• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 97
  • 36
  • 21
  • 9
  • 8
  • 7
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • 3
  • 2
  • Tagged with
  • 197
  • 91
  • 68
  • 36
  • 24
  • 23
  • 21
  • 21
  • 20
  • 19
  • 19
  • 17
  • 16
  • 16
  • 16
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
91

Transcriptional regulation of the mouse gonadotropin-releasing hormone receptor gene in pituitary gonadotrope cell lines

Sadie, Hanél 03 1900 (has links)
Thesis (PhD (Biochemistry))--University of Stellenbosch, 2006. / Gonadotropin-releasing hormone (GnRH), acting via its cognate receptor (GnRHR) is the primary regulator of mammalian reproductive function. Pituitary sensitivity to GnRH can be directly correlated with GnRHR levels on the surface of the pituitary gonadotrope cells, which can be regulated at transcriptional, post-transcriptional and post-translational levels. This study investigated mechanisms of transcriptional regulation of mouse GnRHR expression in two mouse gonadotrope cell lines, αT3-1 and LβT2, using a combination of endogenous mRNA expression studies, promoter-reporter studies, a two-hybrid protein-protein interaction assay, Western blotting, and in vitro protein-DNA binding studies. In the first part of the study, the role of two GnRHR promoter nuclear receptor binding sites (NRSs) and their cognate transcription factors in basal and Protein Kinase A (PKA)-stimulated regulation of GnRHR promoter activity was investigated in αT3-1 cells. The distal NRS was found to be crucial for basal promoter activity in these cells. While the NRSs were not required for the PKA response in these cells, results indicate a modulatory role for the transcription factors Steroidogenic Factor-1 (SF-1) and Nur77 via these promoter elements. The second part of the study focused on elucidating the mechanism of homologous regulation of GnRHR transcription in LβT2 cells, with a view to defining the respective roles of PKA and Protein Kinase C (PKC) in the transcriptional response to GnRH. In addition, the respective roles of the NRSs, the cyclic AMP response element (CRE) and the Activator Protein-1 (AP-1) promoter cis elements, together with their cognate transcription factors, in basal and GnRH-stimulated GnRHR promoter activity, were investigated. Homologous upregulation of transcription of the endogenous gene was confirmed, and was quantified by means of real-time RTPCR. The GnRH response of the endogenous gene and of the transfected promoter-reporter construct required PKA and PKC activity, and the GnRH response of the promoter-reporter construct was found to be dependent on a functional AP-1 site. Furthermore, GnRH treatment resulted in increased binding of phosphorylated cAMP-response element binding protein (phospho-CREB) and decreased expression and binding of SF-1 to their cognate cis elements in vitro, and stimulated a direct interaction between SF-1 and CREB, suggesting that these events are also required for the full transcriptional response to GnRH. This study is the first providing detail regarding the mechanism of transcriptional regulation of GnRHR expression in LβT2 cells by GnRH. Based on results from this study, a model has been proposed which outlines for the first time the kinase pathways, the promoter cis elements and the cognate transcription factors involved in homologous regulation of GnRHR transcription in the LβT2 cell line. As certain aspects of this model have been confirmed for the endogenous GnRHR gene, the model is likely to be physiologically relevant, and provides new ideas and hypotheses to be tested in future studies.
92

Dissection of GnRH receptor-G protein coupling

White, Colin D. January 2009 (has links)
Hypothalamic gonadotropin-releasing hormone (GnRH) (GnRH I) is the central regulator of the mammalian reproductive system. Most vertebrates studied also possess a second form of GnRH, GnRH II. GnRH I acts on its cognate G proteincoupled receptor (GPCR) on pituitary gonadotropes and activates Gq/11-mediated signalling pathways to stimulate the biosynthesis and the release of luteinising hormone (LH) and follicle-stimulating hormone (FSH). Both GnRHs have also been suggested to inhibit cellular proliferation, an action which has largely been proposed to be mediated by the coupling of the receptor to Gi/o. However, the range of G proteins activated by the GnRH receptor and the signalling cascades involved in inducing antiproliferation remain controversial. To delineate the G protein coupling selectivity of the mammalian GnRH receptor and to identify the signalling pathways involved in GnRH I-mediated cell growth inhibition, I examined the ability of the receptor to interact with Gq/11, Gi/o and Gs in Gαq/11 knockout MEF cells. My results indicate that the receptor is unable to interact with Gi/o but can signal through Gq/11. Additionally, my data do not support the suggestion of GnRH receptor-Gs interaction. Furthermore, I show that the GnRH Iinduced inhibition of cell growth is dependent on Gq/11, src and extracellular signal regulated kinase (ERK) but is independent of the activity of protein kinase C (PKC), Ca2+, jun-N-terminal kinase (JNK) or P38. Based on these findings and previous research within our group, I propose a mechanism whereby GnRH I may induce antiproliferation. Previous studies from our laboratory suggest that the GnRH receptor can adopt distinct active conformations in response to the binding of GnRH I and GnRH II. These data thus account for our hypothesis of ligand-induced selective signalling (LiSS). Given my previous results, I examined the ability of the GnRH receptor to couple to G12/13. My work indicates that the receptor can directly activate G12/13 and the downstream signalling cascades associated with this G protein family. Indeed, I provide evidence, in several cellular backgrounds, to suggest that GnRH receptor- G12/13-mediated signalling is involved in the regulation of GnRH-induced MAPK activity, SRE-driven gene transcription and cytoskeletal reorganisation. Furthermore, I propose a role for these G proteins in the transcriptional regulation of LHβ and FSHβ. Finally, I confirm previous results from our laboratory indicating that the GnRH receptor may interact with src Tyr kinase and show that GnRH I but not GnRH II may, independently of Gq/11, stimulate the Tyr phosphorylation and thus the activation of this protein. I propose that this differential signalling accounts for the distinct effects of GnRH I and GnRH II on cellular morphology and SREpromoted transcriptional activity. The research presented within this thesis provides evidence to refute published conclusions based on largely circumstantial experimental data, describes novel GnRH receptor signalling pathways and offers support for the concept of LiSS. It may assist in the development of new therapeutic compounds which selectively target one GnRH-mediated signalling pathway while bypassing others.
93

EFFECTS OF ELEVATING BLOOD LIPIDS IN ENERGY DEFICIENT ANESTROUS DAIRY HEIFERS ON PITUITARY RESPONSE TO GONADOTROPIN RELEASING HORMONE CHALLENGE.

Johnson, Mark Steven, 1955- January 1985 (has links)
No description available.
94

Influência de diferentes isoformas de fosfodiesterases no controle da maturação de oócitos bovinos / Influence of different phosphodiesterase isoforms on the control of bovine oocyte maturation

Zaffalon, Fabiane Gilli 31 July 2014 (has links)
A maturação in vitro do oócito é um dos fatores limitantes na produção in vitro de embriões. In vivo, esta maturação é um processo altamente orquestrado no qual a meiose é retomada pela onda de gonadotrofina que antecede a ovulação e que induz à queda dos níveis de AMPc no oócito. No entanto, os oócitos aspirados ao serem retirados dos folículos ovarianos retomam espontaneamente a maturação comprometendo a competência de seu desenvolvimento. O AMPc é sintetizado pela adenilato ciclase (AC) e degradado pelas fosfodiesterases (PDE), existindo algumas relacionadas à degradação do AMPc e outras do GMPc. Sendo assim, a proposição deste trabalho foi averiguar a contribuição de diferentes isoformas de fosfodiesterases na retomada da meiose e nos níveis de GMPc, AMPc e ainda, determinar quando há manutenção de AMPc em níveis elevados observando sua influência na competência oocitária e ativação da MAPK. Para isso, os complexos cumulus-oócito (CCOs) foram maturados in vitro na ausência, presença ou associação de inibidores de PDEs-AMPc e GMPc específicas e FSHr. As amostras foram avaliadas em relação a: 1) taxa de maturação; 2) níveis intracelulares de AMPc e GMPc nos CCOs; 3) taxa de desenvolvimento de blastocistos ; 4) ativação da MAPK em oócitos e células do cumulus. Os resultados obtidos no primeiro experimento indiaram que o inibidor da PDE3 foi o mais eficaz (p<0,05) em atrasar a retomada da meiose, às nove horas de maturação, porém, isolado ou em associação com o inibidor da PDE8, não foi capaz de alterar (p>0,05) os níveis de AMPc. No experimento dois, o inibidor da PDE5 isolado não influenciou a retomada da meiose (p>0,05), porém, quando associado aos inibidores da PDE3 e 8 houve atraso na retomada (p<0,05) e ainda alteraram os níveis de GMPc e AMPc (p<0,05) nas primeiras horas de maturação. O experimento três mostrou a influencia do FSHr durante a MIV, o qual estimulou a retomada da meiose, mas em associação com inibidores da PDE5 e 8 atrasa a retomada (p<0,05). Além disso, o FSHr provoca aumento do nível de AMPc e sua associação com inibidores de PDE5 e PDE8 ocasionou elevação adicional (p<0,05). As condições de cultivo estudadas no experimento quatro mostraram que a maturação induzida (pré-MIV de duas horas com agentes para elevar AMPc seguindo de 22 horas de MIV com FSH associado a inibidores de PDEs) atrasaram a retomada da meiose às nove horas de maturação, mas não afetaram progressão da meiose às 24, 28 e 30 horas. Os tratamentos, porém, não melhoraram a competência oocitária após a fertilização in vitro e ocasionaram pequenas variações na ativação da MAPK em oócitos e células do cumulus. / In vitro maturation of oocytes is a limiting factor in the in vitro production of bovine embryos. In vivo, this maturation is a highly orchestrated process in which meiosis resumption by the gonadotropin surge that precedes ovulation induces the decrease in cAMP levels in the oocyte. However, when oocytes are removed from follicles, they spontaneously resume maturation compromising the competence for its development. cAMP is synthesized by adenylyl cyclase (AC) and degraded by phosphodiesterases (PDE), and there are some PDEs related to degradation of cAMP and/or cGMP. Thus, the purpose of this work was to investigate the contribution of different isoforms of phosphodiesterases in the resumption of meiosis and levels of cAMP and, also, to determine differences in signaling pathways when maintaining high levels of cAMP and its influence on oocyte competence. For this purpose, cumulus-oocyte complexes (COC) were matured in vitro in the presence, absence or combination of inhibitors of cAMP- and cGMP-specific PDEs and FSH. Samples be were evaluated in relation to: 1) maturation rate, 2) intracellular levels of cAMP and cGMP in COCs, 3) rate of blastocyst development and 4) activation of MAPK in oocytes and cumulus cells. The results of the first experiment showed that the PDE3 inhibitor is more effective (p <0.05) in delaying meiosis resumption, at nine hours of maturation, but was not capable of altering cAMP levels (p> 0.05) either alone or in combination with the PDE8 inhibitor. In experiment two, the PDE5 inhibitor alone did not affect the meiosis resumption (p> 0.05), however, when associated with PDE3 and PDE8 inhibitors it delayed their resumption (p <0.05) and also altered cGMP and cAMP levels of (p <0.05) in the early hours of maturation. The third experiments showed the influence of FSHr during IVM, which stimulated the resumption of meiosis, but in combination with PDE5 and PDE8 inhibitors meiosis was delayed (p <0.05). Furthermore, FSHr causes increased levels of cAMP and its association with PDE5 and PDE8 inhibitors caused an additional increase (p <0.05). Culture conditions studied in experiment four showed that induced maturation (pre-IVM for two hours with agents to elevate cAMP followed by 22 hours IVM with FSH associated with PDE inhibitors) delayed the resumption of meiotic maturation at nine hours, but has no effect on meiosis progression at 24, 28 and 30 hours. The treatments, however, did not improve oocyte competence after in vitro fertilization and caused minor variations in the activation of MAPK in oocytes and cumulus cells.
95

A influência do biglicam mediada por receptores do tipo Toll-like 2 e 4 no processo de invasão das células trofoblásticas. / The influence of biglycan mediated by Toll-like receptors 2 and 4 in the invasion of trophoblast cells.

Borbely, Alexandre Urban 25 October 2013 (has links)
O biglicam é um proteoglicano é altamente expresso em células trofoblásticas de patologias placentárias com invasividade exacerbada. No entanto, as funções do biglicam no trofoblasto ainda não foram elucidadas. Sendo assim, verificamos a expressão e as funções de biglicam e seus receptores Toll-like (TLR)-2 e TLR-4 nas células trofoblásticas durante a gestação. As células do citotrofoblasto extraviloso (CTEV) foram positivas para todas as moléculas, menos para o biglicam em placentas a termo. Adição exógena de biglicam promoveu migração e invasão das células trofoblásticas. O biglicam estimulou a fosforilação de AKT nos sítios Thr308 e Ser473 nas células trofoblásticas. A migração e a invasão biglicam-dependentes e as fosforilações de AKT foram inibidas após a adição de anticorpos bloqueadores anti-TLR-2 e anti-TLR-4. O silenciamento gênico de AKT1 em células SGHPL-5 aboliu os efeitos do biglicam na motilidade. Em conclusão, o biglicam aumenta a motilidade de células trofoblásticas após sinalização por AKT através da ativação de TLR-2 e TLR-4. / Biglycan is a highly expressed proteoglycan in trophoblast cells from invasiveness-changed placental pathologies. However, biglycan functions in the trophoblast were not yet identified. Therefore, it was verified the expression and functions of biglycan and its receptors Toll-like (TLR)-2 and TLR-4 in trophoblast cells throughout pregnancy. The extravillous cytotrophoblast cells (EVT) were positive to all the molecules, although biglycan was negative in term placentas. Exogenous biglycan promoted migration and invasion of trophoblast cells. Biglycan stimulated AKT phosphorilation at Thr308 and Ser473 sites in trophoblast cells. The biglycan-dependent migration, invasion and AKT phosphorilation were inhibited upon addiction of anti-TLR-2 and anti-TLR-4 blocking antibodies. AKT1 genic silencing in SGHPL-5 cells abolished the motility effects. In conclusion, biglycan increases the motility of trophoblast cells after AKT signaliing throughout TLR-2 and TLR-4 activation.
96

Clonagem, caracterização e análise filogenética das subunidades alfa e beta do hormônio folículo estimulante de Pirarucu (Arapaima gigas) visando sua síntese em células CHO / Cloning, characterization and phylogenetic analisys of the alpha and beta subunits of the follicle stimulating hormone of Pirarucu (Arapaima gigas) in view of its synthesis in cho cells

Carvalho, Roberto Feitosa de 16 June 2014 (has links)
O Pirarucu (Arapaima gigas) é um peixe gigante da família Arapaimidae, nativo das bacias amazônicas que pode chegar a dois metros de comprimento e pesar mais de 200 Kg. Está presente no Equador, na Colômbia, no Peru, na Bolívia e no Brasil. Atualmente a espécie está ameaçada de extinção devido à pesca predatória e ao aumento da presença humana em seus viveiros naturais. No presente trabalho, os cDNAs da subunidade (ag-GTHα) e da subunidade β do hormônio folículo estimulante de A. gigas (ag-FSH) foram isolados e clonados pela primeira vez, possibilitando uma melhor compreensão da diversidade e evolução desta glicoproteína em peixes e a futura síntese biotecnológica deste hormônio para fins reprodutivos e alimentícios. Tanto o cDNA do ag-GTHα quanto aquele do ag-FSHβ foram sintetizados pela reação de transcriptase reversa (RT) e pela reação em cadeia da polimerase (PCR) utilizando como molde RNA total proveniente das glândulas hipofisárias de A. gigas. O cDNA da subunidade ag-GTHα possui uma sequência codificadora (Open Reading Frame, ORF) de 348 pb, o que corresponde a uma proteína de 115 aminoácidos com um suposto peptídeo sinal de 24 aminoácidos e com um peptídeo maduro de 91 aminoácidos. Dez resíduos de cisteínas responsáveis pela formação de cinco pontes dissulfeto, dois sítios de N-glicosilação e três resíduos de prolinas apresentaram-se altamente conservados quando comparados com outras espécies de peixes. A comparação baseada em sequências de aminoácidos de GTHα de 38 espécies de peixes revelou alta identidade do A. gigas com membros das seguintes ordens: Acipenseriformes, Anguiliformes, Siluriformes e Cypriniformes (87,1-89,5%), e, a menor identidade com os Gadiformes e Cyprinodontiformes (55%). A identidade com a análoga sequência de GTHα de Homo sapiens foi de 67%. Para a subunidade ag-FSHβ, a ORF correspondente foi de 381 pb, produzindo uma proteína de 126 aminoácidos com um peptídeo sinal de 18 e um peptídeo maduro de 108 aminoácidos. Quando comparado com as sequências de Anguilla marmorata, Acipenser gueldenstaedtii e Homo sapiens, o peptídeo maduro de ag-FSHβ mostrou conter 12 resíduos de cisteínas responsáveis pela formação de seis pontes dissulfeto, duas prolinas e um sítio de glicosilação perfeitamente conservados, apresentando identidades de 63, 50 e 45% respectivamente em suas sequências de aminoácidos. As árvores filogenéticas construídas mostraram, em geral, que o A. gigas, da ordem dos Osteoglossiformes, se apresenta como grupo irmão dos Clupeocefala, enquanto os Elopomorpha (Anguiliformes) formam o grupo mais basal de todos os teleósteos aqui analisados. / Pirarucu (Arapaima gigas) is a giant fish of the Arapaimidae family native to the Amazon river basin, that can reach 3 meters in length, weighing up to 250 Kg. It is present in Equador, Colombia, Peru, Bolivia and Brazil. This species is in danger of disappearing due to exploitation by the fishing industry and increasing human presence in its natural habitat. In the present work the cDNAs of the gonadotropin -subunit (ag-GTH) and of follicle-stimulating hormone β subunit (ag-FSHβ) were isolated and cloned for the first time. As a consequence, a better understanding of the diversity and evolution of this glycoprotein in fish and its future biotechnological synthesis for reproductive and alimentary purposes will be possible. Both cDNAs of ag-GTHα and ag-FSHβ have been synthesized via reverse transcriptase reaction (RT-PCR) and polymerase chain reaction (PCR) using as a template total RNA extracted from A. gigas pituitary glands. Ag-GTHα- subunit has a coding sequence (open reading frame, ORF) of 348 bp, corresponding to a 115 amino acid protein, with a putative signal peptide of 24 aminoacids and a mature peptide of 91 amino acids. Ten cysteine residues, responsible for the formation of five disulfide linkages, two N-glycosylation sites and three proline residues were found highly conserved when compared to other fish species. A comparison based on the amino acid sequence of the GTHα- subunit from 38 different species of fish showed high identity of A. gigas with members of the following orders: Acipenseriformes, Siluriformes and Cypriniformes (87.1-89.5%), while the lowest identity was found with Gadiformes and Cyprinodontiformes (55%). In comparison with the analogous sequence of Homo sapiens an identity of 67% was found. For the ag-FSHβ subunit an ORF of 381 bp, coding for a 126 amino acid protein, with a signal peptide of 18 and a mature peptide of 108 amino acids, was found. When compared with the Anguilla marmorata, Acipenser gueldenstaedtii and Homo sapiens, the mature peptide of ag-FSHβ showed the presence of the twelve cysteine residues responsible for the formation of six disulfide linkages, two proline residues and one glycosylation site, all of them perfectly conserved. The identity with the mentioned species were 63, 50 and 45% respectively. The obtained phylogenetic trees have shown, in general, that A. gigas of the order of Osteoglossiformes appears as sister group of Clupeocephala, while Elopomorpha (Anguilliformes) forms the most basal group of all analyzed teleosts.
97

Testicular angiogenesis in rats: developmental changes and hormonal stimulation by human chorionic gonadotrophin.

January 1998 (has links)
by Chung Hoi Sing. / Thesis (M.Phil.)--Chinese University of Hong Kong, 1998. / Includes bibliographical references (leaves 92-106). / Abstract also in Chinese. / ABSTRACT --- p.i / 摘要 --- p.iii / ACKNOWLEDGMENT --- p.v / Chapter 1. --- Introduction / Chapter 1.1 --- Angiogenesis in general --- p.1 / Chapter 1.1.1 --- The concept of angiogenesis --- p.1 / Chapter 1.1.2 --- The process of angiogenesis --- p.1 / Chapter 1.2 --- Measurement of angiogenesis --- p.3 / Chapter 1.2.1 --- In vivo assays --- p.3 / Chapter 1.2.2 --- In vitro assays --- p.5 / Chapter 1.3 --- Angiogenic factors --- p.6 / Chapter 1.4 --- Angiogenesis in the female reproductive system --- p.7 / Chapter 1.5 --- Evidence of hormonally-regulated angiogenesis in endocrine tissues --- p.10 / Chapter 1.5.1 --- Ovary --- p.10 / Chapter 1.5.2 --- Thyroid --- p.11 / Chapter 1.6 --- Angiogenesis in the testis --- p.12 / Chapter 1.6.1 --- Structure of testicular vasculature --- p.12 / Chapter 1.6.2 --- Angiogenic factors in the testis --- p.13 / Chapter 1.6.3 --- Vascular effects of hCG/LH in the testis --- p.17 / Chapter 1.6.4 --- Postnatal development of testicular vasculature --- p.17 / Chapter 1.7 --- Aims of the present study --- p.19 / Chapter 2. --- Materials and methods / Chapter 2.1 --- Animals --- p.20 / Chapter 2.2 --- Experimental design --- p.20 / Chapter 2.2.1 --- Testicular angiogenesis in adult rats - hormonal stimulation by hCG --- p.20 / Chapter 2.2.1.1 --- Changes with time after hCG treatment --- p.20 / Chapter 2.2.1.2 --- Effect of Leydig cell depletion --- p.22 / Chapter 2.2.1.3 --- Effect of Leydig cell suppression by subcutaneous testosterone-filled silastic implants --- p.22 / Chapter 2.2.1.4 --- Effect of testicular macrophage activation --- p.24 / Chapter 2.2.1.5 --- Effect of testicular macrophage depletion --- p.26 / Chapter 2.2.2 --- Developmental changes in testicular angiogenesis --- p.29 / Chapter 2.3 --- Perfusion of testes with fixative or Indian Ink --- p.29 / Chapter 2.4 --- Processing of the testes for histological sections --- p.30 / Chapter 2.5 --- Immunohistochemical staining for proliferating cell nuclear antigen (PCNA) --- p.31 / Chapter 2.6 --- Immunohistochemical staining for vascular endothelial growth factor --- p.32 / Chapter 2.7 --- Quantification of PCNA-positive endothelial cells --- p.33 / Chapter 2.8 --- Quantification of blood vessel density --- p.34 / Chapter 2.9 --- Estimation of intertubular area in testis section --- p.35 / Chapter 2.10 --- Preparation of liposome-entrapped dichloromethylene diphosphonate (Cl2MDP-lp) --- p.38 / Chapter 2.11 --- Radioimmunoassay of serum tsetosterone --- p.38 / Chapter 2.12 --- Statistical analyses --- p.40 / Chapter 3. --- Results / Chapter 3.1 --- hCG-induced increase in endothelial cell proliferation in adult rat testes --- p.41 / Chapter 3.1.1 --- Testicular histology --- p.41 / Chapter 3.1.2 --- Changes in the number of PCNA-positive endothelial cells --- p.41 / Chapter 3.1.3 --- Changes in blood vessel density --- p.44 / Chapter 3.1.4 --- Changes in testis weight and serum testosterone concentration --- p.44 / Chapter 3.2 --- Effect of Leydig cell depletion by ethane dimethane sulphonate (EDS) on hCG-induced endothelial cell proliferation in adult rat testes --- p.48 / Chapter 3.2.1 --- Testicular histology --- p.48 / Chapter 3.2.2 --- Changes in the number of PCNA-positive endothelial cells --- p.48 / Chapter 3.2.3 --- Changes in serum testosterone concentration and testis weight --- p.52 / Chapter 3.3 --- Effect ofLeydig cell suppression by testosterone-filled subcutaneous silastic implants on hCG-induced endothelial cell proliferation in adult rat testes --- p.54 / Chapter 3.3.1 --- "Changes in serum testosterone concentration, testis weight, and testicular intertubular area" --- p.54 / Chapter 3.3.2 --- Changes in the number of PCNA-positive endothelial cells --- p.58 / Chapter 3.3.3 --- Changes in the level of vascular endothelial growth factor (VEGF) immunoreactivity in the testis --- p.60 / Chapter 3.4 --- Effect of testicular macrophage activation by polystyrene latex beads on hCG-induced endothelial cell proliferation in adult rat testes --- p.60 / Chapter 3.4.1 --- Testicular histology --- p.60 / Chapter 3.4.2 --- Changes in the number of PCNA-positive endothelial cells --- p.63 / Chapter 3.4.3 --- Changes in testis weight and serum testosterone concentration --- p.65 / Chapter 3.5 --- Effect of testicular macrophage depletion by liposome-entrapped C12MDP treatment on hCG-induced endothelial cell proliferation in adult rat testes --- p.67 / Chapter 3.5.1 --- Testicular histology --- p.68 / Chapter 3.5.2 --- Changes in the number of PCNA-positive endothelial cells --- p.68 / Chapter 3.5.3 --- Changes in testis weight and serum testosterone --- p.72 / Chapter 3.6 --- Endothelial cell proliferation in rat testes during postnatal development --- p.74 / Chapter 3.6.1 --- Changes in the number of PCNA-positive endothelial cells --- p.74 / Chapter 3.6.2 --- Changes in blood vessel density --- p.74 / Chapter 3.6.3 --- Changes in testis weight and intertubular area of the testes --- p.77 / Chapter 4. --- Discussion / Chapter 4.1 --- hCG-induced endothelial cell proliferation and changes in blood vessel density --- p.79 / Chapter 4.2 --- Role of Leydig cells in hCG-induced endothelial cell proliferation in adult rat testes --- p.82 / Chapter 4.3 --- Role of testicular macrophages in hCG-induced endothelial cell proliferation in adult rat testes --- p.86 / Chapter 4.4 --- Testicular angiogenesis during postnatal development --- p.88 / Chapter 5. --- References --- p.92
98

Expression control of zebrafish gonadotropin receptors in the ovary. / CUHK electronic theses & dissertations collection

January 2012 (has links)
卵泡刺激素(FSH)和促黃體激素(LH)是脊椎動物體內的促性腺激素(GTH)。它們通過其相應的GTH受體(GTHR)- FSH受體(FSHR)及LH/絨毛膜性腺激素受體(LHCGR),來調控雌性脊椎動物的主要性腺活動,如卵泡生成和類固醇生成。因此,GTHR的表達水平可控制卵泡細胞對於GTH的反應程度,從而影響脊椎動物的繁殖能力。 / 然而,跟哺乳動物中的資料相比,這些受體的表達調控機制在硬骨魚類中仍然很模糊。此前,我們已經證明了斑馬魚卵泡之fshr和lhcgr的表達譜差異,顯示出lhcgr的表達滯後於fshr的表達。此表達時間之差異引申出兩條有趣的問題:一)甚麼激素能分別調節fshr和lhcgr的表達? 二)這些調控的機制是甚麼?因此,我們發起本研究來解答這些問題。 / 利用培養出來的斑馬魚卵泡細胞,我們展示了雌二醇(E2)是一個有力的GTHR調控激素。雖然E2同時刺激了fshr和lhcgr的表達,但E2對於lhcgr的表達調控效力遠遠比對fshr的高。由於雌激素核受體(nER)的特異拮抗劑(ICI 182,780)能完全抵消E2的效果,表明了E2是通過傳統的nER來直接促進了lhcgr的表達。有趣的是,不能穿越細胞膜的雌二醇-牛血清白蛋白偶聯複合物(E2-BSA)能完全模仿E2的效果,因此我們的證據提出這些nER可能位於細胞膜上。此外,我們運用各種藥劑發現了多種信號分子跟E2調控GTHR的能力有關,包括cAMP、PKA、PI3K、PKC、MEK、MAPK及p38 MAPK。當中以cAMP-PKA的信號傳導最有可能在E2的雙相調控效果起了直接作用,而E2的行動也極依賴其他信號分子的允許作用。 / 除了E2,人絨毛膜促性腺激素(hCG; LH的類似物)、垂體腺苷酸環化酶激活多肽(PACAP)、表皮生長因子(EGF)和胰島素樣生長因子-I(IGF-I)也能有效地調節斑馬魚卵泡細胞的GTHR表達。hCG能大幅下調其受體lhcgr的表達,顯示hCG能令卵泡細胞對GTH脫敏。與此同時,PACAP能瞬時模仿hCG的行動,表明了PACAP很可能是hCG的瞬態下游信號。EGF是一個強烈抑制lhcgr表達的因子,而IGF-I是一個潛在的fshr表達增強因子,均說明了旁分泌因子對GTHR表達調控有關鍵作用。除了這些激素或因子的獨立調控作用,我們進一步發現了E2的效果可能會被它們覆蓋或調節。它們對nER的調控作用可能會造成這種現象。PACAP瞬時減少了esr2a及esr2b的表達量,而EGF則顯著地下調了esr2a。 / 作為第一個在硬骨魚卵巢中對GTHR調控的全面研究,它無疑豐富了我們對卵泡生成過程中GTH的功能及GTHR表達調控的認識。此外,我們成功將目前的研究平台應用於雙酚A(BPA)的研究,進一步展示了本研究平台的潛力,有助於我們未來對各種內分泌干擾物(EDC)的作用機制進行研究。 / Follicle-stimulating hormone (FSH) and luteinizing hormone (LH) are the gonadotropins (GTHs), which bind to their cognate GTH receptors (GTHRs), FSH receptor (FSHR) and LH/choriogonadotropin receptor (LHCGR), to mediate major gonadal events in female vertebrates, including folliculogenesis and steroidogenesis. The expression level of GTHRs, therefore, controls the responsiveness of follicle cells to GTHs and hence governs the vertebrate reproduction. / However, compared with the information in mammals, the expression control of these receptors in teleosts remains largely unknown. Previously, we have demonstrated the differential expression profiles of fshr and lhcgr in the zebrafish folliculogenesis, showing that lhcgr expression lags behind fshr expression. This temporal difference between fshr and lhcgr expression has raised two interesting questions: 1) What hormones regulate the differential expression of fshr and lhcgr? and 2) What are the control mechanisms of these regulations? The present study was initiated to answer these questions. / With the primary zebrafish follicle cell cultures, we demonstrated that estradiol (E2) was a potent differential regulator of GTHRs. Although E2 increased both fshr and lhcgr expression, the up-regulatory potency of E2 on lhcgr was much greater than that on fshr. E2 directly promoted lhcgr expression via classical nuclear estrogen receptors (nERs) since nER-specific antagonist (ICI 182,780) completely abolished the E2 effect. Interestingly, our evidence suggested that these nERs could be localized on the plasma membrane because the membrane-impermeable form of estrogen (E2-BSA) fully mimicked the actions of E2. Furthermore, by applying various pharmaceutical agents, we revealed the involvement of multiple signaling molecules, including cAMP, PKA, PI3K, PKC, MEK, MAPK and p38 MAPK. The cAMP-PKA pathway likely played a direct role in the biphasic actions of E2 while the E2 actions were also greatly dependent on the permissive actions of other signaling molecules. / Apart from the sex steroid E2, human chorionic gonadotropin (hCG; as a LH analogue), pituitary adenlyate cyclase-activating peptide (PACAP), epidermal growth factor (EGF) and insulin-like growth factor-I (IGF-I) also significantly regulated GTHR expression in the zebrafish follicle cells. hCG drastically down-regulated its receptor, lhcgr, suggesting that hCG could desensitize the follicle cells to respond to GTH. Meanwhile, PACAP transiently mimicked the actions of hCG, indicating that PACAP was likely a transient downstream mediator of hCG. EGF was another strong suppressor of lhcgr expression while IGF-I was a potential fshr expression enhancer, which highlighted the crucial roles of paracrine factors in the regulation of GTHRs. In addition to the regulatory effect of these individual hormones or factors, we further revealed that the E2 action could be overridden or modulated by them. Their regulatory effects on the expression of nERs might contribute to this phenomenon. PACAP transiently reduced esr2a and esr2b expression while EGF significantly down-regulated esr2a. / As the first comprehensive study of GTHR regulation in the teleost ovary, the present study certainly enriched our knowledge in the functions of GTHs and the expression control of GTHRs during folliculogenesis. By applying the current research platform on the study of bisphenol A (BPA), an endocrine-disrupting chemical (EDC), the present study further highlighted the potential of this research platform to contribute to the future action mechanism studies of various EDCs. / Detailed summary in vernacular field only. / Detailed summary in vernacular field only. / Detailed summary in vernacular field only. / Detailed summary in vernacular field only. / Detailed summary in vernacular field only. / Liu, Ka Cheuk. / Thesis (Ph.D.)--Chinese University of Hong Kong, 2012. / Includes bibliographical references (leaves 159-212). / Electronic reproduction. Hong Kong : Chinese University of Hong Kong, [2012] System requirements: Adobe Acrobat Reader. Available via World Wide Web. / Abstract also in Chinese. / Abstract (in English) --- p.i / Abstract (in Chinese) --- p.iii / Acknowledgement --- p.v / Table of contents --- p.vi / List of figures and tables --- p.xii / Symbols and abbreviations --- p.xv / Chapter Chapter 1 --- General Introduction / Chapter 1.1 --- Hypothalamic-pituitary-gonadal axis / Chapter 1.1.1 --- Overview --- p.1 / Chapter 1.1.2 --- Gonadotropin-releasing hormone --- p.1 / Chapter 1.2 --- Folliculogenesis / Chapter 1.2.1 --- Structure of ovarian follicles --- p.2 / Chapter 1.2.2 --- Stages of folliculogenesis --- p.3 / Chapter 1.3 --- Gonadotropins and gonadotropin receptors / Chapter 1.3.1 --- History of teleost gonadotropin and gonadotropin receptors --- p.5 / Chapter 1.3.2 --- Structure --- p.6 / Chapter 1.3.3 --- Function --- p.7 / Chapter 1.3.4 --- GTH-GTHR specificity --- p.9 / Chapter 1.3.5 --- Signal transduction --- p.10 / Chapter 1.3.6 --- Expression profile of gonadotropin receptors --- p.11 / Chapter 1.3.7 --- Regulation of gonadotropin receptors --- p.12 / Chapter 1.4 --- Objectives and significances of the project --- p.14 / Chapter 1.5 --- Figure legends --- p.16 / Chapter 1.6 --- Figures --- p.18 / Chapter Chapter 2 --- Differential Regulation of Gonadotropin Receptors (fshr and lhcgr) by Estradiol in the Zebrafish Ovary Involves Nuclear Estrogen Receptors That Are Likely Located on the Plasma Membrane / Chapter 2.1 --- Introduction --- p.24 / Chapter 2.2 --- Materials and methods / Chapter 2.2.1 --- Animals --- p.25 / Chapter 2.2.2 --- Hormones and chemicals --- p.26 / Chapter 2.2.3 --- Primary follicle cell culture and drug treatment --- p.26 / Chapter 2.2.4 --- Ovarian fragment incubation --- p.27 / Chapter 2.2.5 --- Total RNA extraction and real-time qPCR --- p.27 / Chapter 2.2.6 --- Western blot analysis --- p.27 / Chapter 2.2.7 --- SEAP reporter gene assay --- p.28 / Chapter 2.2.8 --- Data analysis --- p.28 / Chapter 2.3 --- Results / Chapter 2.3.1 --- Differential stimulation of fshr and lhcgr expression in ovarian fragments and follicle cells by estradiol but not testosterone --- p.28 / Chapter 2.3.2 --- Potentiation of follicle cell responsiveness to hCG by E2 pretreatment --- p.30 / Chapter 2.3.4 --- Evidence for transcription but not translation-dependent up-regulation of lhcgr by E2 --- p.30 / Chapter 2.3.5 --- Evidence for the involvement of nuclear estrogen receptors but not G protein-coupled estrogen receptor 1 (Gper) in E2-stimulated lhcgr expression --- p.31 / Chapter 2.3.6 --- Evidence for possible localization of estrogen receptors on the plasma membrane --- p.32 / Chapter 2.3.7 --- MAPK dependence of E2 effect on lhcgr expression --- p.32 / Chapter 2.4 --- Discussion --- p.33 / Chapter 2.5 --- Table --- p.38 / Chapter 2.6 --- Figure legends --- p.39 / Chapter 2.7 --- Figures --- p.43 / Chapter Chapter 3 --- Signal Transduction Mechanisms of the Biphasic Estrogen Actions in the Regulation of Gonadotropin Receptors (fshr and lhcgr) in the Zebrafish Ovary / Chapter 3.1 --- Introduction --- p.50 / Chapter 3.2 --- Materials and methods / Chapter 3.2.1 --- Animals --- p.52 / Chapter 3.2.2 --- Hormones and chemicals --- p.52 / Chapter 3.2.3 --- Primary cell culture and drug treatment --- p.52 / Chapter 3.2.4 --- Total RNA extraction and real-time qPCR --- p.52 / Chapter 3.2.5 --- Fractionation of follicle cells --- p.52 / Chapter 3.2.6 --- Western blot analysis --- p.52 / Chapter 3.2.7 --- Statistical analysis --- p.53 / Chapter 3.3 --- Results / Chapter 3.3.1 --- Biphasic roles of cAMP-PKA pathway --- p.53 / Chapter 3.3.2 --- Effects of p38 MAPK inhibition --- p.54 / Chapter 3.3.3 --- Effects of PKC and PI3K inhibition --- p.54 / Chapter 3.4 --- Discussion --- p.55 / Chapter 3.5 --- Figure legends --- p.59 / Chapter 3.6 --- Figures --- p.61 / Chapter Chapter 4 --- Gonadotropin (hCG) and pituitary adenylate cyclase-activating peptide (PACAP) down-regulate basal and E2-stimulated gonadotropin receptors (fshr and lhcgr) in the zebrafish ovary via a cAMP-dependent but PKA-independent pathway / Chapter 4.1 --- Introduction --- p.66 / Chapter 4.2 --- Materials and methods / Chapter 4.2.1 --- Animals --- p.69 / Chapter 4.2.2 --- Hormones and chemicals --- p.69 / Chapter 4.2.3 --- Primary cell culture and drug treatment --- p.69 / Chapter 4.2.4 --- Total RNA extraction and real-time qPCR --- p.69 / Chapter 4.2.5 --- Statistical analysis --- p.69 / Chapter 4.3 --- Results / Chapter 4.3.1 --- Down-regulation of fshr and lhcgr by hCG --- p.69 / Chapter 4.3.2 --- Differential regulation of fshr and lhcgr by PACAP --- p.70 / Chapter 4.3.3 --- Inhibition of E2-regulated fshr and lhcgr expression by hCG --- p.71 / Chapter 4.3.4 --- Suppressive effects of PACAP on E2-induced fshr and lhcgr expression --- p.71 / Chapter 4.3.5 --- Role of cAMP in hCG and PACAP actions --- p.72 / Chapter 4.4 --- Discussion --- p.73 / Chapter 4.5 --- Figure legends --- p.78 / Chapter 4.6 --- Figures --- p.80 / Chapter Chapter 5 --- Paracrine regulation of gonadotropin receptors (fshr and lhcgr) by ovarian growth factors: epidermal growth factor (EGF) and insulin-like growth factor-I (IGF-I) / Chapter 5.1 --- Introduction --- p.85 / Chapter 5.2 --- Materials and methods / Chapter 5.2.1 --- Animals --- p.88 / Chapter 5.2.2 --- Hormones and chemicals --- p.88 / Chapter 5.2.3 --- Primary cell culture and drug treatment --- p.88 / Chapter 5.2.4 --- Total RNA extraction and real-time qPCR --- p.88 / Chapter 5.2.5 --- Statistical analysis --- p.88 / Chapter 5.3 --- Results / Chapter 5.3.1 --- Biphasic down-regulation of lhcgr by EGF --- p.89 / Chapter 5.3.2 --- Evidence for EGFR involvement --- p.89 / Chapter 5.3.3 --- Minor role of MEK-MAPK3/1 pathway in the EGF effect on lhcgr expression --- p.90 / Chapter 5.3.4 --- Up-regulation of fshr by IGF-I --- p.90 / Chapter 5.3.5 --- Evidence for IGF-IR involvement --- p.91 / Chapter 5.3.6 --- Role of PI3K-Akt pathway in IGF-I action --- p.91 / Chapter 5.3.7 --- Role of EGF and EGFR in E2-induced GTHR expression --- p.91 / Chapter 5.3.8 --- Role of IGF-I and IGF-IR in E2-induced GTHR expression --- p.91 / Chapter 5.4 --- Discussion --- p.92 / Chapter 5.5 --- Figure legends --- p.98 / Chapter 5.6 --- Figures --- p.100 / Chapter Chapter 6 --- Regulation of estrogen receptor subtypes (esr1, esr2a and esr2b): a possible mechanism to modulate estradiol-stimulated lhcgr expression in the zebrafish ovary / Chapter 6.1 --- Introduction --- p.107 / Chapter 6.2 --- Materials and methods / Chapter 6.2.1 --- Animals --- p.110 / Chapter 6.2.2 --- Hormones and chemicals --- p.110 / Chapter 6.2.3 --- Staging ovarian follicles --- p.110 / Chapter 6.2.4 --- Primary cell culture and drug treatment --- p.110 / Chapter 6.2.5 --- Total RNA extraction and real-time qPCR --- p.110 / Chapter 6.2.6 --- Statistical analysis --- p.111 / Chapter 6.3 --- Results / Chapter 6.3.1 --- Expression profiles of estrogen receptors (ERs) in zebrafish folliculogenesis --- p.111 / Chapter 6.3.2 --- Homologous regulation of nERs by E2 --- p.111 / Chapter 6.3.3 --- Regulation of nERs by endocrine hormones (hCG and PACAP) --- p.112 / Chapter 6.3.4 --- Regulation of nERs by ovarian paracrine growth factors (EGF and IGF-I) --- p.112 / Chapter 6.3.5 --- Role of cAMP in nER regulation --- p.113 / Chapter 6.3.6 --- Role of PKA in nER regulation --- p.113 / Chapter 6.4 --- Discussion --- p.114 / Chapter 6.5 --- Figure legends --- p.119 / Chapter 6.6 --- Figures --- p.121 / Chapter Chapter 7 --- Estrogenic Action Mechanisms of Bisphenol A / Chapter 7.1 --- Introduction --- p.127 / Chapter 7.2 --- Materials and methods / Chapter 7.2.1 --- Animals --- p.129 / Chapter 7.2.2 --- Hormones and chemicals --- p.129 / Chapter 7.2.3 --- Primary cell culture and drug treatment --- p.129 / Chapter 7.2.4 --- Total RNA extraction and real-time qPCR --- p.129 / Chapter 7.2.5 --- Statistical analysis --- p.130 / Chapter 7.3 --- Results / Chapter 7.3.1 --- Expression of fshr and lhcgr interfered by BPA --- p.130 / Chapter 7.3.2 --- Signaling mechanism of BPA-induced lhcgr up-regulation --- p.130 / Chapter 7.3.3 --- Dependence of transcription and translation in BPA-induced lhcgr expression --- p.131 / Chapter 7.3.4 --- Evidence for the involvement of nuclear estrogen receptors in the BPA actions --- p.131 / Chapter 7.3.5 --- Interference on E2-induced lhcgr expression by BPA --- p.131 / Chapter 7.4 --- Discussion --- p.132 / Chapter 7.5 --- Figure legends --- p.136 / Chapter 7.6 --- Figures --- p.138 / Chapter Chapter 8: --- General Discussion / Chapter 8.1 --- Estradiol as a differential regulator of gonadotropin receptors --- p.143 / Chapter 8.2 --- Conserved role of estradiol with differential action mechanisms in lhcgr regulation of mammals and teleosts --- p.144 / Chapter 8.3 --- Involvement of classical estrogen receptors that are likely located on the plasma membrane --- p.145 / Chapter 8.4 --- Biphasic response of lhcgr to estradiol and the underlying signal transduction mechanisms --- p.145 / Chapter 8.5 --- Desensitization of follicle cells to gonadotropins by hCG --- p.146 / Chapter 8.6 --- Paracrine control of gonadotropin receptors by ovarian growth factors --- p.147 / Chapter 8.7 --- Interaction of the estrogen action with other endocrine and paracrine signals --- p.148 / Chapter 8.8 --- Action mechanism studies of an endocrine-disrupting chemical: bisphenol A --- p.150 / Chapter 8.9 --- Conclusion --- p.151 / Chapter 8.10 --- Figure legends --- p.153 / Chapter 8.11 --- Figures --- p.155 / References --- p.159
99

Maternal serum alpha-fetoprotein and total beta-human chorionic gonadotrophin in twin pregnancies during mid-trimester: their implications for adverse pregnancy outcomes.

January 1997 (has links)
Cheung Kwok Lung. / Thesis (M.Phil.)--Chinese University of Hong Kong, 1997. / Includes bibliographical references (leaves 123-136). / ABSTRACT (English) --- p.i / ACKNOWLEDGMENTS --- p.1 / LIST OF FIGURES --- p.3 / LIST OF TABLES --- p.5 / LIST OF ABBREVIATIONS --- p.7 / Chapter I. --- INTRODUCTION AND OBJECTIVES --- p.8 / Chapter II. --- LITERATURE REVIEWS --- p.11 / Chapter II.A. --- Maternal Serum Alpha-fetoprotein Screeningin Singleton Pregnancies --- p.11 / Chapter II.A.1. --- Physiology of Alpha-fetoprotein --- p.12 / Chapter II.A.2. --- Historical Background of Screening by Alpha- fetoprotein --- p.12 / Chapter II.A.3. --- Factors that Influence Maternal Serum Alpha- fetoprotein Concentration --- p.13 / Chapter ILA.4. --- Elevated Maternal Serum Alpha-fetoprotein Concentration and Adverse Pregnancy Outcomes and Complications --- p.14 / Chapter II.A.4.a. --- Low Birth Weight --- p.16 / Chapter II.A.4.b. --- Fetal Loss --- p.17 / Chapter II.A.4.c. --- Pregnancy Induced Hypertension --- p.18 / Chapter II.B. --- Maternal Serum Human Chorionic Gonadotrophin Screening in Singleton Pregnancies --- p.18 / Chapter II.B.1. --- Physiology of Human Chorionic Gonadotrophin --- p.18 / Chapter II.B.2. --- Historical Background of Screening by Human Chorionic Gonadotrophin --- p.20 / Chapter II.B.3. --- Factors that Influence Maternal Serum Human Chorionic Gonadotrophin --- p.21 / Chapter II.B.4. --- Elevated Maternal Serum Human Chorionic Gonadotrophin Concentration and Pregnancy Complications --- p.21 / Chapter II.B.5. --- Maternal Serum AFP and hCG Concentrations and Adverse Outcomes or Complications in Twin Pregnancies --- p.23 / Chapter II.C. --- Mechanism for the Association between Adverse Outcomes and Elevated Maternal Serum Alpha- fetoprotein and Human Chorionic Gonadotrophin --- p.25 / Chapter III. --- METHODS --- p.28 / Chapter III.A. --- Study Population --- p.28 / Chapter III.B. --- Sample Collection and Analysis --- p.29 / Chapter III.C. --- Clinical Information --- p.30 / Chapter III.D. --- Microparticle Enzyme Immunoassay --- p.30 / Chapter III.D.1. --- Principles --- p.30 / Chapter III.D.1.a. --- Reaction Process --- p.31 / Chapter III.D.1.b. --- MEIA Assembly --- p.33 / Chapter III.D.1.c. --- Operation --- p.34 / Chapter III.D.2. --- AFP Assay --- p.34 / Chapter III.D.2.a. --- AFP Reagents --- p.34 / Chapter III.D.2.b. --- Sample Dilution --- p.36 / Chapter III.D.3. --- Total p-hCG Assay --- p.37 / Chapter III.D.3.a. --- Total p-hCG Reagents --- p.37 / Chapter III.D.3.b. --- Sample Dilution --- p.39 / Chapter III.D.4. --- Intra- and Inter-assay Variation --- p.39 / Chapter III.E. --- Data Handling --- p.42 / Chapter III.F. --- Statistical Analysis --- p.42 / Chapter III.F.1. --- Calculations of Median Values of Maternal Serum Alpha-fetoprotein and Human Chorionic Gonadotrophin Concentrations --- p.42 / Chapter III.F.2. --- Analysis for Adverse Outcomes or Complications --- p.43 / Chapter III.F.3. --- Adjustment of Alpha-fetoprotein and Human Chorionic Gonadotrophin for Gestational Age and Maternal Weight --- p.46 / Chapter IV. --- RESULTS --- p.48 / Chapter IV.A. --- Median Values of Maternal Serum Alpha-fetoprotein Human Chorionic Gonadotrophin --- p.48 / Chapter IV.B. --- Prediction of Adverse Outcomes by Maternal Serum Alpha-fetoprotein and Human Chorionic Gonadotrophin --- p.60 / Chapter IV.B. l. --- Preterm Delivery --- p.60 / Chapter IV.B.2. --- Spontaneous Preterm Delivery --- p.64 / Chapter IV.B.3. --- Premature Delivery --- p.68 / Chapter IV.B.4. --- Spontaneous Premature Delivery --- p.68 / Chapter IV.B.5. --- Other Outcomes or Complications --- p.72 / Chapter IV.B.6. --- Single Predictor for Most Adverse Outcomes --- p.74 / Chapter IV.C. --- Adjustment of Maternal Serum Alpha-fetoprotein and Human Chorionic Gonadotrophin for Maternal Weight and Gestational Age --- p.75 / Chapter IV.C.1. --- Distribution of Alpha-fetoprotein and Human Chorionic Gonadotrophin during Mid-trimester --- p.76 / Chapter IV.C.2. --- Adjustment of Alpha-fetoprotein for Maternal Weight and Gestational Age --- p.79 / Chapter IV.C.3. --- Adjustment of Human Chorionic Gonadotrophin for Maternal Weight and Gestational Age --- p.80 / Chapter IV.D. --- Predictiveness of Alpha-fetoprotein and Human Chorionic Gonadotrophin for Adverse Outcomes after Adjusted for Maternal Weight and Gestational Age --- p.83 / Chapter IV.D.l. --- Preterm Delivery --- p.86 / Chapter IV.D.2. --- Spontaneous Preterm Delivery --- p.86 / Chapter IV.D.3. --- Premature Delivery --- p.92 / Chapter IV.D.4. --- Spontaneous Premature Delivery --- p.92 / Chapter IV.D.5. --- Other Adverse Outcomes or Complications --- p.98 / Chapter IV.D.6. --- Single Predictor for Most Adverse Outcomes --- p.98 / Chapter V. --- DISCUSSIONS --- p.100 / Chapter V.A. --- Median Values of Maternal Serum Alpha-fetoprotein and Human Chorionic Gonadtrophin --- p.100 / Chapter V.B. --- Maternal Serum Alpha-fetoprotein and Human Chorionic Gonadotrophin Screening for Adverse Outcomes --- p.103 / Chapter V.C. --- Adjustment of Alpha-fetoprotein and Human Chorionic Gonadotrophin for Maternal Weight and Gestational Age --- p.109 / Chapter V.D. --- Predictiveness of Alpha-fetoprotein and Human Chorionic Gonadotrophin for Adverse Outcomes after Maternal Weight and Gestational Age Adjustment --- p.112 / Chapter V.E. --- Conclusions --- p.113 / Chapter V.F. --- Future Directions --- p.116 / APPENDIX 1 DATA BASE OF CLINICAL INFORMATION --- p.117 / APPENDIX 2 SEVERITY AND CLASSIFICATION OF PREGNANCY INDUCED HYPERTENSION --- p.122 / REFERENCES --- p.123
100

Gonadotrophins and cytokines in ovarian epithelial cancer / John Alexander Latimer.

Latimer, John Alexander. January 1997 (has links)
Bibliography: p. 159-193. / x, 200 p. : / Title page, contents and abstract only. The complete thesis in print form is available from the University Library. / This thesis compares the rates of mitotic activity of the ovarian surface epithelium (OSE) and the peritoneal mesothelium (PM) and the effects of ovarian hyperstimulation using a rodent model. The study provides also information about cytokine expression and production in benign and malignant ovarian tissue, both in humans and animals. / Thesis (M.D.)--University of Adelaide, Dept. of Obstetrics and Gynaecology, 1997?

Page generated in 0.0596 seconds