to oxidative stress inthe fetal cardiovascular system. While treatmentwith allopurinol of severely asphyxic humanneonates reduced free radical formation <strong>and</strong>improved cardiovascular status (1), allopurinoltreatment started postnatally was deemed toolate to prevent oxidative damage (2). As a result,in pregnancy complicated with severe fetalasphyxia, recommendations to treat the fetusvia the mother, rather than the neonate, withallopurinol are currently being entertained. Thisstudy investigated the effects of maternaltreatment with allopurinol on indices of oxidativestress in the fetal heart following repeated UCCin late gestation sheep.Methods Under halothane anaesthesia, 10sheep fetuses <strong>and</strong> their mothers wereinstrumented at 0.8 of gestation with vascular<strong>and</strong> amniotic catheters, an inflatable occluderaround the umbilical cord, <strong>and</strong> a Transonic flowprobe around an umbilical artery. At least 5 dayslater, all fetuses were submitted to an I/Rchallenge produced by 5 x 10 min UCC at 10min intervals, each designed to reduce umbilicalblood flow (UBF) by 80-90% from baseline <strong>and</strong>to lead to a progressive fall in fetal pHa to 6.9. In5 fetuses, the I/R challenge was induced duringmaternal i.v. treatment with allopurinol (SigmaLtd., 30 mg.kg -1 over 20 minutes). In theremaining 5 fetuses, the I/R challenge wasinduced during maternal infusion with salinevehicle at the same rate. Infusion started 10 minbefore the 4 th UCC <strong>and</strong> finished immediatelyafter the end of the 4 th UCC. The fetal heartswere collected 48h after I/R <strong>and</strong> snap frozen formeasurement of oxidant <strong>and</strong> antioxidantproteins by Western blot. Hearts collected from5 non-instrumented fetal sheep at 0.8 gestationserved as controls. Statistical comparisons weremade using one-way ANOVA with the Tukeypost-hoc test. Significance was accepted whenp


Pre-existing hypoxia is associated with a delayed but moresustained rise in T/QRS ratio during prolonged umbilical cordocclusion in near-term fetal sheepLaura Bennet, Bert Wibbens, Jenny A. Westgate, Harmen H. De Haan,Guido Wassink, <strong>and</strong> Alistair J. Gunn<strong>Fetal</strong> Physiology <strong>and</strong> Neuroscience Group, Dept of Physiology, The University of Auckl<strong>and</strong>,Auckl<strong>and</strong>, New Zeal<strong>and</strong>.Abstract35There is limited information on whether pre-existing fetalhypoxia alters hemodynamic responses <strong>and</strong> changes inT/QRS ratio <strong>and</strong> ST waveform shape during subsequentsevere asphyxia. Chronically instrumented near-termsheep fetuses (124±1 days) were identified as eithernormoxic PaO 2 > 17 mmHg (n=9) or hypoxic PaO 2 ≤ 17mmHg (n=5), <strong>and</strong> then received complete occlusion of theumbilical cord for 15-min. Umbilical cord occlusion lead tosustained bradycardia, severe acidosis <strong>and</strong> transienthypertension followed by profound hypotension in bothgroups. Pre-existing hypoxia did not affect changes inmean arterial blood pressure, but was associated with amore rapid initial fall in femoral blood flow <strong>and</strong> vascularconductance <strong>and</strong> with transiently higher fetal heart rate at2 minutes <strong>and</strong> from 9 to 11 minutes of occlusioncompared with previously normoxic fetuses. Occlusionwas associated with a significant but transient rise inT/QRS ratio; pre-existing hypoxia was associated with asignificant delay in this rise (maxima 3.7 ± 0.4 vs. 6.2 ± 0.5minutes), but a slower rate of fall. There was a similarelevation in troponin-T levels 6 hours after occlusion in thetwo groups (median (range) 0.43 (0.08, 1.32) vs 0.55(0.16, 2.32) µg/L, N.S.). In conclusion, mild pre-existinghypoxia in normally grown singleton fetal sheep isassociated with enhanced centralisation of circulationafter umbilical cord occlusion <strong>and</strong> delayed elevation of theST waveform <strong>and</strong> slower fall, suggesting that chronichypoxia improves myocardial dynamics during asphyxia.NotesPage 91


NotesPage 92


Abstract36High incidence of ketosis <strong>and</strong> hyper-homocysteinemia in thelast trimester of <strong>Japan</strong>ese mothers. without any complicationsFukuoka Hideoki*, Watanabe Hiroko**, Nagai Yasushi***, OgasawaraChiyoko***, Asano Shigetaka** Institute for Epigenetic Regulation of <strong>Fetal</strong> Development Waseda University, Tokyo, <strong>Japan</strong>** Dapt of Nursing Science, the Graduate School of Medicine Kyoto University***Nagai Clinic, Saitama, <strong>Japan</strong>.In these 2 or 3 decades, the <strong>Japan</strong>ese rate of low birthweight infant has been growing to 9.44 % in 2004, <strong>and</strong> theaverage birth weight of boys <strong>and</strong> girls has been loweringunder 3000g in 2005. The next generation has beensuspected to face the high risk of adult diseases in future.To study the maternal nutritional state, in one urb<strong>and</strong>istrict hospital on the outskirts of Tokyo, we studied 198mothers at the gestational age 12th, 20th, 32nd <strong>and</strong> 36thweek, with DHQ (Diet History Questionnaire) <strong>and</strong> serumhomocystein <strong>and</strong> ketone bodies, under the approval of theEthical Committees of University of Tokyo. In <strong>Japan</strong>, theestimated energy requirement for 20’s pregnant mothersis 2550 Cal for the last trimester. DHQ study, however,disclosed no change of energy intake from 1st, 2nd until3rd trimester (average energy intake: 1723, 1754, 1792Cal, respectively), <strong>and</strong> ,by comparison, that of nonpregnant ladies was 1722 Cal. Many mothers weresuspected to be in the severe energy deficit <strong>and</strong> abnormalmetabolic state. Under this doubt, we measured theketone bodies. ( acetoacetate, 3βhydroxbutylate <strong>and</strong>acetone). High ketone bodies were detected in 11.7 <strong>and</strong>29.3 % at 12th <strong>and</strong> 32nd gestational week. In the lasttrimester, 89% mothers had the folate intake under 400 μg<strong>and</strong> 13% showed serum homocystein over 5.5±0.4 μg/L.High homocystein means the possibility of high3βadenosylhomocystein(3βADH) inside cells. The<strong>Japan</strong>ese maternal nutritional state is not so ideal withlong lower energy <strong>and</strong> folate intake <strong>and</strong> high incidence ofketosis, inducing lowering the birth weight. The averagebirth weight in no ketosis mothers was higher than theketosis group in each BMI without any clear relation offolate <strong>and</strong> energy intake. But low intake of folate withhigher homocystein suggests possibility of disruptedturnover of one carbon metabolism <strong>and</strong> DNA methylationin the fetus.NotesPage 94


AbstractGenomic imprinting in the placenta involves histone tailmodifications independent of DNA methylation37Kohzoh Mitsuya 1 <strong>and</strong> Kunihiro Okamura 21Biofunctional Science, Tohoku University Biomedical Engineering Research Organization(TUBERO)2Department of Obstetrics <strong>and</strong> Gynecology, Tohoku University School of MedicineImprinted genes are expressed from only one of theparental chromosomes <strong>and</strong> are marked epigenetically byDNA methylation <strong>and</strong> histone modifications. Theimprinting center 2 (IC2) on mouse distal chromosome 7is flanked by several paternally repressed genes, with themore distant ones imprinted exclusively in the placenta.We found that most of these genes lack parent-specificDNA methylation, <strong>and</strong> genetic ablation of methylationdoes not lead to loss of their imprinting in the trophoblast(placenta). The silent paternal alleles of the genes aremarked in the trophoblast by repressive histonemodifications (dimethylation at Lys9 of histone H3 <strong>and</strong>trimethylation at Lys27 of histone H3), which are disruptedwhen IC2 is deleted, leading to reactivation of the paternalalleles. Thus, repressive histone methylation is recruitedby IC2 (potentially through a noncoding antisense RNA) tothe paternal chromosome in a region of at least 700 kb<strong>and</strong> maintains imprinting in this cluster in the placenta,independently of DNA methylation. We propose that anevolutionarily older imprinting mechanism limited toextraembryonic tissues was based on histonemodifications, <strong>and</strong> that this mechanism was subsequentlymade more stable for use in embryonic lineages by therecruitment of DNA methylation. A possible role forhistone tail modifications in epigenetic inheritance will befurther discussed.NotesPage 95


Abstract38LACTOBACILLI SUPERNATANT INHIBITS TNF-α PRODUCTIONAND COX 2 EXPRESSION IN LPS-ACTIVATED PLACENTALTROPHOBLASTS1,3 M Yeganegi, 3 C Watson, 2 S Kim, 2 G Reid, 1 J Challis <strong>and</strong> 1,3 A Bocking1 Dept. Physiology & Ob/Gyn, U Toronto, Canada; 2 Dept. Microb. & Immun., U Western Ontario,London, Canada <strong>and</strong> 3 SLRI, Mt. Sinai Hosp., Toronto, CanadaObjective:Bacterial Vaginosis (BV) is characterized by absence ofendogenous Lactobacillus <strong>and</strong> an increased risk ofpreterm birth (PTB). BV-associated pathogenic bacteriaupregulate pro-inflammatory cytokines, leading toincreases in prostagl<strong>and</strong>ins. Lactobacilli are known toinhibit pro-inflammatory cytokines in mouse macrophages.We hypothesized that Lactobacillus rhamnosus GR-1interferes with the cascade leading to prostagl<strong>and</strong>insynthesis by downregulating pro-inflammatory cytokineproduction <strong>and</strong> COX 2 protein expression in humanplacental trophoblasts.Methods:Placental trophoblasts were isolated from term electivec-section placentae <strong>and</strong> divided into six groups: 1) Notreatment; 2) Treatment with LPS; 3,4) Treatment withlactobacilli supernatant or MRS media; 5,6) Pretreatmentwith lactobacilli supernatant or MRS followed by LPStreatment. COX 2 expression was measured by WesternBlot <strong>and</strong> cytokine concentrations by ELISA. Cell viabilitywas tested by LDH analysis.Results:LPS stimulation caused a marked increase in TNF-αproduction <strong>and</strong> COX 2 expression in placental trophoblasts.Pretreatment with lactobacilli supernatant downregulatedthese increases. Treatment with supernatant alone had noeffect on cytokine production or COX 2 expression. Therewere no changes in IL-1β concentrations with anytreatment. LDH analysis showed minimal apoptosis inlactobacilli treated cells.Conclusion:Probiotic lactobacilli inhibit both TNF-α production <strong>and</strong>COX 2 expression in placental trophoblasts. This studyprovides evidence for potential mechanisms by whichlactobacilli reduces the risk of BV-associated PTB.NotesPage 96


INTERACTION BETWEEN LIPOXYGENASE PATHWAY ANDPROGESTERONE ON 11β-HYDROXYSTEROIDDEHYDROGENASE TYPE 2 IN CULTURED HUMAN TERMPLACENTAL TROPHOBLASTSK Sato 1,2 , H Chisaka 2 , K Okamura 2 , JRG Challis 11 Department of Physiology, Obstetrics, Gynecology <strong>and</strong> Medicine, University of Toronto,Ontario, Canada 2 Department of Obstetrics <strong>and</strong> Gynecology, Tohoku University GraduateSchool of Medicine, Sendai, <strong>Japan</strong>Abstract39Objective:Placental 11β-hydroxysteroid dehydrogenase type 2(11β-HSD2) is localized to placental syncytiotrophoblasts<strong>and</strong> plays an important role in pregnancy maintenance<strong>and</strong> fetal maturation by inactivating excess glucocorticoids.In the event of intrauterine infection, lipoxygenase (LOX)metabolites are produced in the placenta <strong>and</strong> contribute topreterm labor <strong>and</strong> adverse fetal outcomes. On the otherh<strong>and</strong>, LOX metabolites are involved in production ofprogesterone, which is required for pregnancymaintenance. In this study, we evaluated the interactionsbetween LOX pathway <strong>and</strong> progesterone on 11β-HSD2.Specifically, we hypothesized that LOX metabolites wouldalter 11β-HSD2 <strong>and</strong> this effect would be mediated byprogesterone.Methods:We cultured human term placental trophoblasts in thepresence/absence of LOX inhibitors, Nordihydroguaiareticacid (NDGA), AA861 <strong>and</strong> Baicalein; LOX metabolites,Leukotriene B 4 (LTB4) <strong>and</strong> 12(S)-hydroxyeicosatetraenoate(12-HETE); progesterone (P4) <strong>and</strong> progesterone receptorantagonist RU486. We used radiometric conversion assay,real-time RT-PCR, Western blot analysis <strong>and</strong> ELISA.Results:LOX metabolites down-regulated 11β-HSD2 activity <strong>and</strong>mRNA expression. LOX inhibitors up-regulated 11β-HSD2activity <strong>and</strong> mRNA/protein expression, <strong>and</strong> these effectswere attenuated by addition of LOX metabolites. Netprogesterone output was increased by LOX metabolites<strong>and</strong> decreased by LOX inhibitors in a dose dependentmanner. 11β-HSD2 activity <strong>and</strong> mRNA/protein expressionwere down-regulated by progesterone, <strong>and</strong> these effectswere blocked by RU486. Both mRNA <strong>and</strong> proteinexpression of 11β-HSD2 were up-regulated by RU486.The suppressive effect of 12-HETE on 11β-HSD2 activitywas reversed by RU486.Conclusion:We conclude that 11β-HSD2 in human placentaltrophoblasts is decreased by progesterone <strong>and</strong> increasedby inhibition of endogenous LOX metabolites, <strong>and</strong> that acomponent of the effect of LOX metabolites on 11β-HSD2is mediated by their stimulation of endogenousprogesterone output.NotesPage 97


Abstract40Distribution of the thyroid hormone transporter, MCT8, in fetal <strong>and</strong>placental tissues of murine, human <strong>and</strong> ovine speciesJenny L Macrae, Theo J Visser*, Graham J Burton, F B Peter Wooding,Abigail L Fowden <strong>and</strong> Alison J ForheadDepartment of Physiology, Development <strong>and</strong> Neuroscience, University of Cambridge,Cambridge CB2 3EG, UK; *Department of Internal Medicine, Erasmus Medical Center,Rotterdam, The Netherl<strong>and</strong>s.Membrane transporters, such as monocarboxylatetransporter MCT8, allow thyroid hormones access tonuclear receptors in target tissues. Thyroid hormonesare important regulators of fetal growth <strong>and</strong> development;however, MCT8 expression in fetal tissues is unknown.This study investigated the distribution of MCT8 in fetal<strong>and</strong> placental tissues from murine, human <strong>and</strong> ovinespecies.Mouse embryos <strong>and</strong> placentae were collected at 16 <strong>and</strong>19d of gestation (term 20d). Tissues were also obtainedfrom newborn <strong>and</strong> adult mice. Human placentae in thefirst trimester (5-6w) <strong>and</strong> at term (40w), <strong>and</strong> embryos at9w, were collected during clinical procedures. A varietyof tissues were obtained from sheep fetuses at 130 <strong>and</strong>144d of gestation (term 145d). All animal <strong>and</strong> clinicalsamples were obtained according to UK legislation.Tissue expression of MCT8 was determined byimmunohistochemistry using a polyclonal rabbit antibodyagainst human MCT8 <strong>and</strong> visualised with 3-3’-diaminobenzamine. In all species, immunostaining wasabolished by co-incubation with blocking peptide.In mouse embryos, MCT8 was localised to cardiac <strong>and</strong>skeletal myocytes, hepatocytes, brown adipocytes, bonecells, pulmonary epithelium, intestinal epithelium, renaltubules, dorsal root ganglia, skin epidermis <strong>and</strong> choriodplexus. In the murine placenta, MCT8 was localisedspecifically to the yolk sac. There were no differences inlocalisation between embryonic, neonatal <strong>and</strong> adulttissues.In partial sections of human embryos, similar expressionof MCT8 protein was identified in cardiac <strong>and</strong> skeletalmyocytes, bone cells, skin epidermis <strong>and</strong> areas of thebrain. In the human placenta, MCT8 was expressed inendometrial gl<strong>and</strong> epithelium in the first trimester, <strong>and</strong> infetal villous trophoblast in the first trimester <strong>and</strong> at term.In ovine fetuses, MCT8 was detected in all tissuesexamined. Expression was observed in cardiac <strong>and</strong>skeletal myocytes, hepatocytes, brown adipocytes,pulmonary epithelium, renal tubules, adrenal gl<strong>and</strong>,pancreatic islets <strong>and</strong> skin epidermis. In the ovineplacenta, MCT8 appeared to be localised to the fetaltrophoblast layer. There was no difference in tissuelocalisation of MCT8 between the gestational agesstudied.Therefore, in murine, human <strong>and</strong> ovine species, MCT8 iswidely expressed in the fetus <strong>and</strong> placenta, especially intissues sensitive to thyroid hormones before birth.Supported by The Royal <strong>Society</strong> <strong>and</strong> Isaac Newton Trust.NotesPage 98


Abstract41DETERMINANTS OF THE NUMBER OF CARDIOMYOCYTES INSHEEP: THE INFLUENCE OF FETAL AND POSTNATALGROWTHVictoria Stacy, Megan Probyn, Robert DeMatteo, Nigel Wreford, RichardHarding, M. Jane BlackDepartment of Anatomy <strong>and</strong> Cell Biology, Monash University, VIC 3800, AustraliaAims:The muscle cells of the heart (cardiomyocytes) becometerminally differentiated <strong>and</strong> cease proliferating soon afterbirth; therefore, the ultimate number of cardiomyocytes isdetermined during fetal <strong>and</strong> early postnatal life. Our aimwas to determine the effects of fetal <strong>and</strong> postnatal growthin singleton <strong>and</strong> twin lambs on the number of ventricularcardiomyocytes.Methods:Hearts were collected at necropsy from both singleton(n=5) <strong>and</strong> twin (n=5) lambs at 9 weeks of age, whencardiomyocytes are terminally differentiated. The walls ofthe left <strong>and</strong> right ventricles were systematically sampled.The number of cardiomyocytes was stereologicallyestimated in the left <strong>and</strong> right ventricles <strong>and</strong> septum usingan unbiased optical dissector / fractionator technique.Results:At birth, twins were 17% lighter than singletons <strong>and</strong> 28%lighter at 9 weeks (both p


Modulation of Pulmonary Vascular Smooth Muscle Cell(PVSMC) Phenotype in Hypoxia: Role of cGMP-DependentProtein Kinase (PKG) .Weilin Zhou, Chiranjib Dasgupta, Sewite Negash, J Usha Raj.Division of Neonatology, Harbor-UCLA Medical Center, Los Angeles Biomedical ResearchInstitute at Harbor-UCLA, Torrance, CA 90502.Abstract42Chronic hypoxia triggers pulmonary vascular remodeling,which is associated with changes in PVSMC phenotypefrom a contractile, differentiated to a synthetic,de-differentiated type. We have reported previously thatacute hypoxia results in decrease in PKG activity inPVSMC (AJP, 2004), hence we investigated whetheraltered expression of PKG in hypoxia explain SMCphenotype modulation. Hypoxia-induced reduction in PKGexpression correlated strongly with the repressedexpression of SMC phenotypic markers myosin heavychain, calponin, vimentin, α-smooth muscle actin <strong>and</strong>thrombospondin, indicating that exposure to hypoxiaresulted in phenotype modulation to de-differentiatedstate <strong>and</strong> PKG may be involved in SMC phenotypemodulation. The results from PKG-specific smallinterfering RNA (siRNA) transfection <strong>and</strong> treatment withDT-3 (a peptide inhibitor of PKG, 30μM) in FPVSMCindicate that the expression of SMC phenotype proteinexpression decreased by hypoxia was also similarlyimpacted by PKG inhibition. Over-expression of PKG inFPVSMC by transfection of FPVSMC with a full lengthPKG construct tagged with GFP (PKG-GFP) reversed theeffect of hypoxia on the expression of SMC phenotypemarker proteins. These results suggest that PKG could beone of the determinants for the expression of SMCphenotype maker proteins <strong>and</strong> may be involved in themaintenance of the differentiated phenotype in pulmonaryvascular SMCs in hypoxia.NotesPage 101


Abstract43Human fetal cardiovascular baro-reflex responses in maternalsteroid administration analyzed by pulsed Doppler minutetissue displacement measurementNorio SHINOZUKADepartment of Obstetrics <strong>and</strong> Gynecology, <strong>Japan</strong> Red Cross Medical CenterObjectivesInvestigation of the cardiovascular baro-refelx response inhuman fetus has been methodologically limited. Dopplerminute tissue displacement measurement, we devised,enables to measure fetal aortic pulses at microns order.To clarify fetal baro-reflex changes in human fetus, thechanges in aortic pulse pressure amplitude <strong>and</strong> heart rateare analyzed in the fetus treated with trans-maternalsteroid administrationMethodsHardware: We developed the multi-channel Dopplerdevice with high accuracy tissue displacementmeasurement system that enabled to calculate minutetissue displacement at the order of micrometer. A ColorDoppler Ultrasound machine(Mochida SonoColor) wasmodified to h<strong>and</strong>le multi-depth Doppler raw signals withdigital quadrature detector. This system can measure 32channels of tissue Doppler signals at 1.5mm intervalstoward the beam direction simultaneously (Freq.5MHz:Rep.Freq:1.6kHz SampleFreq.400Hz).Measurements: Twelve fetuses treated with antenataltrans-maternal steroid administration(range26w0d-32w0d)were analyzedProtocol of trans-maternal steroid administration :Dexamethasone 12mg x 2<strong>Fetal</strong> descending aortic wall pulse wave was measured atpre-steroid <strong>and</strong> 24h after steroid administrationThe amplitude of aortic wall pulse wave (AWPW) <strong>and</strong>heart rate were calculated (60 second data obtained atquiet state )Fig 1ResultsThe baseline FHR was decreased from 154±10 to144±10bpm (p < 0.05) <strong>and</strong> AWPW amplitude wasincreased from 579±123 to 890±174μm (p


EFFECTS OF MATERNAL BETAMETHASONE ADMINISTRATIONON REGIONAL BLOOD FLOW IN THE INTRAUTERINE GROWTHRESTRICTED (IUGR) OVINE FETUSSupramaniam VG 1 , Jenkin G 1 , Wallace EM 2 , Miller SL 3Monash Immunology & Stem Cell Laboratories, Departments of Obstetrics <strong>and</strong> Gynaecology 2<strong>and</strong> Physiology 3 Monash University, Victoria, 3800, AustraliaAbstract44Glucocorticoid administration to pregnant women at riskof delivering preterm has beneficial effects on fetal lungmaturation. However, glucocorticoids may also causenon-pulmonary effects, particularly on the fetalcardiovascular system. These effects may beexacerbated in IUGR fetuses. The aim of this study wasto investigate the effect of maternal betamethasoneadministration on regional blood flow in the IUGR fetus.Methods:Pregnant sheep carrying twins underwent surgery at105-110 days gestation. Both twins were instrumentedwith catheters in the carotid artery, <strong>and</strong> femoral artery<strong>and</strong> vein. Single umbilical artery ligation (SUAL) wasperformed in one twin to induce IUGR. On day 5 aftersurgery, betamethasone (11.4mg) was administered imto the ewe (BM1) <strong>and</strong> repeated 24 hours later (BM2). Toassess blood flow, coloured microspheres were injectedinto the fetal venous circulation at time points relative toBM1: -24h, -1h, +5h, +11h <strong>and</strong> +33h. Animals weresacrificed after BM2 injection.Results:SUAL fetuses weighed less than the controls(1.4±0.01kg versus 1.69 ± 0.03kg) <strong>and</strong> had a greaterbrain-to-body weight ratio (22.64±1.19 versus17.83±1.75), indicative of brain sparing. Mean basalblood flow to the brain was 900ml/min/100g in controls<strong>and</strong> 740ml/min/100g in SUAL fetuses. At +5h therewas a 20% fall in total brain blood flow in SUAL fetuses<strong>and</strong> a 7% fall in controls. However, at +33h blood flowincreased by 97% (SUAL) <strong>and</strong> 21% (control). The trendfor a decrease in blood flow at +5h was observed in allbrain regions examined; i.e. white matter, grey matter,hippocampus, thalamus, brainstem <strong>and</strong> cerebellum. Inthe SUAL fetuses only, regional blood flow wasincreased at +33 h (11h after BM2) in all areas, rangingfrom 60% to 110% above basal levels. There weresignificant differences between control <strong>and</strong> SUALfetuses in blood flow in the heart, lungs <strong>and</strong>placentomes.Conclusions:Blood flow to the brain of SUAL fetuses, in response tobetamethasone, differs from that of control fetuses, Thesignificant reperfusion after a second BM administrationin all regions may be detrimental to the alreadycompromised fetal brain.NotesPage 103


Abstract45A Novel extraction method of fetal electrocardiogram from thecomposite abdominal signal.Yoshitaka Kimura *1 ,Takuya Ito *1 , Michiyo Nakamura *2 , Kaori Uchida *2 ,Hiroshi Chisaka *2 ,Norihiro Katayama, Mitsuyuki Nakao <strong>and</strong> KunihiroOkamura *2*1 Tohoku University Biomedical Engineering Research Organization*2 Department of Gynecology <strong>and</strong> Obstetrics, Tohoku UniversityGraduate School of MedicineIn contrast to the ultrasound measurement of fetal heartmotion , the electrocardiogram (ECG) provides clinicallysignificant information concerning the electrophysiologicalstate of a fetus. The problem of the fetal ECG extractionfrom maternal skin electrode signals was not only thesevere noise contained in the measured signal but alsothe non-linear property of fetal or maternal ECG itself. Inthis paper, a novel extraction method of fetal ECG fromabdominal composite signals is proposed. This methodconsists of the cancellation of the mother’s ECGcomponents <strong>and</strong> blind source separation with referencesignal (BSSR). The BSSR is a fixed-point algorithm theLagrange function of which includes the higher ordercross-co-relation between the extracted signal <strong>and</strong> thereference signal as the cost term rather than a constraint.By practical application, the proposed method has beenshown to be able to extract the P <strong>and</strong> T waves in additionto the R wave. The reliability <strong>and</strong> accuracy of this methodwas confirmed by comparing the extracted signals withthe directly recorded ECG at the second stage of labor.The gestational age dependency of the physiologicalparameters of the extracted fetal ECG also coincided wellwith that of the magneto-cardiogram, which proves theclinical applicability of this proposed method.NotesPage 104


AbstractThe effects of the tocolytics atosiban <strong>and</strong> nifedipine on fetalmovements, heart rate, <strong>and</strong> blood flow.46Roel de Heus, MD, Eduard J.H. Mulder, MSc, PhD, Jan B. Derks, MD, PhD,Gerard H.A. Visser, MD, PhDDepartment of Woman <strong>and</strong> Baby, University Medical Centre Utrecht, The Netherl<strong>and</strong>sObjective: to study the direct fetal effects of tocolysiswith atosiban or nifedipine combined with a course ofbetamethasone.Methods Women with preterm labour (n=40), gestationalage between 25 <strong>and</strong> 33 weeks <strong>and</strong> no previous tocolytictreatment, were prospectively r<strong>and</strong>omised for treatmentwith atosiban or nifedipine combined with a course ofbetamethasone. One-hour recordings of fetal heart rate(FHR) <strong>and</strong> its variation <strong>and</strong> fetal movements were madeon each of five successive days (day0- day4) starting inthe early morning. <strong>Fetal</strong> blood flow velocity patterns werestudied daily by Doppler ultrasound.Main outcome measures: Primary outcome measureswere effects on FHR <strong>and</strong> its variation, short term variation(STV). Secondary endpoints were effects on fetalmovement <strong>and</strong> fetal blood flow parameters, umbilicalartery (UA) <strong>and</strong> medial cerebral artery (MCA).Results: Baseline characteristics of 31 women notdelivered at day 0 <strong>and</strong> no escape tocolysis, did not differbetween the study groups. Multilevel analysis showed nosignificantly contribution of either tocolytic to the models ofFHR parameters <strong>and</strong> fetal movement parameters. Therewas a strong significant time course in basal FHR <strong>and</strong>STV during day 0 to 4 of the morning recordings.Compared to baseline (day 0) a significant rise in STVoccurred on day 1 (p=0.01). Basal FHR decreasedsignificantly on day 1 (p< 0.01) <strong>and</strong> day 2 (p< 0.01)compared to baseline (day 0). The use of tocolytics didnot contributed significantly to the models of the UApulsatility index (PI) (p=0.91) <strong>and</strong> MCA PI (p=0.62). Nosignificant changes in time course of PI parameters werefound.Conclusion This study demonstrates for the first time thedirect effects of atosiban on fetal movement, fetal heartrate <strong>and</strong> fetal blood flow. Tocolysis with either atosiban ornifedipine combined with betamethasone administrationappears to have no adverse direct adverse fetal effects.Figure. Mean morning values ± SEM of the study parameters. Open circles represent women treated withatosiban <strong>and</strong> closed circles women treated with nifedipine. The first arrow indicates start of tocolytic treatment<strong>and</strong> betamethasone administration. The second arrow indicates the second dose betamethasone.NotesPage 105


Abstract47THE EFFECT OF SULPHASALAZINE ON LPS-INDUCEDCHANGES IN FETAL OXYGENATION AND CAROTID BLOODFLOWBurcu Saglam 1 , Graham Jenkin 2 , Suzanne L. Miller 3 , <strong>and</strong> Euan M. Wallace 11 Department of Obstetrics <strong>and</strong> Gynaecology, 2 Monash Immunology <strong>and</strong> Stem Cell Laboratories<strong>and</strong> 3 Deparment of Physiology, Monash University, Victoria, Australia.Background:Intrauterine infection is associated with fetal brain injurythought to be caused, at least in part, by the resultinginflammatory response. However, infection is alsoassociated with fetal hypoxia <strong>and</strong> it is possible that thiscontributes to the resulting brain injury, either directly onvia changes in cerebral blood flow <strong>and</strong> subsequentoxidative stress. We have used an ovine model of pretermfetal infection, using exposure to lipopolysaccharide (LPS),to investigate the effect of the administration ofsulfasalazine (SSZ), an NFκB inhibitor, on LPS-inducedchanges in fetal cerebral blood flow.Methods:Seven twin-pregnant ewes were r<strong>and</strong>omly assigned toreceive either SSZ (n=3) or placebo (n=4) orally. Surgerywas performed at ~105 days gestation for placement of afemoral artery <strong>and</strong> vein catheter <strong>and</strong> a carotid artery flowprobe into each fetus. In each twin pair, whether SSZ orplacebo, one fetus received three daily i.v. infusions ofLPS (100ng/kg) <strong>and</strong> the co-twin received the samevolume of saline. Carotid blood flow, fetal blood gases <strong>and</strong>pH were monitored daily.Results:(1) The administration of SSZ was associated with analmost two-fold increase in baseline fetal carotid bloodflow in both saline <strong>and</strong> LPS-adminsitered groups. (2) Theadministration of LPS was associated with fetal acidaemia,hypoxaemia <strong>and</strong> hypercapnia within 4-6 hours in fetusesof both placebo <strong>and</strong> SSZ ewes. In the fetuses of ewespre-treated with SSZ the degree of these changes wassignificantly worse than in fetuses of placebo treated ewes.(3) LPS administration was associated with significantincreases in fetal carotid blood flow, by 70% in placebotreated ewes <strong>and</strong> by 85% in SSZ treated ewes. Thisresponse in fetal carotid blood flow to LPS was similar inthe two groups.Conclusion:These preliminary observations suggest that maternalSSZ administration alters fetal cerebrovascular function,which may be detrimental. Further, LPS administrationsignificantly increases fetal carotid blood flow – an effectnot mitigated by SSZ. Indeed, the maternal administrationof SSZ worsens the fetal acideaemic, hypoxaemic <strong>and</strong>hypercapnic response to LPS. Together, theseobservations suggest that SSZ would not be suitable as aanti-inflammatory in the management of intrauterineinfection.<strong>Fetal</strong> Carotid Blood Flow<strong>Fetal</strong> carotid blood flow (ml/min)10080604020LPS #1LPS #2 LPS #3SSZ+LPSPlacebo+LPSSSZ-SalineSSZ-Placebo0-12 0 12 24 36 48 60 72Time (hours)NotesPage 106


Poster1The role of the neural sympathetic <strong>and</strong> parasympatheticsystems in diurnal <strong>and</strong> sleep state cardiovascular rhythms inthe late gestation ovine fetusEllen C. Jensen, Laura Bennet, Sarah-Jane Guild, Lindsea C. Booth, JennyA. Westgate, <strong>and</strong> Alistair J. GunnDepartment of Physiology, Faculty of Medical <strong>and</strong> Health Sciences, The University of Auckl<strong>and</strong>,Auckl<strong>and</strong>, New Zeal<strong>and</strong>Diurnal <strong>and</strong> sleep state rhythms for fetal heart rate (FHR)<strong>and</strong> FHR variability (FHRV) are well established by lategestation in mammalian species. However, despite thecommon use of FHR <strong>and</strong> FHRV for fetal surveillance, theunderlying efferent mechanisms controlling these rhythmsremain incompletely understood. In the present study weevaluated the contributions of the neural parasympathetic<strong>and</strong> sympathetic nervous systems in chronicallyinstrumented fetal sheep at a mean (SE) of 121.6 (0.9)days gestation.Eight fetuses underwent chemical sympathectomy using6-hydroxydopamine the day after surgery, 8 fetusesunderwent vagotomy at surgery <strong>and</strong> 8 fetuses were shamcontrols. Three days after surgery FHR, FHRV, meanarterial blood pressure (MAP), carotid blood flow, <strong>and</strong>electroencephalogram (EEG) activity were measuredcontinuously for 24 hours. Changes between sleep states,i.e. high voltage slow activity <strong>and</strong> low voltage fast activity,were determined in a 6 h interval.Control fetal sheep showed consistent diurnal rhythms inFHR <strong>and</strong> FHRV, with maximal activity close to midnight.Both the sympathectomy <strong>and</strong> vagotomy groups alsoshowed diurnal rhythms in FHR <strong>and</strong> FHRV, butsympathectomy shifted the peak in FHR to much later(0500 h, p


Np95, a master regulator of the epigenome, is essentiallyrequired for embryonic development2Sharif J. 1,2 , Muto M. 3 , Nakamura M. 2 , Takebayashi S. 4 , Okano M. 4 , KosekiH. 3 , Nagamune T. 1 , Mitsuya K. 5 <strong>and</strong> Okamura K. 21 Department of Chemistry & Biotechnology, School of Engineering, The University of Tokyo,Tokyo, <strong>Japan</strong>, 2 Department of Obstetrics & Gynecology, Tohoku University School of Medicine,Sendai, Miyagi, <strong>Japan</strong>, 3 RIKEN Research Center for Allergy <strong>and</strong> Immunology, Yokohama,<strong>Japan</strong>, 4 RIKEN Center for Developmental Biology, Kobe, Hyogo, <strong>Japan</strong> <strong>and</strong> 5 TohokuUniversity Biomedical Engineering Research Organization, Sendai, Miyagi, <strong>Japan</strong>PosterAIMSAcquirement <strong>and</strong> maintenance of a correct epigeneticstate of the genome is indispensable for biologicalprocesses such as germ cell development, fertilization<strong>and</strong> embryogenesis. The insights gained from assistedreproductive technology (ART) <strong>and</strong> somatic cell nucleartransfer clearly indicate the importance of epigenetics inembryonic <strong>and</strong> postnatal development. We havepreviously identified a novel epigenetic regulatory protein,Np95. In this report, we discuss the role of Np95 inembryogenesis <strong>and</strong> mammalian development.METHODSWe have studied genomewide DNA methylation level inthe Np95 KO ES cells by bisulphite DNA conversiontechniques. We generated Np95-null homozygoticembryos (B6 x JF1 cross) <strong>and</strong> examined any possibleimprinting defects. We investigated the expression ofrepeat associated transcripts in Np95 mutant embryos byquantitative real-time PCR. We also examined cell cycledependent cellular localization of Np95 by immunostaining.RESULTSDigestion with methylation specific enzymes revealed thatNp95 KO ES cells are severely hypomethylated. TheNp95-null homozygotic embryos were found to beembryonic lethal at d9.5. IAP, LINE <strong>and</strong> otherheterochromatin associated repeat elements constitutemore than 40% of the mouse genome. In Np95 mutantembryos, IAP, LINE L1 <strong>and</strong> SINE B1 transcripts weredramatically upregulated. This suggested that Np95 isessential for heterochromatin regulation. To examine therole of Np95 in modulation of single copy genes in theeuchromatin, we studied the imprinted H19/Igf2 <strong>and</strong>Lit1/Kcnq1ot1 loci. We found that in both these lociparent-of-origin specific expression of H19 <strong>and</strong> Lit1 waslost. The adjacent Igf2 <strong>and</strong> Cdkn1c genes werecompletely repressed. Loss of imprinting was associatedwith severe loss of CpG methylation in the H19-DMD <strong>and</strong>Dlk1/Gtl2 IG-DMR <strong>and</strong> the histone modifications in theseloci were also affected. These results indicated that Np95is essential to regulate the euchromatin along with theheterochromatin. Coimmunoprecipitation <strong>and</strong>immunostaining analyses revealed that Np95 physicallyinteracts with Dnmt1 during the middle-S1 phase <strong>and</strong>suggested a possible mechanism that involved Dnmt1.CONCLUSIONWe show that Np95 acts as a master regulator of themammalian epigenome by regulating both theheterochromatin <strong>and</strong> euchromatin compartments of thegenome. We also describe an essential role for Np95 inmammalian development.NotesPage 109


Poster3EVALUATION OF THE PLACENTAL VASCULARITY USINGFRACTAL PARAMETERS: COULD THEY HELP IN EARLYDIAGNOSIS OF FETAL GROWTH RESTRICTION?C. Guiot, E. Piccoli^, F. Saccom<strong>and</strong>i & T. Todros^Dept Neuroscience, University of Torino, Torino, Italy <strong>and</strong> ^Dept of Gynecologic <strong>and</strong> ObstetricDisc., University of Torino, Torino, ItalyPlacental vascularization has been extensivelyinvestigated in the past (Giles et al, 1985, Salafia et al,1997) . Most of the studies referred to villous arteries <strong>and</strong>arterioles, <strong>and</strong> compared placentas from ‘normal’,uneventful pregnancies at term with cases of <strong>Fetal</strong> GrowthRestrictions (FGR) placentas. Studies of the placentalcapillaries were also performed. (Mayhew et al, 2004,Macara et al, 1996; Krebs et al, 1996; Todros et al, 1999).Results were conflicting.All the previous studies were performed by countingmethods, i.e. based only on the comparison of thevascular density evaluated on placental samples, <strong>and</strong>required a certain amount of placental tissue. Otherparameters, such as the ‘fractal dimension D <strong>and</strong> thelacunarity L , were proposed to evaluate thecharacteristics of the vascular networks, i.e. their fractalproperties, which are expected to portrait some specific,intrinsic characteristics of the system (.M<strong>and</strong>elbrot 1967,1982). The main advantage of such parameters is thatthey are invariant in self-similar structures, i.e. D has thesame value independently of the number of generations ofbranches developed by the tree.Up to now only one paper related to the placenta <strong>and</strong>using fractal parameters was published.( Bergman & Ullberg, 1998), confirming the self-similarityof the placental vascular tree.In the present study we investigate placentas deliveredfrom normal <strong>and</strong> FGR pregnancies. Biopsy on placentaltissue fixed <strong>and</strong> paraffin included after performing aimmunoistochemical reaction with anto-CD31, which is amarker specific for vascular endothelium, are investigatedat magnification x40 (Neurolucida, MBF Bioscience), <strong>and</strong>vessels up to 2-5 microns can be observed, includingplacental capillaries. D <strong>and</strong> L are estimated using Fraclac,a downloadable computing code (http://rsb.info.nih.gov/ij/)on the 2D specimen (a 3D extension is already understudy) .Results show that, although in a very narrow range,the parameter values are related to fetal growth. Theyencourage further investigation aimed at proposing a newtool for early detection of FGR on histologic probes fromchorial villi samples withdrown for genetic tests.NotesPage 110


<strong>Fetal</strong> <strong>and</strong> perinatal growth curves: what can we learn fromthem?4Caterina Guiot, Tullia Todros ^, Antonio Gliozzi * , Pier Paolo Delsanto*Dept Neuroscience <strong>and</strong> ^ Dept Gynecol Obstet Disciplines, University of Torino, Torino,Italy*Dept Physics, Politecnico of Torino, Torino, ItalyPosterMany different mathematical models, based onobservational datasets, have been proposed for decades.Various parameters X k ( mass, height, long bones length,biparietal diameter, etc) have been investigated bothlongitudinally, i.e. on single individuals, <strong>and</strong> transversally,i.e., averaged over a given homogeneous population.According to a recent formalism [1] developed on thebasis of the ontogenic growth model by West <strong>and</strong>collaborators [2], fetal <strong>and</strong> perinatal datasets can beclassified <strong>and</strong> fitted in the framework of a completelygeneral scheme. As Fig 1 shows, plotting the logarithmicgrowth rate ( a = d ln X k / dt ) vs. its time derivative ( b=da/dt ) , curves are obtained which result from acombination of a polynomial expansion <strong>and</strong> complete orpartial loops. The former account for an overall growthterm as predicted by the classification scheme of ref. [1],while the latter are due to intrinsic cyclicities in the growthprocess. They are usually related to various feedbackmechanisms, as already observed qualitatively [3] , <strong>and</strong>possibly due to adaptation to the surrounding environment,hormonal control cycles, etc.A thorough interpretation of plots such as in Fig. 1, canlead to a deeper underst<strong>and</strong>ing of the variousmechanisms involved. In addition, the model could beutilized to discriminate abnormal growth processes dueto intrinsic (e.g. genetic) factors from the ones resultingfrom a mismatch with environmental conditions.[1] P. Castorina, P.P. Delsanto, C. Guiot, Phys Rev Letter 96,188701 (2006)[2] G. B. West, J. H. Brown, B. J. Enquist, Nature 413, 628 (2001)[3] C.B. Davenport. J Gen Physiol 10,205 (1926)NotesPage 111


Poster5Nitric Oxide <strong>and</strong> CD4+25+Regulatory T cell production fromiNOS knockout mice <strong>and</strong> their F1 mice treated with LPS.Hidenori Takahashi, Toshiaki Okawa, Kiya Fujimori, Akira SatoFukushima Medical University, Ob/Gyn, Fukushima, <strong>Japan</strong>,OBJECTIVE: To evaluate the effect of inherited iNOS onNitric Oxide (NO) production by placental tissue <strong>and</strong>CD4+25+ regulatory T cell (Treg) from spleen by using ofiNOS knockout (iNOSKO: iNOS-/-) mice <strong>and</strong> their F1 femalemice half inherited iNOS(paternal(+p) or maternal(+m))were treated with lipopolysaccharide (LPS).STUDY DESIGN: Pregnant mice with genetic compositionof placenta for mating as (A) male iNOSKO× femaleiNOSKO; iNOS-/-, (B) male iNOSKO × femaleC3H(iNOS+/+) ; iNOS-/+m, (C) male C3H×femaleiNOSKO; iNOS+p/- day 14 of gestation were sacrificed 6hours after intraperitoneal injection(i.p.) of LPS (400 μg/kg) or vehicle (n=8 per group). Uterine rings from eachmice were equilibrated in Krebs-Henseleit solution. Insome rings, a piece of placenta was left attached to theuterine wall . After 60 min incubation one ml sample of thesolution was taken <strong>and</strong> analyzed for NOx production.Their F1 female mice were sacrificed 6 hours after LPSi.p.(400 μg/ kg). Their blood serum were analyzed for NOx<strong>and</strong> mononuclear cells isolated from spleen were for Tergby FACS. Statistical analysis was performed using oneway ANOVA.RESULTS: (1)NOx Production of uterine rings attachedwith 3 types placenta: Increasing order of magnitude ofNOx production without LPS; (A) iNOS(-/-) =(C)iNOS(-/+m)< (B) iNOS(+p/-) (p


Nitric Oxide production in placental tissue from iNOS knockoutmice with LPS <strong>and</strong> their offsprings.6Hidenori Takahashi, Toshiaki Okawa, Kiya Fujimori, Akira SatoFukushima Medical University, Ob/Gyn, Fukushima, <strong>Japan</strong>,PosterOBJECTIVE: To evaluate the effect of inherited iNOS onNitric Oxide (NO) production by placental tissue by usingof iNOS knockout (iNOSKO: iNOS-/-) mice <strong>and</strong> their F1female mice half inherited iNOS(paternal(+p) ormaternal(+m)) were treated with lipopolysaccharide(LPS).STUDY DESIGN: Pregnant mice with genetic compositionof placenta for mating as (A) male iNOSKO× femaleiNOSKO; iNOS-/-, (B) male iNOSKO × femaleC3H(iNOS+/+) ; iNOS-/+m, (C) male C3H×femaleiNOSKO; iNOS+p/- day 14 of gestation were sacrificed 6hours after intraperitoneal injection(i.p.) of LPS (400 μg/kg) or vehicle (n=8 per group). Uterine rings from eachmice were equilibrated in Krebs-Henseleit solution. Insome rings, a piece of placenta was left attached to theuterine wall . After 60 min incubation one ml sample of thesolution was taken <strong>and</strong> analyzed for NOx production.Their F1 female mice were sacrificed 6 hours after LPSi.p.(400 μg/ kg). Their blood serum were analyzed for NOx.Statistical analysis was performed using one way ANOVA.RESULTS: (1)NOx Production of uterine rings attachedwith 3 types placenta: Increasing order of magnitude ofNOx production without LPS; (A) iNOS(-/-) =(C)iNOS(-/+m)< (B) iNOS(+p/-) (p


Poster7Maternal severe undernutrition during both late gestation <strong>and</strong>lactation period induce hypertension in male rat offspring.Hidenori Takahashi, Toshiaki Okawa, Keiya Fujimori, Akira SatoDepartment of Obstetrics <strong>and</strong> Gynecology Fukushima Medical University School of Medicine,FukushimaOBJECTIVE:Our objective was to investigate the effects of eithersevere undernutrition during late gestation or lactationperiod on blood pressure <strong>and</strong> the development of vascularfunction in male rat offspring.STUDY DESIGN:We use normal pregnant Wistar rats (Group A),nutritionally restricted by feeding with 30% of the normalgestation-matched dietary intake from day 17 of gestationto delivery (Group B) <strong>and</strong> 30% restricted after delivery tothe end of lactation period (Group C).The offspring was measured blood pressure at 12 <strong>and</strong> 24weeks by using indirect tail-cuff method.Rings of thoracic aorta with intact endothelium from themale offspring of A <strong>and</strong> B at 8 weeks, were equilibrated at2 g passive tension in organ chambers filled withKrebs-Henseleit solution. Concentration-responserelationships to Norepinephrine (NE) <strong>and</strong> angiotensinⅡ(ATⅡ) were obtained in the absence or presence ofN(omega)-nitro-L-arginine methyl ester (L-NAME) or aselective AT Ⅱ type-1 receptor blocker (Valsartan).Responses to cumulative concentrations of sodiumnitroprusside (SNP) <strong>and</strong> to 10-5M oxyhemoglobin (Hb,nitric oxide scavenger) were also determined.RESULTS: Body weight was significantly reduced in Boffspring compared to A <strong>and</strong> C in male offspring at day 1(p C >A.NE concentration-dependently stimulated tension ofaortic rings from in A <strong>and</strong> B offspring, which was notsignificantly (n=6). Maximal contractions to NE weresignificantly stimulated by L-NAME in A (p


Regulation of maternal feeding during lactation period maycontrol adulthood hypertension.8Hidenori Takahashi, Toshiaki Okawa, Keiya Fujimori, Akira SatoDepartment of Obstetrics <strong>and</strong> Gynecology Fukushima Medical University School of Medicine,FukushimaPosterOBJECTIVE:Exposure to undernutrition during fetal life has beenproposed as an underlying cause of adult hypertension,but the effect of either high fat nourishment orundernutrition during lactation period on blood pressure isunclear.Our objective was to investigate the most effectivematernal nourishment <strong>and</strong> feeding period for offspringinduced adulthood hypertension in using high-fat diet(HFD).STUDY DESIGN:We use 5 types pregnant Wistar rats as fed with normalnutrition (Group A), nutritionally restricted by feeding with30% of the normal gestation-matched dietary intake fromday 17 of gestation to delivery (Group B), 30% restrictedafter delivery to the end of lactation period (Group C), witha high fat diet (HFD) during gestation to lactation period(Group D) <strong>and</strong> with HFD nutritionally 30% restricted fromthe day of delivery to the end of lactation period (GroupE).The offspring was measured Body Weight (BW) <strong>and</strong>measured blood pressure at 12, 24 <strong>and</strong> 60 weeks byusing indirect tail-cuff method. Statically analysis wasperformed using one-way ANNOVA.RESULTS: BW was significantly reduced in B offspringcompared to another (A, C, D, E) male offspring at day 1(p B > C>A. (pD>E>C>A. (p


Poster9Maternal regulation of high fat nourishment during lactationperiod reduce a hypertension of male offspringHidenori Takahashi, Toshiaki Okawa, Keiya Fujimori, Akira SatoDepartment of Obstetrics <strong>and</strong> Gynecology Fukushima Medical University School of Medicine,FukushimaOBJECTIVE:Exposure to undernutrition or high fat nourishment duringfetal life has been proposed as an underlying cause ofadult hypertension, but the effect of maternal feedingregulation during lactation period on blood pressure ofoffspring is unclear.Our objective was to investigate the effects of eitherhigh-fat diet (HFD) during gestation to lactation period orrestrictive fed a HFD during lactation period on bloodpressure in male rat offspring.STUDY DESIGN:We use 3 types pregnant Wistar rats as fed with normalnutrition (Group A), with a high fat diet (HFD) duringgestation to lactation period (Group B) <strong>and</strong> with HFDnutritionally restricted by feeding with 30% of the normallactation-matched dietary intake from the day of deliveryto the end of lactation period (Group C).The male offspring was measured blood pressure at 12,24 <strong>and</strong> 60 weeks by using indirect tail-cuff method.Statically analysis was performed using one-wayANNOVA.RESULTS: Body weight was significantly reduced in Coffspring compared to A <strong>and</strong> B in male offspring at day 28after delivery (p C>A. (p


PosterEpigenetic stability <strong>and</strong> developmental plasticity in clonedplacentae during somatic cell nuclear transfer10Naomi Matsuda 1 , Jafar Sharif 1 , Kimiko Inoue 2 , Narumi Ogonuki 2 , HiromiMiki 2 , Hiroshi Chisaka 1 , Kunihiro Okamura 1 , Atsuo Ogura 2 , <strong>and</strong> KohzohMitsuya 31Department of Obstetrics <strong>and</strong> Gynecology, Tohoku University School of Medicine, 1-1Seiryomachi, Aobaku, Sendai 980-8574, <strong>Japan</strong>2Bioresource Engineering Division, RIKEN Bioresource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki305-0074, <strong>Japan</strong>3Biofunctional Science, Tohoku University Biomedical Engineering Research Organization(TUBERO), 2-1 Seiryomachi, Aobaku, Sendai 980-8575, <strong>Japan</strong>Early developmental process largely involves epigeneticgene regulation. One of the most remarkablereprogramming of the epigenome can be seen duringsomatic cell nuclear transfer (SCNT) whereby the nucleusfrom a differentiated cell could be reset by oocytecytoplasm, resulting in the production of live offspring. Inthis study, we have focused on the epigenetic aspects ofthe placentae derived from cloned mice, in which morethan 3-fold enlargement was observed in placental weightat term. All the twelve cloned placentae derived fromcloned conceptuses exhibit loss of genomic imprintingwhich is closely linked to histone tail modificationsindependent of DNA methylation, implying a possible rolefor histone modification remodeling in nuclearreprogramming. Moreover, we have identified multipletranscripts that are repressed specifically in the clonedplacentae irrespective of the degree of enlargement <strong>and</strong>of the presence of embryo proper. It has also been evidentthat the imprinted Dlk1 transcripts were less abundant inthe cloned placentae that were delivered with fetus butwere extensively silenced in all the four placentaerecovered without fetus. Our results contrast with theprevious observations indicating the variegated epigeneticanomalies in cloned animals <strong>and</strong> highlight considerablestability of the epigenome during SCNT <strong>and</strong>developmental plasticity throughout early embryogenesis<strong>and</strong> placentation.NotesPage 117


Poster11Pre-existing hypoxia is associated with a delayed but moresustained rise in T/QRS ratio during prolonged umbilical cordocclusion in near-term fetal sheepLaura Bennet, Bert Wibbens, Jenny A. Westgate, Harmen H. De Haan,Guido Wassink, <strong>and</strong> Alistair J.Gunn <strong>Fetal</strong> Physiology <strong>and</strong> Neuroscience Group, Dept of Physiology, The University ofAuckl<strong>and</strong>, Auckl<strong>and</strong>, New Zeal<strong>and</strong>.There is limited information on whether pre-existing fetalhypoxia alters hemodynamic responses <strong>and</strong> changes inT/QRS ratio <strong>and</strong> ST waveform shape during subsequentsevere asphyxia. Chronically instrumented near-termsheep fetuses (124±1 days) were identified as eithernormoxic PaO 2 > 17 mmHg (n=9) or hypoxic PaO 2 ≤ 17mmHg (n=5), <strong>and</strong> then received complete occlusion of theumbilical cord for 15-min. Umbilical cord occlusion lead tosustained bradycardia, severe acidosis <strong>and</strong> transienthypertension followed by profound hypotension in bothgroups. Pre-existing hypoxia did not affect changes inmean arterial blood pressure, but was associated with amore rapid initial fall in femoral blood flow <strong>and</strong> vascularconductance <strong>and</strong> with transiently higher fetal heart rate at2 minutes <strong>and</strong> from 9 to 11 minutes of occlusioncompared with previously normoxic fetuses. Occlusionwas associated with a significant but transient rise inT/QRS ratio; pre-existing hypoxia was associated with asignificant delay in this rise (maxima 3.7 ± 0.4 vs. 6.2 ± 0.5minutes), but a slower rate of fall. There was a similarelevation in troponin-T levels 6 hours after occlusion in thetwo groups (median (range) 0.43 (0.08, 1.32) vs 0.55(0.16, 2.32) µg/L, N.S.). In conclusion, mild pre-existinghypoxia in normally grown singleton fetal sheep isassociated with enhanced centralisation of circulationafter umbilical cord occlusion <strong>and</strong> delayed elevation of theST waveform <strong>and</strong> slower fall, suggesting that chronichypoxia improves myocardial dynamics during asphyxia.NotesPage 118


Poster<strong>Fetal</strong> brain, liver <strong>and</strong> renal blood flow assessment using 3Dpower Doppler ultrasound12Fong-Ming Chang, Chiung-Hsin Chang, Chen-Hsiang Yu, Lin Kang,Pei-Ying Tsai <strong>and</strong> Huei-Chen Ko*Department of Obstetrics <strong>and</strong> Gynecology, <strong>and</strong> Research Institute of Behavioral Medicine*,National Cheng Kung University Hospital, Tainan 70428 TaiwanPurpose: To assess the blood flow of fetal brain, liver,kidney at second <strong>and</strong> third trimesters in normal pregnancyusing 3D power Doppler ultrasound.Materials <strong>and</strong> Methods: We underwent a series ofprospective, cross-sectional, consecutive <strong>and</strong> r<strong>and</strong>omizedstudies to assess the fetal organ blood flow in normalsingleton fetuses from 20 to 40 weeks of gestation at theultrasound lab of a tertiary medical center. All the mothers<strong>and</strong> fetuses are free of any medical or obstetricalcomplications. We assessed the vascularization index (VI),flow index (FI) <strong>and</strong> vascularization-flow index (VFI) of fetalbrain, liver <strong>and</strong> kidney using the quantitative 3D powerDoppler ultrasound. All the fetuses were followed todelivery to ensure they were born normal.Results: All the VI, FI <strong>and</strong> VFI of fetal brain, liver <strong>and</strong>kidney increased significantly with gestational age (GA).Using GA as the independent variable, <strong>and</strong> the VI, FI <strong>and</strong>VFI as the dependent variable, the linear regressionequations as well as the correlation coefficients for VI, FI<strong>and</strong> VFI of fetal brain, liver <strong>and</strong> kidney versus GA were allsignificant (all P


Poster13Molecular Genetic Study on Angiotensin-Converting EnzymeGene <strong>and</strong> Preeclampsia in Taiwanese:Two polymorphismsTested <strong>and</strong> as HaplotypesFong-Ming Chang , Ming-Hui Chen, Chiung-Hsin Chang, Chia-Fu Li * ,Chen-Hsiang Yu, <strong>and</strong> Huei-Chen Ko **Department of Obstetrics <strong>and</strong> Gynecology, National Cheng Kung University Medical College,Tainan, Taiwan, *831 Army Hospital, Pei-Tou, Taipei, Taiwan, <strong>and</strong> ** Research Institute ofBehavioral Medicine, National Cheng Kung University Medical College, Tainan, TaiwanPurpose: To test whether the angiotensin-convertingenzyme (ACE) gene polymorphisms are associated withpreeclampsia-eclampsia (PE).Method: The study included 99 patients with PE <strong>and</strong> 146healthy controls. From genomic DNA, two polymorphismsin genes for ACE were typed. Allelic frequencies werecompared between PE patients <strong>and</strong> controls. Forstatistical analysis, chi-squared tests were used for twopolymorphic sites <strong>and</strong> as haplotypesResults : The genotype frequencies of the 287 bpinsertion-deletion (I/D) polymorphic site at the intron 16 ofthe ACE gene in PE patients were different with those incontrols (χ 2 = 6.68, p=0.035). However, the allelefrequency of the 287 bp I/D polymorphic site at the intron16 was not different with controls. In addition, thegenotype frequencies <strong>and</strong> the allele frequencies of Pst Isite at intron 7 in PE patients were not different withcontrols. Furthermore, the haplotype frequencies of twopolymorphic sites were not different in PE patients <strong>and</strong>controls.Conclusion:Although significant difference exists in thegenotype frequencies of the 287 bp I/D polymorphic siteat the intron 16 of the ACE gene between PE patients <strong>and</strong>controls, no significant differences can be detected for allthe other tests, either as single locus or as haplotype. Wespeculate the disease-gene association between PE <strong>and</strong>the ACE gene in Taiwanese population. Further studiesare warranted.NotesPage 120


Clinical application of electromechanical delay time for indirectevaluation of fetal blood pressure during labor <strong>and</strong> delivery. Apreliminary study of 18 full-term infants.Yasuyuki Kawagoe, MD, Hiroshi Sameshima, MD, Tsuyomu Ikenoue, MD.Department of Obstetrics <strong>and</strong> Gynecology <strong>and</strong> Perinatal Center, Faculty of Medicine, Universityof Miyazaki, <strong>Japan</strong>.Poster14Objective: We previously showed a reciprocal relationshipbetween the electro-mechanical delay time <strong>and</strong> bloodpressure in fetal goat models. We applied this techniqueto clinical settings to determine whether the chronologicalchanges in delay time during labor <strong>and</strong> delivery correlatewith fetal heart rate monitoring patterns <strong>and</strong> acid-basestatus.Methods: Informed consent was obtained from 18 womenwith full-term, singleton pregnancy. Delay time wasobtained on a beat-by-beat basis from electrocardiogramto pulse oximeter waveform <strong>and</strong> averaged during eachbaseline period defined by the interpretation of fetal heartrate monitoring. According to the > 10% change ormore from the control value, chronological changes duringlabor <strong>and</strong> delivery were categorized into shortened,unchanged, <strong>and</strong> prolonged. Incidence of abnormal fetalheart rate monitoring patterns, hypoxemia, acidemia, lowApgar score was compared according to the chronologicalchanges in delay time.Results: Delay time was available in 82±11% of the time ofrecordings. Chronological changes were assessed in 15fetuses, 2 (13%) showed prolonged, 7 (47%) showedshortened, <strong>and</strong> 6 (40%) showed unchanged.Comparisons of the shortened <strong>and</strong> unchanged revealedthat severe variable deceleration was significantlyincreased <strong>and</strong> half or more fetuses showed mildlyhypoxemic in the shortened category.Conclusions: Shortening of delay time during delivery,theoretically indicating a hypertensive condition, wasmore likely associated with the presence of severevariable decelerations <strong>and</strong> fetal hypoxemia, suggestingthat the delay time may supplement the interpretation offetal heart rate monitoring.NotesPage 121


Poster15Temporal Changes in Mediators of Microvascular Tone areInfluenced by Sex <strong>and</strong> Glucocorticoid Exposure in PretermInfantsStark MJ, Clifton VL, Wright IMRMother <strong>and</strong> Babies Research Centre, Hunter Medical Research Institute, University ofNewcastle, NSW, AustraliaIan.Wright@hnehealth.nsw.gov.auAIM: Sexually dimorphic differences in microvascularfunction following preterm birth, with dysregulation ofvascular tone, may contribute to circulatory compromise<strong>and</strong> a male excess of morbidity <strong>and</strong> mortality. The fetusexhibits sex-specific differences in glucocorticoidmetabolism with downstream pathways involved in thecontrol of vascular tone. We hypothesized that therewould be temporal changes in local mediators ofmicrovascular tone <strong>and</strong> that these would be influenced bysex <strong>and</strong> antenatal steroid exposure in extremely preterminfants.METHOD: Forty-four infants (male: n=29, female: n=15)of 24-28 weeks gestation were studied at 24, 72, <strong>and</strong> 120hours of age. Venous endothelin-1 <strong>and</strong> urinary nitric oxidemetabolites <strong>and</strong> normetanephrine were determined byEIA. Venous haemoglobin-bound carbon monoxide(COHb) was determined by spectrophotometry. Data wasanalysed by repeated measures ANOVA.RESULTS: Endothelin-1 altered with time [F(2,30)=2.65,p=0.048] but not sex or steroid exposure. No temporal orsex-specific differences in urinary nitric oxide metaboliteswereobserved but levels were lower in infants exposed toantenatal betamethasone [F(1,15)=4.75, p=0.046].Urinary normetanephrine was lower in male infants[F(1,10)=11.03, p=0.008] <strong>and</strong> in those infants exposed toantenatal glucocorticoids [F(1,30)=2.51, p=0.041] but notemporal change was observed. Venous COHbdecreased with time [F(2,76)=18.9, p


The effect of carperitide, human atrial natriuretic peptide, onacute heart failure caused by endocardial cushion defect withatrioventricular block in a premature infant: a case reportYuichiro Miura, Tadashi Matsuda, Masaki Sato, Shizuko Akiyama, RyutaKitanishi, Takushi Hanita, Tatsuya Watanabe, Hidenobu OhtaDivision of Perinatal Medicine, Tohoku University Hospital, <strong>Japan</strong>Poster16[Introduction]Systemic vasodilator therapy has proved useful in thetreatment of low cardiac output in adult <strong>and</strong> child patients.Both carperitide <strong>and</strong> nitroglycerin have vasodilatingactions, which are exhibited by elevation of cyclicguanosine monophosphate. In addition to the vasodilatingaction, carperitide has more pathophysiological rolesincluding increase in the glomerular filtration rate <strong>and</strong>inhibition of the renin-angiotensin-aldosterone (RAA)system.We describe a premature infant with acute heart failuredue to complete atrioventricular block <strong>and</strong> atrioventricularregurgitation. In this case, elevation of the mean bloodpressure (MBP) <strong>and</strong> adequate diuresis were achievedwith carperitide while nitroglycerin was of no effect even athigh dose, indicating that carperitide effectively inhibitedthe accelerated RAA system, which nitroglycerin did not.[Case report]A 22-year-old woman was pointed out fetal heart anomalyat 27weeks of gestation. The fetal echocardiographyshowed complete endocardial cushion defect <strong>and</strong> completeatrioventricular block; the fetal heart rate was 50-60 bpm,<strong>and</strong> the cardio-thoracic area rate was 40.8%. Cesare<strong>and</strong>elivery was performed at 31 weeks <strong>and</strong> 1day of gestationbecause fetal pleural effusion <strong>and</strong> ascites appeared.A female infant weighing 2,182g was delivered. Theheart rate was 40-60 bpm <strong>and</strong> the MBP was as low as 23mmHg. External pacing at 120-140 bpm was introducedimmediately after birth. However, the MBP remained low<strong>and</strong> finally elevated to 31 mmHg after additionaladministrations of dopamine (10 µg/kg/min) <strong>and</strong>hydrocortisone (5 mg/kg), resulted in an increasing flow ofatrioventricular regurgitation. Nitroglycerin <strong>and</strong> diureticswere given for improvement of the regurgitation <strong>and</strong>oliguria but no significant effect was observed.After 30 hours of unsuccessful management forpersistent hypotension <strong>and</strong> hypoperfusion, carperitide(0.05 µg/kg/min) was administered. Immediately after theadministration, the decrease of the atrioventricularregurgitation <strong>and</strong> the increase of the diastolic flow velocityin the renal artery started to be detected by pulseddoppler echocardiography. By increasing carperitide up to0.20 µg/kg/min, appropriate MBP (>30 mmHg) <strong>and</strong>urination (>3 ml/kg/h) were achieved.[Comments]Carperitide has a potential effect not only as avasodilator but also as an inhibitor of RAA system in thetreatment for low cardiac output in premature infants.NotesPage 123


Poster17Effect of Maternal Stress on <strong>Fetal</strong> Heart Rate Assessed byVibroacoustic StimulationIkuko Makino 1) , Yoshio Matsuda 2) , Marie Yoneyama 2) , Kyoko Hirasawa 3) ,Koichiro Takagi 1) , Horoaki Ohta 2) , Yukuo Konishi 3)1) Depertment of Obstetrics <strong>and</strong> Gynecology Tokyo Medical University, Medical Center East2) Department of Obstetrics <strong>and</strong> Gynecology Tokyo Women’s Medical University3) Department of Infants’ Brain & Cognitive Development Tokyo Women’s Medical UniversityObjective: To determine whether maternal stress level,state <strong>and</strong> trait anxiety levels <strong>and</strong> stress hormones, affectfetal heart rate (FHR) pattern after vibroacousticstimulation (VAS) at 30 weeks of gestation.Materials <strong>and</strong> methods: Twenty-four healthy pregnantwomen with a singleton pregnancy were enrolled.Maternal anxiety was assessed using the SpielbergerState-Trait Anxiety Inventory (STAI). Women in the highanxiety groups had scores equal to or above the median.Corticotropin releasing hormone (CRH) <strong>and</strong>adrenocorticotrophic hormone (ACTH) in maternal plasma<strong>and</strong> cortisol <strong>and</strong> chromogranin A in saliva were measuredas maternal stress hormone. FHR pattern after VAS weredivided into three response types; type I: a long period ofacceleration, or one acceleration lasting > 1 minute or atleast two accelerations lasting > 15 seconds’ duration,type II: a biphasic response with acceleration followed bydeceleration, type III: no response or prolongeddeceleration.Results: Only of these hormones, the value of CRH in thehigh trait anxiety group was significantly higher than in thelow trait anxiety group. In pregnant women with high traitanxiety, the FHR response after VAS showed mostly atype II pattern rather than normal response as type I(p=0.036) <strong>and</strong> the mean amplitude of FHR accelerationafter VAS was significantly lower in the high trait anxietythan in the low trait anxiety group (p=0.012).Conclusions: These findings suggest that stress inpregnant women with high anxiety may influence the FHRpatterns after VAS. The maternal environment exertsinfluence on the fetal autonomic nervous system.NotesPage 124


PosterNon-Invasive long-term fetal ECG monitoring 18E.M. Graatsma, J.B.Derks, E. Mulder, G. VisserDept of Perinatology, University Medical Center Utrecht,The Netherl<strong>and</strong>sNon-invasive fetal heart rate (FHR) monitoring using thefetal ECG (fECG) signal as obtained from the maternalabdomen has several advantages compared to FHRmonitoring using ultrasound. Main advantage is itspotential of obtaining prolonged recordings. Previously,antepartum fECG-monitoring has been hampered bytechnical difficulties (poor signal pickup, <strong>and</strong> a decline insignal quality around the formation of the vernix caseosabetween GA 28-32 wks). Most technical problems appearto have been solved now.We report on our first experience with an updatedfECG-monitor that is currently being tested for clinical use.We hypothesized that recording quality would be betterduring the night.abdominal electrodes. Recordings were consideredsuccessful if data loss was limited to a maximum of 60%).When total recording time (5pm-8am) was considered,62/75 (83%) recordings were successful. The number ofsuccessful recordings increased to 68/75 (91%) when thenight part (11pm-7am) was analyzed separately. Mean SRfor total recording time was 75%, but was higher overnight(85%).SR declined transiently between 26-34 wks, which may bedue to the vernix formation. Fortunately, despite theoverall decline in SR at this age range, the overnight SRwas sufficient to perform numerical FHR analysis.fECG recordings were made in 75 women at GA 20-39wks with singleton pregnancies between 5pm <strong>and</strong> 8am(total duration 15h) using a fECG-recorder (MonicaHealthcare). Patients used the recorder at home (n=40) orin hospital (n=35). fECG data was obtained from fiveThe newly developed fECG-recorder appeared anaccurate tool for long-term antepartum FHR monitoring,especially during the night. In the future the fECG will beof particular interest for fetal monitoring in at riskpregnancies.NotesPage 125


Poster19 The Relationship of Microchimerism to AbortionTomoko Sato 1) , Keiya Fujimori 2) , Akira Sato 2) , Hitoshi Ohto 3) , KenichiHata 4) , Hiroko Sasaki 4) , Mihoko Yazawa 4) , Shizuka Honda 5) , MayumiNoguchi 5)1) Ohara General Hospital, Fukushima, <strong>Japan</strong> 2) Fukushima Medical University OB/GYN,Fukushima, <strong>Japan</strong> 3) Fukushima Medical University Transfusion/Immunology, Fukushima, <strong>Japan</strong>4) Meiji Hospital, Fukushima, <strong>Japan</strong> 5) Nishiguchi Clinic, Fukushima, <strong>Japan</strong>Objective: Feto-maternal transfusion occurs in mostpregnancies. Researchers have recently reported anassociation between fetal cell microchimerism <strong>and</strong>autoimmune diseases; the phenomenon was related tothe persistence of fetal DNA postpartum. The purpose ofthis study was to determine the perseverance of fetal cellmicrochimerism after artificial or spontaneous abortion inthe first trimester.Methods: A total of 84 women who had never had anabortion or a male delivery were registered. Peripheralblood samples were obtained from 67 women whounderwent a dilatation <strong>and</strong> curettage in the first trimester.Samples were collected from each woman at three points:before, 7 days after, <strong>and</strong> 30 days after the abortion. Ychromosome-specific, nested PCR targeting SRY wasused to test DNA extracted from lymphocytes in the buffycoat. DNA was also extracted from the chorion todetermine sex. The sensitivity of our assay was thedetection of approximately one male cell in 100,000female cells.Results: A total of 31 male (48%) <strong>and</strong> 36 female (52%)chorions were obtained. Male DNA was found in 54.8%of women who had a male chorion before the abortion,<strong>and</strong> decreased to 6.5% at 7 days after the abortion. At 30days after the abortion, the DNA had completelydisappeared. Male DNA was never detected at any pointfrom women who had a female chorion. No differencewas found between artificial <strong>and</strong> spontaneous abortion.Conclusion: <strong>Fetal</strong> cell microchimerism was not detectedafter any abortion. Male cellular DNA disappeared frommaternal circulation 30 days after the abortion.NotesPage 126


PosterThe effect of cerebral hypothermia on cortisol <strong>and</strong> ACTHresponses after umbilical cord occlusion in preterm fetal sheep.20Joanne Davidson, Mhoyra Fraser, Andrew S. Naylor, Vincent Roelfsema,Alistair J. Gunn, Laura Bennet.<strong>Fetal</strong> Physiology <strong>and</strong> Neuroscience Group, Dept of Physiology, The University of Auckl<strong>and</strong>,Auckl<strong>and</strong>, New Zeal<strong>and</strong>The hypothalamus-pituitary-adrenal (HPA) axis isessential for adaptation to stress. It is unknown whetherthese endocrine responses are impaired or exaggeratedby mild systemic hypothermia induced as part of headcooling. We therefore examined HPA responses inchronically instrumented preterm fetal sheep (104 d ofgestation, term is 147 d), allocated to sham occlusion(n=7), 25 min of complete umbilical cord occlusion (n=7)or occlusion <strong>and</strong> head cooling with mild systemichypothermia (n=7, mean±SEM esophageal temperature37.6±0.3°C vs. 39.0±0.2°C; P


Poster21The prevention of fetal brain <strong>and</strong> lung injuries based oninterleukin-6 levels in amniotic fluid before birthTakushi Hanita, Tadashi Matsuda, Masaki Sato, Shizuko Akiyama, RyutaKitanishi, Yuichiro Miura, Tatsuya Watanabe, Hidenobu OhtaDivision of Perinatal Medicine, Tohoku University Hospital, <strong>Japan</strong>.Objective:To determine whether interleukin-6 (IL-6) levels inamniotic fluid before birth could predict postnataldevelopment of periventricular leukomalacia <strong>and</strong> chroniclung disease in premature infants.Study design:A total of 16 pregnant women in cases of clinicalchorioamnionitis (CAM) were underwent transabdominalamniocentesis <strong>and</strong> divided into two groups on the basis ofIL-6 levels in amniotic fluid, the high IL-6 group (≥17 ng/ml,n=8) <strong>and</strong> the low IL-6 group (


Effect of intrauterine inflammation on induction of antenatalperiventricular leukomalacia in chronically instrumented fetalsheepMasatoshi SaitoDepartment of Obstetrics & Gynecology, Tohoku University Graduate School of Medicine,Sendai, <strong>Japan</strong>Poster22Objective:Our purpose was to determine effects of intrauterineinflammation on induction of antenatal periventricularleukomalacia (PVL) in premature fetal sheep.Methods:Fetuses were infused with 40 µg/day ofgranulocyte-stimulating factor intravenously at 105-109days gestation <strong>and</strong> 20 mg of lipopolysaccharide (LPS) intothe amniotic cavity at 107 days gestation. At 24 hoursafter the LPS infusion, the hemorrhage group received anacute withdrawal of 40% of the fetoplacental blood volume(n=5), whereas an isovolemic exchange transfusion wasperformed in the control group (n=5). Changes in the totalhemoglobin (Hb), oxy-Hb, <strong>and</strong> deoxy-Hb levels in thecerebral tissue were assessed using near-infraredspectroscopy throughout the study period <strong>and</strong> comparedstatistically (ANOVA). Five days after the insult, fetuseswere processed for histologic analysis.Results:Four of 5 fetuses in the hemorrhage group <strong>and</strong> four of 5fetuses in the control group exhibited PVL. In all fetuses inthe both groups, necrotizing membranitis <strong>and</strong> funisitiswere observed <strong>and</strong> both the brain total-Hb <strong>and</strong> deoxy-Hbwere continuously increased from 6 hours after the LPSinfusion. After the insult, the brain total-Hb, oxy-Hb <strong>and</strong>deoxy-Hb in the both groups decreased seriously (p


Poster23Maternal Low Protein Diet Aggravates <strong>Fetal</strong> White MatterDamage Caused by InfectionTakuya Ito *1 , Kaori Uchida *2 , Michiyo Nakamura *2 , Hiroshi Chisaka *2 ,Yoshitaka Kimura *1 <strong>and</strong> Kunihiro Okamura *2*1 Tohoku University Biomedical Engineering Research Organization*2 Department of Gynecology <strong>and</strong> Obstetrics, Tohoku UniversityGraduate School of MedicineMaternal infection causes release of cytokines such aschorioamnionitis. Such cytokines are known to damageoligodendrocytes or oligodendrocyte precursor cells(OL/OPC) <strong>and</strong> affect the white matter in fetal brain. Lossof white matter has been related to various mentaldisorders. However, white matter damage in fetus doesnot necessarily occur in all cases of maternal infection.Therefore, there may be a critical factor that determinesthe occurrence of white matter damage in fetal brainduring infection. In this study we have focused on therelationship between maternal nutritional <strong>and</strong> fetal braindamage. Our hypothesis about undernutrition is based onthe well-known results from epidemiological survey of theDutch Hunger Winter in 1944.We fed low protein diet (L) or normal diet (N) to femaleC57BL/6N mice <strong>and</strong> performed transvaginaladministration of LPS (0.1mg/ml 30µl) or PBS (30µl) fromthe 14 th day of pregnancy. In total, we investigated thefollowing four groups (L_LPS, L_PBS, N_LPS, N_PBS).We collected brain samples from E17 fetuses <strong>and</strong> P7neonates, <strong>and</strong> made cross-sections for immunohistochemistry.We performed immunostaining with olig2, PDGFR alpha,MBP, GFAP <strong>and</strong> nestin antibodies <strong>and</strong> examineddifferentiation to Glia. We also investigated activecaspase3 <strong>and</strong> performed TUNEL assay for measurementof apoptosis. Upon performing the above studies, wephotographed periventricular white matter area in thebrain, measured the number of positive cells <strong>and</strong> madestatistical analyses.We detected no difference in E17 fetus brain uponadministration of LPS with/ without undernutrition.However, in P7 neonatal mice GFAP (+) <strong>and</strong> nestin (+)cells / GFAP (+) cells increased upon administration ofLPS both with normal diet or undernutrition diet. Decreaseof MBP (+) cells was observed in L_LPS mice only.Transvaginal administration of LPS caused increase ofGFAP (+) <strong>and</strong> nestin (+) cells / GFAP (+) cells. Theseresults indicate that maternal infection may affect fetalbrain in an adverse way. Brain damage observed in onlyL_LPS mice suggests maternal under nutrition mayinduce hyperactive response for infection in fetal brain.NotesPage 130


PosterPrenatal diagnosis of bradycardia using fetal electrocardiogramvia maternal abdomen24Yoshitaka Kimura *1 , Michiyo Nakamura *2 , Kaori Uchida *2 , Takuya Ito *1 ,Hiroshi Chisaka *2 , <strong>and</strong> Kunihiro Okamura *2*1 Tohoku University Biomedical Engineering Research Organization*2 Department of Gynecology <strong>and</strong> Obstetrics, Tohoku UniversityGraduate School of Medicine<strong>Fetal</strong> bradycardia is often associated with severediseases such as fetal distress, sick sinus syndrome, <strong>and</strong>atrioventricular block. Generally, it is difficult to make adiagnosis before birth because we cannot perform directfetal electrocardiogram. Here we report that we havecarried out fetal electrocardiogram via maternal abdomento patients showing severe bradycardia, <strong>and</strong> successfullyperformed appropriate prenatal diagnoses of arrhythmia.All the patients subjected to this study were referred to ourhospital for severe bradycardia. We performed fetalelectrocardiogram via maternal abdomen. One patientwas diagnosed with sick sinus syndrome. After vaginaldelivery, the baby underwent pacemaker implantation.Another patient was diagnosed with premature atrialcontraction. After one week of vaginal delivery, thearrhythmia disappeared. The last patient was diagnosedwith atrioventricular block. The baby underwentpacemaker implantation after vaginal delivery.Prenatal diagnosis of bradycardia using fetalelectrocardiogram can help us to determine the mode <strong>and</strong>the timing of the delivery, leading to appropriate therapyafter birth.NotesPage 131


Poster25Endothelin-1 inhibits both L-type Ca 2+ current <strong>and</strong> ATPsensitiveK + current in neonatal rat ventricular myocytesYasuhiro Katsube, Nobuko Suzuki, Makoto Watanabe, Masanori Abe,Miharu Hajikano, Mitsuhiro Kamisago, Ryuji Fukazawa, Shunichi OgawaDepartment of Pediatrics, Nippon Medical SchoolBackground: Endothelin-1 (ET-1), a potent vasoactivepeptide, has a wide range of biological effects in variousorgans <strong>and</strong> tissues. In the heart, ET-1 is released byendothelial, vascular smooth muscle <strong>and</strong> myocardial cells,particularly in cardiovascular disorders such ascongestive heart failure <strong>and</strong> ischemic heart disease. Inthese pathophysiological conditions, intracellular ATPcontents decrease, followed by the simultaneous releaseof catecholamines <strong>and</strong> ET-1, which may contribute to theregulation of cardiac function. We previously presentedthat ET-1 inhibits voltage-gated <strong>and</strong> ATP-sensitive K +channel in resistance pulmonary artery from neonatalrabbit, suggesting that ET-1 may act as a mediator ofvasospasm <strong>and</strong> hypertension even in immature tissue.Objective: In this study, we investigated the effects ofET-1 on L-type Ca 2+ current (I Ca,L ) <strong>and</strong> ATP-sensitive K +current (I K,ATP ) in immature ventricular myocytes.Methods: Cells were enzymatically isolated from theventricles of neonatal rats (aged 3 to 10 days). Whole-cellwas measured as the peak inward current at a testpotential of +10 mV from a holding potential of -40 mV.I K,ATP was measured at a test potential of +60 mV from aholding potential of -70 mV. All experiments wereperformed at 37°C.Results: ET-1 (10-100 nM) decreased not only basal I Ca,L ,but also forskolin (a direct activator of adenylate cyclase)-stimulated <strong>and</strong> isoproterenol (β-adrenoceptor agonist)-stimulated I Ca,L . On the other h<strong>and</strong>, ET-1 (10 nM) almostcompletely inhibited the ATP-sensitive K + channel opener,pinacidil-induced outward current (i.e., I K,ATP ). Furthermore,ET-1 inhibited the shortening of cardiomyocyte actionpotential induced by pinacidil. These effects were reducedin the presence of BQ-123, an ET A -receptor antagonist,which suggests that the action of ET-1 is mediated by theET A receptor.Conclusions: In the neonatal heart, ET-1 inhibits not onlycontractility, but also cardioprotecive action by inhibitingI Ca,L <strong>and</strong> I K,ATP .voltage <strong>and</strong> current clamp techniques were applied. I Ca,LNotesPage 132


PosterPlacenta accreta/increta in unscarred uterus 26Akagi K, Shima T, Ishigaki N, Oota S, Hayasaka A, Fujita N, Asano K <strong>and</strong>Wada YNational Health Organization Sendai Medical Center, <strong>Japan</strong>When the obstetrician encounter placenta previa after aprior cesarean section, careful preoperative managementwill take place because of the significant possibility ofplacenta accreta/increta. However, it is difficult toanticipate placenta accreta/increta in patients without prioruterine surgery or cesarean delivery. When placentaaccreta/increta do occur in these patients, promptintervention will be needed on site.Methods: We retrospectively reviewed our hospitalrecords for patient with placenta accreta/increta caseswithout prior uterine surgery including cesarean delivery.Clinically diagnosed cases <strong>and</strong> pathologically confirmedcases were included.Results: There were 8 cases of placenta accreta/increta(0.06% of all deliveries) without prior cesarean birth oruterine surgery between July 1992 <strong>and</strong> June <strong>2007</strong>. 5cases (62.5%) were occurred after 2004. In 4 cases,postpartum bleeding was controlled <strong>and</strong> retained placentawas successfully treated by methotrexate. In two cases,massive bleeding after vaginal delivery was onlycontrolled by hysterectomy on the same day. In twocases, cesarean section was performed because ofposterior wall low lying placenta. In one of these twocases, placenta was firmly adherent to the uterus <strong>and</strong>hysterectomy was done without attempting to remove theplacenta. Although bleeding was controlled by packing theuterus on surgery in other case, hysterectomy was donebecause of massive bleeding on the next day. At leastone uterine curettage was done in 4 cases (50%) butmaternal age was over 35 only in 2 cases (25%).Conclusions: The most important risk factors for placentaaccreta/increta is prior uterine surgery <strong>and</strong> the mostcommon setting is placenta previa after a prior cesare<strong>and</strong>elivery. In our study, there were 0.06% incidence ofplacenta accrete/increta without previous uterine surgery.Therefore, the obstetrician has to be cautious of thepossibility of placenta accrete/increta even in the absenceof previous uterine surgery.NotesPage 133


Poster27 QT interval during pregnancy in Long QT syndromeYuri Tokito, Akiko Omoto, Naoshi Yamada, Naoko Iwanaga, KaoruYamanaka, Keiko Ueda, Masayo Nozawa, Reiko Neki, Wataru Shimizu,Tomoaki IkedaNational Cardiovascular Center, Suita Osaka, <strong>Japan</strong>ObjectivesThe hereditary long QT syndrome (LQTS) ischaracterized by prolonged ventricular repolarization,manifesting arrhythmia-related recurrent syncope,aborted cardiac arrest, <strong>and</strong> sudden death. Pregnancy isassociated with frequent cardiac events in the women withLQTS. In the animal study, QT interval was significantlyprolonged in pregnanct mice compared withnon-pregnancy mice. (Mansoureh Eghbali et al,Circulation Research 2005) We retrospectivery analizedelectro cardiogram(ECG) of pregnant women with LQTSin perinatal period.Methods17 pregnancies in 7 LQTS cases were treated from 1992to 2006 in National Cardio Vascular Center, <strong>Japan</strong>. 15pregnancies succesfuly resated in live infant, 10 of whichwas diagnosed as LQTS subsequently. 2 pregnancieswas terminated as intentional abortion. In 8 pregnanciesfrom 6 cases, ECG was available both in pre- <strong>and</strong>postpartum periods.We measured the QT interval corrected the heart rate(cQT) <strong>and</strong> were compared cQTs between pre- <strong>and</strong>postpartum periods.ResultThe averages of cQT interval in pre-partum periods were568±82msec, longer than that in post-partum periods:504±55msec, in 8 pregnancies. There were no differentsin cQT interval among three different trimester ofpregnancy. In the LQT type2 case, which were seriallymeasured cQT interval, significant decrease in cQTinterval was observer just after the delivery, remaining upto postpartum day 150.ConclusionsQT interval was prolonged during pregnancy in the LQTS,consistant with animal study of pregnant mice. Thisprolongation may be associated with frequent cardiacevents during pregnancy of LQTS.NotesPage 134


High altitude chronic hypoxia modifies cardiovascularresponses in newborn sheep depending on the time <strong>and</strong>duration of the hypoxic insult during gestation.Poster28a,d EA Herrera, a G Ebensperger, b RA Riquelme, a C Torres-Farfan, a VRReyes, c JT Parer, d DA Giussani, a AJ Llanos.a Facultad de Medicina, b Facultad Ciencias Químicas y Farmacéuticas, Universidad de Chile,Chile; c University of California San Francisco, USA; d University of Cambridge, UK.Chronic hypoxia is a common insult during fetaldevelopment inducing structural <strong>and</strong> functional changes inpulmonary <strong>and</strong> systemic circulations. Hypothesis:Maternal hypoxia induces neonatal cardiovascularchanges depending on the time <strong>and</strong> duration of thehypoxic insult during fetal life. Methods: Pregnant eweswere divided into three groups: conception, pregnancy<strong>and</strong> delivery in lowl<strong>and</strong> (500m, LLL); conception inlowl<strong>and</strong> <strong>and</strong> at 30% gestation taken to highl<strong>and</strong> hypoxia(3,600m) until delivery <strong>and</strong> then taken back to lowl<strong>and</strong>(LHL); <strong>and</strong> conception, pregnancy <strong>and</strong> delivery inhighl<strong>and</strong> hypoxia (3,600m, HHH). LLL <strong>and</strong> LHL werestudied at lowl<strong>and</strong> <strong>and</strong> HHH at highl<strong>and</strong>. Newborn lambswere catheterised under general anesthesia <strong>and</strong>submitted to acute hypoxic episodes. Results: LHL grouppresent a high abortion (24%) <strong>and</strong> stillbirth (11%) rate, notseen in the other groups. Under basal conditions, LHL <strong>and</strong>HHH had higher pulmonary arterial pressures (PAP,20.8+1.2 <strong>and</strong> 20.2+2.4 vs. 13.6+0.5 mmHg, p


NotesPage 136


List of delegatesYuka AbeDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>abe-9.27.-yuka@s3.dion.ne.jpSamana AliDepartment of Pediatrics.Aga Khan UniversityStadium Road, P.O. BOX 3500Karachi 74800, Pakistansamana.ali@aku.eduKozo AkagiNational Health Organization,Sendai Medical Center2-8-8 Miyagino Miyagino-kuSendai 983-8520<strong>Japan</strong>akagik@snh.go.jpMaiko AraiDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>maiko-k@mail.tains.tohoku.ac.jpKazunori BabaCenter for Maternal, <strong>Fetal</strong> <strong>and</strong><strong>Neonatal</strong> Medicine, SaitamaMedical Center,Saitama Medical University1981 Kamoda, Kawagoe,Saitama <strong>Japan</strong> 350-8550baba-tokyo@umin.netLaura BennetDepartment of Physiology,The University of Auckl<strong>and</strong>85 Park Road, Grafton,Auckl<strong>and</strong> 1023New Zeal<strong>and</strong>l.bennet@auckl<strong>and</strong>.ac.nzAlan D BockingUniversity of TrontCanadaabocking@mtsinai.on.caFong-Ming ChangOb/Gyn, Natl Cheng Kung Univ MedCollege & Hospital138 Victory Road 70428Taiwanfchang@mail.ncku.edu.twHiroshi ChisakaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>chisaka@mail.tains.tohoku.ac.jpJason H Collins MdPregnancy InstituteP.O. Box 806 New Roads LA 70760U.S.A.haydel1@bellsouth.netJoanne DavidsonDepartment of Physiology,The University of Auckl<strong>and</strong>85 Park Road, Grafton,Auckl<strong>and</strong> 1023New Zeal<strong>and</strong>joanne.davidson@auckl<strong>and</strong>.ac.nzRobert De MatteoMonash UniversityDepartment of Anatomy <strong>and</strong> CellBiologyWellington Road, Clayton 3168Australiarobert.dematteo@med.monash.edu.auJan DerksUniversity Medical Center UtrechtKerkstraat 6 Utrecht 3581rdNetherl<strong>and</strong>sjbderks@hotmail.comJ.B.Derks@umcutrecht.nlAlison ForheadUniversity of CambridgeDepartment of Physiology,Development <strong>and</strong> NeuroscienceDowning Street,Cambridge CB2 3EGU.K.ajf1005@cam.ac.ukMhoyra FraserLiggins Institute, University of Auckl<strong>and</strong>Liggins Institute, Faculty of Medical<strong>and</strong> Health Sciences Universityof Auckl<strong>and</strong>,Private Bag 92019, Auckl<strong>and</strong> 1New Zeal<strong>and</strong>m.fraser@auckl<strong>and</strong>.ac.nzKeiya FujimoriDept. Ob & Gyn. Fukushima MedicalUniversity1-Hikarigaoka Fukushima 960-1295<strong>Japan</strong>fujimori@fmu.ac.jpHideoki FukuokaWaseda University1-1-1, Nishiwaseda, Shinjuku-KuTokyo 169-0051<strong>Japan</strong>hideoki.fukuoka@gmail.comHelena M GardinerImperial College52 Valiant House, VicarageCrescent London SW11 3LUU.K.helena.gardiner@imperial.ac.ukSherly GeorgeDepartment of Physiology,The University of Auckl<strong>and</strong>85 Park Road, Grafton,Auckl<strong>and</strong> 1023New Zeal<strong>and</strong>sa.george@auckl<strong>and</strong>.ac.nzCaterina GuiotUniversity of TorinoDept Neuroscience C. Raffaello30 10125Italycaterina.guiot@unito.itAlistair J GunnDepartment of Physiology,The University of Auckl<strong>and</strong>85 Park Road, Grafton,Auckl<strong>and</strong> 1023New Zeal<strong>and</strong>aj.gunn@auckl<strong>and</strong>.ac.nzTakushi HanitaDivision of Perinatal Medicine,Tohoku University Hospital,<strong>Japan</strong>1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>punita@olive.ocn.ne.jpMark HansonUniversity of SouthamptonInstitute of Developmental Sciences,Mailpoint 887, SouthamptonGeneral Hospital, Tremona Road,Southampton, Hants SO16 6YDU.K.m.hanson@soton.ac.ukRichard HardingDept of Anatomy <strong>and</strong> Cell BiologyMonash University 3800Australiarichard.harding@med.monash.edu.auPage 138


Steve HarveyUniversity of AlbertaDepartment of Physiology Universityof Alberta T6G2H7Canadasteve.harvey@ualberta.caShinichi HayasakaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>siniti-hayasaka@pref.iwate.jpChika HayashiDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>chayashi@mail.tains.tohoku.ac.jpEri HirokiDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>erimer@zg7.so-net.ne.jpYoshiyasu Hombo1-3-23, Izumino-Demachi,Kanazawashi, Ishikawaken921-8116<strong>Japan</strong>yoshiyasu@spacelan.ne.jpJeng-Hsiu HungDepartment of Obstetrics <strong>and</strong>Gynecology,Taipei Veterans General Hospital,201, Section 2, Shih-Pai Rd,Taipei 112,Taiwanjhhung@vghtpe.gov.twTomoaki IkedaNational Cardiovascular Center5-7-1 Fujishiro-Dai 565-8565<strong>Japan</strong>tikeda44@hotmail.comtikeda@hsp.ncvc.go.jpTsuyomu IkenoueUniversity of Miyazaki5200, Kihara, Kiyotake,Miyazaki 889-1692<strong>Japan</strong>satomako@fc.miyazaki-u.ac.jpTakuya ItoTohoku University BiomedicalEngineering ResearchOrganizationTetsuya ItoNagoya City University,Department of PediatricsMizuho-Ku, Mizuho-Cho,Kawasumi 1 Nagoya 467-8601<strong>Japan</strong>itotetsu@med.nagoya-cu.ac.jpKiyoshi ItoDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>kito@mail.tains.tohoku.ac.jpHiroaki ItohOsaka National Hospital2-1-14 Houenzaka, Chuou-Ku,Osaka, 540-0006<strong>Japan</strong>hitou-endo@umin.ac.jpNoriyuki IwamaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>noribou@mail.tains.tohoku.ac.jpSharif JafarDepartment of Obstetrics <strong>and</strong>Gynecology, Tohoku UniversitySchool of Medicine1-1 Seiryo-machi, Aoba-ku,Sendai-shi, Miyagi 980-8574, <strong>Japan</strong>jafarsharif@tubero.tohoku..ac.jpGraham JenkinMiscl, Bldg 75, Strip 1, 3rd Floor,Monash UniversityClayton Victoria 3800Australiagraham.jenkin@med.monash.edu.auNaru KageyamaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>narukageyama@kcd.biglobe.ne.jpYukika KakutaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>snowsmell@mail.tains.tohoku.ac.jpIneko KatoDepartment of Pediatrics,Nagoya City University1 Kawasumi, Mizuho,Nagoya 467-8601<strong>Japan</strong>ikato@med.nagoya-cu.ac.jpYasuhiro KatsubeNippon Medical SchoolMusashikosugi Hopital1-396 Kosugi-Cho Nakahara-KuKawasaki 211-8533<strong>Japan</strong>katsube@nms.ac.jpIchiro KawabataDirector of the Department of <strong>Fetal</strong><strong>and</strong> Maternal MedicineNational Nagara Medical Center1300-7Nagara Gifu city,Gifu,<strong>Japan</strong>502-8558kawabata@nagara-lan.hosp.go.jpYasuyuki KawagoeDep. of Ob & Gyn, University ofMiyazaki5200 Kihara, Kiyotake-Cho,Miyzaki 880-1692<strong>Japan</strong>yasuk@fc.miyazaki-u.ac.jpMie KawaiDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>Page 139


Kumiko KawanoDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>k-kawano@mail.tains.tohoku.ac.jpKei KawashimaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>Yoshitaka KimuraDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>ykimura@ob-gy.med.tohoku.ac.jpEllen KnappDepartment of Physiology,The University of Auckl<strong>and</strong>85 Park Road, Grafton,Auckl<strong>and</strong> 1023New Zeal<strong>and</strong>e.knapp@auckl<strong>and</strong>.ac.nzAnna Hauntoft KongstedUniversity of CopenhagenGr Negaardsvej 7Frederiksberg 1870Denmarkahtp@life.dkBrian John KoosUcla22-139 Chs 10833 Le Conte Avenue,Los Angeles 90095-1740U.S.A.bkoos@mednet.ucla.eduShiro KozumaUniversity of Tokyo7-3-1 Hongo Bunkyo-Ku Tokyo113-8655<strong>Japan</strong>kozuma-tky@umin.ac.jpTomoyuki KuwataJichi Medical University3311-1, Yakushiji, Shimotsuke,Tochigi 329-0433<strong>Japan</strong>kuwata@jichi.ac.jpIrina G. MakarenkoInstitute of Developmental BiologyRas Vavilov St. 26 Moscow 119334Russiaimakarenk@mail.ruIkuko MakinoTokyo Women’s Medical University2-1-10, Nishiogu,Arakawa-Ku 116-8567<strong>Japan</strong>makinoog@dnh.twmu.ac.jpTadashi MatsudaCenter for Perinatal Medicine,Tohoku University Hospital1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>choku@mail.tains.tohoku.ac.jpYoshio MatsudaTokyo Women’s Medical University2-1-10, Nishiogu,Arakawa-Ku 116-8567<strong>Japan</strong>ym0709@obgy.twmu.ac.jpNaomi MatsudaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>naomim@mail.tains.tohoku.ac.jpSuzanne Lee MillerMonash UniversityDepartment of Physiology,Building 13f Monash University 3800Australiasuzie.miller@med.monash.edu.auTakahiro MinatoDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>t-minato@mail.tains.tohoku.ac.jpMariko MinouraDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>mari-m@mail.tains.tohoku.ac.jpJunko MinouraDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>jminoura@zb.cyberhome.ne.jpKohzoh MitsuyaBiofunctional Science, TohokuUniversity BiomedicalEngineering ResearchOrganization (TUBERO)2-1 Seiryomachi, Aobaku,Sendai 980-8575, <strong>Japan</strong>kmitsuya@tubero.tohoku.ac.jpYuichiro MiuraPerinatal Medicine, TohokuUniversity Hospital1-1, Seiryo-cho, Aoba-ku,Sendai, Miyagi, 980-8574, <strong>Japan</strong>y-miu@mail.tains.tohoku.ac.jpLeon MuldersMaxima Medical Center VeldhovenMarie Curielaan 3Eindhoven 5644 DrNetherl<strong>and</strong>slgm.mulders@chello.nlTakashi MurakamiDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>murakami@mail.tains.tohoku.ac.jpYuji MurataAizenbashi Hospital5-16-15 Nipponbashi Naniwa-kuOsaka-shi Osaka 556-0005<strong>Japan</strong>Jun MurotsukiDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>murotsuki@mail.tains.tohoku.ac.jpPage 140


Tomoyuki NagaiDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>Satoru NagaseDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>nagases@mail.tains.tohoku.ac.jpMichiyo NakamuraDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>minakamura@mail.tains.tohoku.ac.jpMichiya NatoriNational Center For Child Health <strong>and</strong>Development Ohkura2-10-1 Setagaya, Tokyo 157-8535<strong>Japan</strong>natori-m@ncchd.go.jpMarina V NechaevaInstitute of Developmental BiologyRas Vavilov St. 26 Moscow 119-334Russiamnechaeva2003@yahoo.comReiko NekiNational Cardiovascular Center5-7-1, Fujishiro-Dai SuitaOsaka 565-8565<strong>Japan</strong>rneki@hsp.ncvc.go.jpHitoshi NiikuraDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>niikura@mail.tains.tohoku.ac.jpHidekazu NishigoriDept. of Ob & GynIwate Medical University19-1 Uchimaru MoriokaIwate 020-8505<strong>Japan</strong>hknishig@iwate-med.ac.jpYasuhisa NomuraFukushima Nartional Hospital13 AshidadukaSukagawa City 962-8507<strong>Japan</strong>nomuyasu1969@yahoo.co.jpTakashi OkaiDept. of Obst <strong>and</strong> Gyne,Showa University1-5-8 Hatanodai Shinagawa-KuTokyo 142-8666<strong>Japan</strong>okai.t@med.showa-u.ac.jpKunihiro OkamuraDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>okamura@mail.tains.tohoku.ac.jpTeruo OkanoInstitute of Advanced BiomedicalEngineering <strong>and</strong> ScienceTokyo Women’s Medical University8-1kawada-cho, Shinjuku-ku,Tokyo,162-8666 <strong>Japan</strong>Hitoshi OshitaniDepartment of VirologyTohoku University, Graduate Schoolof Medicine1-1Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>Hidenobu OtaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>hideohta@mail.tains.tohoku.ac.jpJulian T. ParerUniversity of California SanFrancisco505 Parnassus AvenueSan Francisco CA 94143-0132U.S.A.pareb@obgyn.ucsf.eduUsha RajLos Angeles Biomedical ResearchInstitute1124 West Carson St.,Rb-1 Torrance, CA 90502U.S.A.raj@labiomed.orgKusol Russameecharoen4-25-5, Mitsukoshi Higashi NakanoMansion, Room No. 602,Nakano-Ku, Tokyo 164-0003<strong>Japan</strong>rkusol@yahoo.comBurcu SaglamMonash University Dept ofObstetrics <strong>and</strong> Gynaecology,Level 5 Monash Medical Centre246 Clayton Road, Clayton,Victoria 3168AustraliaBurcu.Saglam@med.monash.edu.auMasatoshi SaitoDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>masa-saito@mail.tains.tohoku.ac.jpTsukuru SaitoSuzuki Memorial Hospital3-5-5 Satonomori Iwanuma-shiMiyagi 989-2481<strong>Japan</strong>t.saito@suzukihp.or.jpHiroshi SameshimaUniversity of Miyazaki5200 Kihara, KiyotakeMiyazaki 889-1692<strong>Japan</strong>hsameshima@fc.miyazaki-u.ac.jpAkira SatoDept. Ob & Gyn.Fukushima Medical University1-Hikarigaoka Fukushima 960-1295<strong>Japan</strong>akira-s@fmu.ac.jpPage 141


Tomoko SatoOhara General Hospital, Fukushima6-11 Omachi Fukushima-City,Fukushima 960-8611<strong>Japan</strong>tomoko-s@ohara-hp.or.jpKazuyo SatoDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>kazusato@mail.tains.tohoku.ac.jpNaoaki SatoDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>ny-satou@cb3.so-net.ne.jpTakenobu ShimizuDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>Norio Shinozuka4-12 Mitsuka Hiratsuka,Kanagawa 254-0045<strong>Japan</strong>norio@shinozuka.comJunichi SugawaraKyono Art Clinic1-1 Honcyo Aoba-ku Sendai1-2 Miyagi 980-0014<strong>Japan</strong>sugawara@ivf-kyono.or.jpVeena Gayathri SupramaniamMonash UniversityMonash Immunology & Stem CellCentre Strip 1,Monash University 3800Australiaveena.supramaniam@med.monash.edu.auKazunao SuzukiHamamatsu University School ofMedicine H<strong>and</strong>ayama1-20-1 Higashiku Hamamatsu City<strong>Japan</strong>ks101865@f8.dion.ne.jpKeiji SuzukiSaitama Medical Center1981 Tsujido-Machi, Kamoda,Kawagoe, Saitama 350-8550<strong>Japan</strong>dks@saitama-med.ac.jpSatoshi SuzukiDepartment of Pediatrics, NagoyaCity University1 Kawasumi, Mizuho,Nagoya 467-8601<strong>Japan</strong>s.suzu@med.nagoya-cu.ac.jpKichiya SuzukiDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>kichiyasuzuki@mail.tains.tohoku.ac.jpHidenori TakahashiDepartment of Obstetrics <strong>and</strong>Gynecology Fukushima MedicalUniversity School of Medicine,Fukushima1 Hikarigaoka Fukushima city, <strong>Japan</strong>hide-t@fmu.ac.jpToshifumi TakabayashiDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>Yuha TakadaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>takadayh@mail.tains.tohoku.ac.jpNaomi TakahashiDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>Tadao TakanoDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>ttakano@mail.tains.tohoku.ac.jpKojiro TanabeDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>mar22tana@aol.comSouta TanakaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>tanakasouta@hotmail.co.jpShingo Tanigawara<strong>Japan</strong>ese Red Cross SendaiHospitalDepartment of Obstetrics <strong>and</strong>Gynecology2-43-3 Yagiyamahoncyo Taihaku-kuSendai Miyagi 982-8501<strong>Japan</strong>tanigawara@miyagi.med.or.jpYukihiro TeradaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>terada@mail.tains.tohoku.ac.jpHajime TogariDepartment of Pediatrics,Nagoya City University1 Kawasumi, Mizuho,Nagoya 467-8601<strong>Japan</strong>togari@med.nagoya-cu.ac.jpYuri TokiouNational Cardio Vasuculer CenterFujishirodai 5-7-1 SuitaOsaka 565-8565<strong>Japan</strong>yuri_tokitou@hotmail.comPage 142


Claudia Torres FarfanUniversidad De ChileSalvador 486, Providencia, SantiagoChilectorresf@med.uchile.clMasafumi ToyoshimaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>m-toyo@mail.tains.tohoku.ac.jpShinya TsuchidaThe University of Tokyo7-3-1 Hongo Bunkyo-KuTokyo 113-8655<strong>Japan</strong>shinyatsuchida556@hotmail.comKaori UchidaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>kao-u@mwe.biglobe.ne.jpTomohisa UgajinDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>tugajin@mail.tains.tohoku.ac.jpNobuya UnnoKitasato University, School ofMedicine1-15-1, Kitasato, Sagamihara City,Kanagawa 228-8555<strong>Japan</strong>unno@med.kitasato-u.ac.jpHiroki UtsunomiyaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>uskichi@mail.tains.tohoku.ac.jpGuido WassinkDepartment of PhysiologyThe University of Auckl<strong>and</strong>,Auckl<strong>and</strong> 1003New Zeal<strong>and</strong>g.wassink@auckl<strong>and</strong>.ac.nzFlora Y WongRitchie Centre For Bhr,Monash UniversityLevel 5, 246 Clayton RoadClayton 3168Australiaflora.wong@med.monash.edu.auTze-Fang WongDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>wongtzefang@mail.tains.tohoku.ac.jpIan M WrightMother And Babies Research CentreHunter Medical Research Institute,Lookout Road, New Lambton Hts,Newcastle 2287AustraliaIan.Wright@hnehealth.nsw.gov.auShanhai XuDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>shanhaix@mail.tains.tohoku.ac.jpHuixia YangDepartment of Obstetrics <strong>and</strong>Gynecology, Peking UniversityFirst Hospital,Beijing, 100034 Chinayanghuixia688@sina.comNobuo YaegashiDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>yaegashi@mail.tains.tohoku.ac.jpShun YasudaTakeda General Hospital<strong>Japan</strong>shunyasuda335@yahoo.co.jpKosuke YoshinagaDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>y-kou-m@mtb.biglobe.ne.jpHiromitsu YukiDepartment of Obstetrics &Gynecology,Tohoku University Graduate Schoolof Medicine1-1 Seiryo-Machi Aoba-KuSendai 980-8574<strong>Japan</strong>hiromitsu@mail.tains.tohoku.ac.jpTakanori WatanabeSendai City Hospital3-1 Shimizukouji Wakabayashi-kuSendai-shi Miyagi 984-8501<strong>Japan</strong>Page 143


Page 144


Author IndexAkagi, Kozo··························· P26Ali, Samana··························· Abstract13,Abstract14Bennet, Laura ······················· Plenary lecture 5,Abstract35, P11Bocking, Alan ························ Abstract38Chang, Fong–Ming ··············· P12,P13Collins, HJason ····················· Abstract17Davidson, Joanne ················· Abstract24,P20Derks, B Jan ························· Abstract34,Abstract46,P18Forhead, J Alison ·················· Abstract40Fraser, Mhoyra······················ Abstract5Fukuoka, Hideoki ·················· Abstract36Gardiner, Helena··················· Abstract18,Abstract20George, Sherly······················ Abstract31Guiot, Caterina······················ P3,P4Gunn, J Alistair······················ Abstract10,Abstract25Hanita, Takushi ····················· P21Hanson, Mark························ Plenary lecture 2Harding, Richard ··················· Abstract1,Abstract41Harvey, Steve ······················· Abstract23Hombo, Yoshiyasu················ Abstract16Hung, Jeng-Hsiu ··················· Abstract15Ishino, Fumitoshi··················· Invited lectureIto, Takuya ···························· P23Itoh, Hiroaki··························· Abstract3Jafar, Sharif··························· P2Jensen, Ellen ························ Abstract4,P1Katsube, Yasuhiro················· P25Kawagoe, Yasuyuki··············· P14Kimura, Yoshitaka················· Abstract45,P24Kongsted, Hauntoft Anna ······ Abstract2Levine, Richard ····················· Plenary lecture 3Makarenko, G. Irina·············· Abstract29Makino, Ikuko ······················· P17Matsuda, Naomi ··················· P10Matteo, De Robert················ Abstract26Miller, L. Suzanne ················ Abstract28,Abstract30Mitsuya, Kohzoh··················· Abstract37Miura, Yuichiro ····················· P16Murotsuki, Jun······················ Abstract8Nakamura, Michiyo ·············· Abstract9Nechaeva, V. M.··················· Abstract33Neki, Reiko··························· Abstract21Ohta, Hidenobu ···················· Abstract7Okano, Teruo ······················· Invited lectureOshitani, Hitoshi ··················· Geoffrey Dawes LectureParer, T Julian······················ Plenary lecture 1Raj, Usha J··························· Abstract42Saglam, Burcu······················ Abstract27,Abstract47Saito, Masatoshi··················· P22Sato, Kazuyo························ Abstract39Sato, Tomoko······················· P19Shinozuka, Norio·················· Abstract19,Abstract43Supramaniam, Veena ·········· Abstract44Suzuki, Keiji·························· Abstract12Suzuki, Kazunao ·················· Abstract22Takahashi, Hidenori ············· P5,P6,P7,P8,P9Tokitou, Yuri ························· P27Torres-Farfan, C··················· Abstract6,P28Wassink, Guido ···················· Abstract32Wong, Flora·························· Abstract11Wright, Ian···························· P15Yang, Huixia························· Plenary lecture 4Page 146

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

Saved successfully!

Ooh no, something went wrong!