Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Hum Reprod ; 37(6): 1244-1254, 2022 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-35413103

RESUMO

STUDY QUESTION: Could changes in transforming growth factor ß (TGFß) signalling during foetal ovary development alter the expression of polycystic ovary syndrome (PCOS) candidate genes leading to a predisposition to PCOS? SUMMARY ANSWER: TGFß signalling molecules are dynamically expressed during foetal ovary development and TGFß1 inhibits expression of the androgen receptor (AR) and 7 (INSR, C8H9orf3, RAD50, ERBB3, NEIL2, IRF1 and ZBTB16) of the 25 PCOS candidate genes in foetal ovarian fibroblasts in vitro, whilst increasing expression of the AR cofactor TGFß-induced transcript 1 (TGFB1I1 or Hic5). WHAT IS KNOWN ALREADY: The ovarian stroma arises from the mesonephros during foetal ovary development. Changes in the morphology of the ovarian stroma are cardinal features of PCOS. The ovary is more fibrous and has more tunica and cortical and subcortical stroma. It is not known why this is and when this arises. PCOS has a foetal origin and perhaps ovarian stroma development is altered during foetal life to determine the formation of a polycystic ovary later in life. PCOS also has a genetic origin with 19 loci containing 25 PCOS candidate genes. In many adult tissues, TGFß is known to stimulate fibroblast replication and collagen deposition in stroma, though it has the opposite effect in the non-scaring foetal tissues. Our previous studies showed that TGFß signalling molecules [TGFßs and their receptors, latent TGFß binding proteins (LTBPs) and fibrillins, which are extracellular matrix proteins that bind LTBPs] are expressed in foetal ovaries. Also, we previously showed that TGFß1 inhibited expression of AR and 3 PCOS candidate genes (INSR, C8H9orf3 and RAD50) and stimulated expression of TGFB1I1 in cultured foetal ovarian fibroblasts. STUDY DESIGN, SIZE, DURATION: We used Bos taurus for this study as we can ethically collect foetal ovaries from across the full 9-month gestational period. Foetal ovaries (62-276 days, n = 19) from across gestation were collected from pregnant B. taurus cows for RNA-sequencing (RNA-seq) analyses. Foetal ovaries from B. taurus cows were collected (160-198 days, n = 6) for culture of ovarian fibroblasts. PARTICIPANTS/MATERIALS, SETTING, METHODS: RNA-seq transcriptome profiling was performed on foetal ovaries and the data on genes involved in TGFß signalling were extracted. Cells were dispersed from foetal ovaries and fibroblasts cultured and treated with TGFß1. The effects of TGFß regulation on the remaining eight PCOS candidate genes not previously studied (ERBB3, MAPRE1, FDFT1, NEIL2, ARL14EP, PLGRKT, IRF1 and ZBTB16) were examined. MAIN RESULTS AND THE ROLE OF CHANCE: Many TGFß signalling molecules are expressed in the foetal ovary, and for most, their expression levels increased accross gestation (LTBP1/2/3/4, FBN1, TGFB2/3, TGFBR2/3 and TGFB1I1), while a few decreased (FBN3, TGFBR3L, TGFBI and TGFB1) and others remained relatively constant (TGFBRAP1, TGFBR1 and FBN2). TGFß1 significantly decreased expression of PCOS candidate genes ERBB3, NEIL2, IRF1 and ZBTB16 in cultured foetal ovarian fibroblasts. LARGE SCALE DATA: The FASTQ files, normalized data and experimental information have been deposited in the Gene Expression Omnibus (GEO) accessible by accession number GSE178450. LIMITATIONS, REASONS FOR CAUTION: Regulation of PCOS candidate genes by TGFß was carried out in vitro and further studies in vivo are required. This study was carried out in bovine where foetal ovaries from across all of the 9-month gestational period were available, unlike in the human where it is not ethically possible to obtain ovaries from the second half of gestation. WIDER IMPLICATIONS OF THE FINDINGS: From our current and previous results we speculate that inhibition of TGFß signalling in the foetal ovary is likely to (i) increase androgen sensitivity by enhancing expression of AR, (ii) increase stromal activity by stimulating expression of COL1A1 and COL3A1 and (iii) increase the expression of 7 of the 25 PCOS candidate genes. Thus inhibition of TGFß signalling could be part of the aetiology of PCOS or at least the aetiology of polycystic ovaries. STUDY FUNDING/COMPETING INTEREST(S): Funding was received from Adelaide University China Fee Scholarship (M.L.), Australian Research Training Program (R.A.) and the Faculty of Health and Medical Science Divisional Scholarship (R.A.), Adelaide Graduate Research Scholarships (R.A. and N.A.B.), Australia Awards Scholarship (M.D.H.), Robinson Research Institute Career Development Fellowship (K.H.) and Building On Ideas Grant (K.H.), National Health and Medical Research Council of Australia Centre for Research Excellence in the Evaluation, Management and Health Care Needs of Polycystic Ovary Syndrome (N.A.B., M.D.H. and R.J.R.; GTN1078444) and the Centre for Research Excellence on Women's Health in Reproductive life (R.A., R.J.R. and K.H.; GTN1171592) and the UK Medical Research Council (R.A.A.; grant no. G1100357). The funders did not play any role in the study design, data collection and analysis, decision to publish or preparation of the manuscript. The authors of this manuscript have nothing to declare and no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.


Assuntos
Síndrome do Ovário Policístico , Animais , Austrália , Bovinos , Feminino , Feto , Humanos , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Gravidez , Fator de Crescimento Transformador beta
2.
Biol Reprod ; 103(4): 840-853, 2020 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-32678441

RESUMO

Polycystic ovary syndrome (PCOS) appears to have a genetic predisposition and a fetal origin. We compared the expression levels of 25 PCOS candidate genes from adult control and PCOS human ovaries (n = 16) using microarrays. Only one gene was potentially statistically different. Using qRT-PCR, expression of PCOS candidate genes was examined in bovine fetal ovaries from early stages when they first developed stroma through to completion of development (n = 27; 60-270 days of gestation). The levels of ERBB3 mRNA negatively correlated with gestational age but positively with HMGA2, FBN3, TOX3, GATA4, and DENND1A.X1,2,3,4, previously identified as correlated with each other and expressed early. PLGRKT and ZBTB16, and less so IRF1, were also correlated with AMH, FSHR, AR, INSR, and TGFB1I1, previously identified as correlated with each other and expressed late. ARL14EP, FDFT1, NEIL2, and MAPRE1 were expressed across gestation and not correlated with gestational age as shown previously for THADA, ERBB4, RAD50, C8H9orf3, YAP1, RAB5B, SUOX, and KRR1. LHCGR, because of its unusual bimodal expression pattern, had some unusual correlations with other genes. In human ovaries (n = 15; <150 days of gestation), ERBB3.V1 and ERBB3.VS were expressed and correlated negatively with gestational age and positively with FBN3, HMGA2, DENND1A.V1,3,4, DENND1A.V1-7, GATA4, and FSHR, previously identified as correlated with each other and expressed early. Thus, the general lack of differential expression of candidate genes in adult ovaries contrasting with dynamic patterns of gene expression in fetal ovaries is consistent with a vulnerability to disturbance in the fetal ovary that may underpin development of PCOS.


Assuntos
Feto/metabolismo , Ovário/metabolismo , Síndrome do Ovário Policístico/metabolismo , Animais , Bovinos , Feminino , Regulação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Análise Serial de Proteínas
3.
Reproduction ; 157(6): 545-565, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30925461

RESUMO

The ovary has specialised stromal compartments, including the tunica albuginea, interstitial stroma and theca interna, which develops concurrently with the follicular antrum. To characterise the molecular determinants of these compartments, stroma adjacent to preantral follicles (pre-theca), interstitium and tunica albuginea were laser microdissected (n = 4 per group) and theca interna was dissected from bovine antral follicles (n = 6). RNA microarray analysis showed minimal differences between interstitial stroma and pre-theca, and these were combined for some analyses and referred to as stroma. Genes significantly upregulated in theca interna compared to stroma included INSL3, LHCGR, HSD3B1, CYP17A1, ALDH1A1, OGN, POSTN and ASPN. Quantitative RT-PCR showed significantly greater expression of OGN and LGALS1 in interstitial stroma and theca interna versus tunica and greater expression of ACD in tunica compared to theca interna. PLN was significantly higher in interstitial stroma compared to tunica and theca. Ingenuity pathway, network and upstream regulator analyses were undertaken. Cell survival was also upregulated in theca interna. The tunica albuginea was associated with GPCR and cAMP signalling, suggesting tunica contractility. It was also associated with TGF-ß signalling and increased fibrous matrix. Western immunoblotting was positive for OGN, LGALS1, ALDH1A1, ACD and PLN with PLN and OGN highly expressed in tunica and interstitial stroma (each n = 6), but not in theca interna from antral follicles (n = 24). Immunohistochemistry localised LGALS1 and POSTN to extracellular matrix and PLN to smooth muscle cells. These results have identified novel differences between the ovarian stromal compartments.


Assuntos
Biomarcadores/metabolismo , Folículo Ovariano/metabolismo , Ovário/metabolismo , Células Estromais/metabolismo , Células Tecais/metabolismo , Transcriptoma , Animais , Bovinos , Feminino , Folículo Ovariano/citologia , Ovário/citologia , Transdução de Sinais , Células Estromais/citologia , Células Tecais/citologia
4.
Reproduction ; 152(2): 127-37, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27222596

RESUMO

Fibrillins 1-3 are stromal extracellular matrix proteins that play important roles in regulating TGFß activity, which stimulates fibroblasts to proliferate and synthesize collagen. In the developing ovary, the action of stroma is initially necessary for the formation of ovigerous cords and subsequently for the formation of follicles and the surface epithelium of the ovary. FBN3 is highly expressed only in early ovarian development and then it declines. In contrast, FBN1 and 2 are upregulated in later ovarian development. We examined the expression of FBN1-3 in bovine and human fetal ovaries. We used cell dispersion and monolayer culture, cell passaging and tissue culture. Cells were treated with growth factors, hormones or inhibitors to assess the regulation of expression of FBN1-3 When bovine fetal ovarian tissue was cultured, FBN3 expression declined significantly. Treatment with TGFß-1 increased FBN1 and FBN2 expression in bovine fibroblasts, but did not affect FBN3 expression. Additionally, in cultures of human fetal ovarian fibroblasts (9-17weeks gestational age), the expression of FBN1 and FBN2 increased with passage, whereas FBN3 dramatically decreased. Treatment with activin A and a TGFß family signaling inhibitor, SB431542, differentially regulated the expression of a range of modulators of TGFß signaling and of other growth factors in cultured human fetal ovarian fibroblasts suggesting that TGFß signaling is differentially involved in the regulation of ovarian fibroblasts. Additionally, since the changes in FBN1-3 expression that occur in vitro are those that occur with increasing gestational age in vivo, we suggest that the fetal ovarian fibroblasts mature in vitro.


Assuntos
Ativinas/metabolismo , Feto/metabolismo , Fibrilinas/metabolismo , Regulação da Expressão Gênica , Ovário/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Bovinos , Células Cultivadas , Feminino , Feto/citologia , Fibrilina-1/metabolismo , Fibrilina-2/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Ovário/citologia , Gravidez
5.
BMC Genomics ; 15: 24, 2014 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-24422759

RESUMO

BACKGROUND: At later stages of folliculogenesis, the mammalian ovarian follicle contains layers of epithelial granulosa cells surrounding an antral cavity. During follicle development granulosa cells replicate, secrete hormones and support the growth of the oocyte. In cattle, the follicle needs to grow > 10 mm in diameter to allow an oocyte to ovulate, following which the granulosa cells cease dividing and differentiate into the specialised cells of the corpus luteum. To better understand the molecular basis of follicular growth and granulosa cell maturation, we undertook transcriptome profiling of granulosa cells from small (< 5 mm; n = 10) and large (> 10 mm, n = 4) healthy bovine follicles using Affymetrix microarrays (24,128 probe sets). RESULTS: Principal component analysis for the first two components and hierarchical clustering showed clustering into two groups, small and large, with the former being more heterogeneous. Size-frequency distributions of the coefficient of variation of the signal intensities of each probe set also revealed that small follicles were more heterogeneous than the large. IPA and GO enrichment analyses revealed that processes of axonal guidance, immune signalling and cell rearrangement were most affected in large follicles. The most important networks were associated with: (A) Notch, SLIT/ROBO and PI3K signalling, and (B) ITGB5 and extracellular matrix signalling through extracellular signal related kinases (ERKs). Upstream regulator genes which were predicted to be active in large follicles included STAT and XBP1. By comparison, developmental processes such as those stimulated by KIT, IHH and MEST were most active in small follicles. MGEA5 was identified as an upstream regulator in small follicles. It encodes an enzyme that modifies the activity of many target proteins, including those involved in energy sensing, by removal of N-acetylglucosamine from serine and threonine residues. CONCLUSIONS: Our data suggest that as follicles enlarge more genes and/or pathways are activated than are inactivated, and gene expression becomes more uniform. These findings could be interpreted that either the cells in large follicles are more uniform in their gene expression, or that follicles are more uniform or a combination of both and that additional factors, such as LH, are additionally controlling the granulosa cells.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Células da Granulosa/metabolismo , Folículo Ovariano/metabolismo , Animais , Bovinos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Células da Granulosa/citologia , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Oócitos/crescimento & desenvolvimento , Folículo Ovariano/crescimento & desenvolvimento , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Análise de Componente Principal , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Fatores de Transcrição de Fator Regulador X , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
BMC Genomics ; 15: 40, 2014 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-24438529

RESUMO

BACKGROUND: The major function of the ovary is to produce oocytes for fertilisation. Oocytes mature in follicles surrounded by nurturing granulosa cells and all are enclosed by a basal lamina. During growth, granulosa cells replicate and a large fluid-filled cavity (the antrum) develops in the centre. Only follicles that have enlarged to over 10 mm can ovulate in cows. In mammals, the number of primordial follicles far exceeds the numbers that ever ovulate and atresia or regression of follicles is a mechanism to regulate the number of oocytes ovulated and to contribute to the timing of ovulation. To better understand the molecular basis of follicular atresia, we undertook transcriptome profiling of granulosa cells from healthy (n = 10) and atretic (n = 5) bovine follicles at early antral stages (< 5 mm). RESULTS: Principal Component Analysis (PCA) and hierarchical classification of the signal intensity plots for the arrays showed primary clustering into two groups, healthy and atretic. These analyses and size-frequency plots of coefficients of variation of signal intensities revealed that the healthy follicles were more heterogeneous. Examining the differentially-expressed genes the most significantly affected functions in atretic follicles were cell death, organ development, tissue development and embryonic development. The overall processes influenced by transcription factor gene TP53 were predicted to be activated, whereas those of MYC were inhibited on the basis of known interactions with the genes in our dataset. The top ranked canonical pathway contained signalling molecules common to various inflammatory/fibrotic pathways such as the transforming growth factor-ß and tumour necrosis factor-α pathways. The two most significant networks also reflect this pattern of tissue remodelling/fibrosis gene expression. These networks also contain molecules which are present in the canonical pathways of hepatic fibrosis/hepatic stellate cell activation and transforming growth factor-ß signalling and were up regulated. CONCLUSIONS: Small healthy antral follicles, which have a number of growth outcomes, exhibit greater variability in gene expression, particularly in genes associated with cell division and other growth-related functions. Atresia, on the other hand, not only involves cell death but clearly is an active process similar to wound healing.


Assuntos
Atresia Folicular/genética , Perfilação da Expressão Gênica , Células da Granulosa/metabolismo , Folículo Ovariano/metabolismo , Animais , Apoptose , Bovinos , Análise por Conglomerados , Feminino , Atresia Folicular/metabolismo , Regulação da Expressão Gênica , Células da Granulosa/citologia , Folículo Ovariano/crescimento & desenvolvimento , Fenótipo , Análise de Componente Principal , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
7.
FASEB J ; 25(7): 2256-65, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21411746

RESUMO

Although not often discussed, the ovaries of women with polycystic ovary syndrome (PCOS) show all the hallmarks of increased TGF-ß activity, with increased amounts of fibrous tissue and collagen in the ovarian capsule or tunica albuginea and ovarian stroma. Recent studies suggest that PCOS could have fetal origins. Genetic studies of PCOS have also found linkage with a microsatellite located in intron 55 of the extracellular matrix protein fibrillin 3. Fibrillins regulate TGF-ß bioactivity in tissues by binding latent TGF-ß binding proteins. We therefore examined expression of fibrillins 1-3, latent TGF-ß binding proteins 1-4, and TGF-ß 1-3 in bovine and human fetal ovaries at different stages of gestation and in adult ovaries. We also immunolocalized fibrillins 1 and 3. The results indicate that TGF-ß pathways operate during ovarian fetal development, but most important, we show fibrillin 3 is present in the stromal compartments of fetal ovaries and is highly expressed at a critical stage early in developing human and bovine fetal ovaries when stroma is expanding and follicles are forming. These changes in expression of fibrillin 3 in the fetal ovary could lead to a predisposition to develop PCOS in later life.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Ovário/metabolismo , Síndrome do Ovário Policístico/genética , Fator de Crescimento Transformador beta/genética , Animais , Bovinos , Feminino , Fibrilinas , Humanos , Imuno-Histoquímica , Proteínas de Ligação a TGF-beta Latente/genética , Proteínas de Ligação a TGF-beta Latente/metabolismo , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Folículo Ovariano/embriologia , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/metabolismo , Ovário/embriologia , Ovário/crescimento & desenvolvimento , Síndrome do Ovário Policístico/metabolismo , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta2/genética , Fator de Crescimento Transformador beta2/metabolismo , Fator de Crescimento Transformador beta3/genética , Fator de Crescimento Transformador beta3/metabolismo
8.
PLoS One ; 17(7): e0268467, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35802560

RESUMO

During ovarian development, gonadal ridge epithelial-like (GREL) cells arise from the epithelial cells of the ventral surface of the mesonephros. They ultimately develop into follicular granulosa cells or into ovarian surface epithelial cells. Stromal fibroblasts arise from the mesonephros and penetrate the ovary. We developed methods for isolating and culturing fetal ovarian GREL cells and ovarian fibroblasts by expansion of colonies without passage. In culture, these two cell types were morphologically different. We examined the expression profile of 34 genes by qRT-PCR, of which 24 genes had previously been studied in whole fetal ovaries. Expression of nine of the 10 newly-examined genes in fetal ovaries correlated with gestational age (MUC1, PKP2, CCNE1 and CCNE2 negatively; STAR, COL4A1, GJA1, LAMB2 and HSD17B1 positively). Comparison between GREL cells and fetal fibroblasts revealed higher expression of KRT19, PKP2, OCLN, MUC1, ESR1 and LGR5 and lower expression of GJA1, FOXL2, NR2F2, FBN1, COL1A1, NR5A1, CCND2, CCNE1 and ALDH1A1. Expression of CCND2, CCNE1, CCNE2, ESR2 and TGFBR1 was higher in the fetal fibroblasts than in adult fibroblasts; FBN1 was lower. Expression of OCLN, MUC1, LAMB2, NR5A1, ESR1, ESR2, and TGFBR3 was lower in GREL cells than ovarian surface epithelial cells. Expression of KRT19, DSG2, PKP2, OCLN, MUC1, FBN1, COL1A1, COL3A1, STAR and TGFBR2 was higher and GJA1, CTNNB1, LAMB2, NR5A1, CYP11A1, HSD3B1, CYP19A1, HSD17B1, FOXL2, ESR1, ESR2, TGFBR3 and CCND2 was lower in GREL cells compared to granulosa cells. TGFß1 altered the expression of COL1A1, COL3A1 and FBN1 in fetal fibroblasts and epidermal growth factor altered the expression of FBN1 and COL1A1. In summary, the two major somatic cell types of the developing ovary have distinct gene expression profiles. They, especially GREL cells, also differ from the cells they ultimately differentiate in to. The regulation of cell fate determination, particularly of the bi-potential GREL cells, remains to be elucidated.


Assuntos
Células da Granulosa , Mesonefro , Animais , Bovinos , Células Epiteliais , Feminino , Fibroblastos/metabolismo , Células da Granulosa/metabolismo , Ovário/metabolismo
9.
Reprod Fertil ; 2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-36346793

RESUMO

Polycystic ovary syndrome (PCOS) is an endocrine metabolic disorder that appears to have a genetic predisposition and a fetal origin. The fetal ovary has two major somatic cell types shown previously to be of different cellular origins, different morphologies and to differentially express 15 genes. We isolated the somatic gonadal ridge epithelial-like (GREL) cells (n = 7) and ovarian fetal fibroblasts (n = 6) by clonal expansion. Using qRT-PCR, we compared the gene expression levels of PCOS candidate genes with previous data on the expression levels in whole fetal ovaries across gestation. We also compared these levels with those in bovine adult ovarian cells including fibroblasts (n = 4), granulosa cells (n = 5) and surface epithelial cells (n = 5). Adult cell types exhibited clear differences in the expression of most genes. In fetal ovarian cells, DENND1A and ERBB3 had significantly higher expression in GREL cells. HMGA2 and TGFB1I1 tended to have higher expression in fetal fibroblasts than GREL cells. Another 19 genes did not exhibit differences between GREL cells and fetal fibroblasts and FBN3, FSHB, LHCGR, FSHR and ZBTB16 were very lowly expressed in GREL cells and fibroblasts. The culture of fetal fibroblasts in EGF-containing medium resulted in lower expression of NEIL2, but higher expression of MAPRE1 compared to culture in the absence of EGF. Thus, the two fetal ovarian somatic cell types mostly lacked differential expression of PCOS candidate genes.

10.
Biol Reprod ; 82(6): 1021-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20164441

RESUMO

The formation of the follicular antrum and follicular fluid has received scant attention from researchers, yet both are important processes in follicular development. The central hypothesis on follicular fluid formation suggests that production by granulosa cells of hyaluronan and the chondroitin sulfate proteoglycan versican generates an osmotic gradient. This gradient draws in fluid derived from the thecal vasculature. Inter-alpha-trypsin inhibitor is also present in follicular fluid at least in species with large follicles, and inter-alpha-trypsin inhibitor and versican could additionally bind or cross-link with hyaluronan, resulting in the retention of these molecules within the follicular antrum. Barriers to the movement of fluid across the membrana granulosa are apparently minimal, as even relatively large serum proteins are present in follicular fluid. Despite the relative permeability of the follicular wall, aquaporins are present in granulosa cells and could be actively involved in the transport of water into the follicle. The formation of an antrum also requires movement of granulosa cells relative to each other to allow the fluid to accumulate. This presumably involves remodeling of cell-cell junctions and in species with small follicles may involve death of centrally located granulosa cells. Remodeling of the stroma and thecal layers also accompanies growth and expansion of the antrum and presumably involves similar processes that accompany growth of other glands.


Assuntos
Líquido Folicular/fisiologia , Células da Granulosa/fisiologia , Folículo Ovariano/fisiologia , alfa-Globulinas/metabolismo , Animais , Aquaporinas/análise , Bovinos , Movimento Celular , Proteoglicanas de Sulfatos de Condroitina/biossíntese , Cricetinae , Feminino , Células da Granulosa/metabolismo , Humanos , Ácido Hialurônico/biossíntese , Camundongos , Oócitos/metabolismo , Osmose/fisiologia , Ratos , Versicanas/biossíntese
11.
Cell Tissue Res ; 339(3): 613-24, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20033213

RESUMO

Despite the mouse being an important laboratory species, little is known about changes in its extracellular matrix (ECM) during follicle and corpora lutea formation and regression. Follicle development was induced in mice (29 days of age/experimental day 0) by injections of pregnant mare's serum gonadotrophin on days 0 and 1 and ovulation was induced by injection of human chorionic gonadotrophin on day 2. Ovaries were collected for immunohistochemistry (n=10 per group) on days 0, 2 and 5. Another group was mated and ovaries were examined on day 11 (n=7). Collagen type IV alpha1 and alpha2, laminin alpha1, beta1 and gamma1 chains, nidogens 1 and 2 and perlecan were present in the follicular basal lamina of all developmental stages. Collagen type XVIII was only found in basal lamina of primordial, primary and some preantral follicles, whereas laminin alpha2 was only detected in some preantral and antral follicles. The focimatrix, a specialised matrix of the membrana granulosa, contained collagen type IV alpha1 and alpha2, laminin alpha1, beta1 and gamma1 chains, nidogens 1 and 2, perlecan and collagen type XVIII. In the corpora lutea, staining was restricted to capillary sub-endothelial basal laminas containing collagen type IV alpha1 and alpha2, laminin alpha1, beta1 and gamma1 chains, nidogens 1 and 2, perlecan and collagen type XVIII. Laminins alpha4 and alpha5 were not immunolocalised to any structure in the mouse ovary. The ECM composition of the mouse ovary has similarities to, but also major differences from, other species with respect to nidogens 1 and 2 and perlecan.


Assuntos
Corpo Lúteo/metabolismo , Matriz Extracelular/metabolismo , Folículo Ovariano/metabolismo , Animais , Corpo Lúteo/citologia , Feminino , Humanos , Imuno-Histoquímica , Laminina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Folículo Ovariano/citologia , Gravidez
12.
J Histochem Cytochem ; 68(2): 113-126, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31855103

RESUMO

When first formed, the ovary only has an established epithelium at its base or hilum. Later, an epithelium is established around the rest of the ovary. To examine this further, we conducted scanning electron microscopy of the surface of bovine fetal ovaries and immunohistochemistry of ovarian cross-sections. From the earliest time point, the cells on the surface of the base or hilum of the ovary were cuboidal. On the remainder of the ovary, the surface was more irregular. By mid-development, the surface was covered completely with either a stratified or simple epithelium of cuboidal cells. Clefts were observed in the surface and appeared to form due to the expansion of stroma surrounding each open ovigerous cord, elevating the areas surrounding each cord, while leaving the opening of the cord to form the base of each cleft. The continued expansion of the surrounding stroma below the surface appeared not only to close the ovigerous cords from the surface but to compress the clefts into the shape of a groove. Later, most of the ovarian surface was covered with a simple cuboidal epithelium. The changes to the ovarian surface during fetal development coincide with the remodeling of the stroma and cords below.


Assuntos
Epitélio/metabolismo , Desenvolvimento Fetal , Ovário/citologia , Animais , Bovinos , Feminino , Imuno-Histoquímica , Ovário/metabolismo
13.
PLoS One ; 15(2): e0229351, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32078641

RESUMO

Polycystic ovary syndrome (PCOS) affects around 10% of young women, with adverse consequences on fertility and cardiometabolic outcomes. PCOS appears to result from a genetic predisposition interacting with developmental events during fetal or perinatal life. We hypothesised that PCOS candidate genes might be expressed in the fetal ovary when the stroma develops; mechanistically linking the genetics, fetal origins and adult ovarian phenotype of PCOS. In bovine fetal ovaries (n = 37) of 18 PCOS candidate genes only SUMO1P1 was not expressed. Three patterns of expression were observed: early gestation (FBN3, GATA4, HMGA2, TOX3, DENND1A, LHCGR and FSHB), late gestation (INSR, FSHR, and LHCGR) and throughout gestation (THADA, ERBB4, RAD50, C8H9orf3, YAP1, RAB5B, SUOX and KRR1). A splice variant of FSHB exon 3 was also detected early in the bovine ovaries, but exon 2 was not detected. Three other genes, likely to be related to the PCOS aetiology (AMH, AR and TGFB1I1), were also expressed late in gestation. Significantly within each of the three gene groups, the mRNA levels of many genes were highly correlated with each other, despite, in some instances, being expressed in different cell types. TGFß is a well-known stimulator of stromal cell replication and collagen synthesis and TGFß treatment of cultured fetal ovarian stromal cells inhibited the expression of INSR, AR, C8H9orf3 and RAD50 and stimulated the expression of TGFB1I1. In human ovaries (n = 15, < 150 days gestation) many of the same genes as in bovine (FBN3, GATA4, HMGA2, FSHR, DENND1A and LHCGR but not TOX3 or FSHB) were expressed and correlated with each other. With so many relationships between PCOS candidate genes during development of the fetal ovary, including TGFß and androgen signalling, we suggest that future studies should determine if perturbations of these genes in the fetal ovary can lead to PCOS in later life.


Assuntos
Biomarcadores/análise , Desenvolvimento Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Ovário/patologia , Síndrome do Ovário Policístico/patologia , Polimorfismo de Nucleotídeo Único , Adulto , Animais , Bovinos , Feminino , Genes Reguladores , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Ovário/metabolismo , Síndrome do Ovário Policístico/genética , Gravidez , Células Estromais/metabolismo , Células Estromais/patologia
14.
Mol Hum Reprod ; 15(12): 829-41, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19692420

RESUMO

Several studies have demonstrated an association between polycystic ovary syndrome (PCOS) and the dinucleotide repeat microsatellite marker D19S884, which is located in intron 55 of the fibrillin-3 (FBN3) gene. Fibrillins, including FBN1 and 2, interact with latent transforming growth factor (TGF)-beta-binding proteins (LTBP) and thereby control the bioactivity of TGFbetas. TGFbetas stimulate fibroblast replication and collagen production. The PCOS ovarian phenotype includes increased stromal collagen and expansion of the ovarian cortex, features feasibly influenced by abnormal fibrillin expression. To examine a possible role of fibrillins in PCOS, particularly FBN3, we undertook tagging and functional single nucleotide polymorphism (SNP) analysis (32 SNPs including 10 that generate non-synonymous amino acid changes) using DNA from 173 PCOS patients and 194 controls. No SNP showed a significant association with PCOS and alleles of most SNPs showed almost identical population frequencies between PCOS and control subjects. No significant differences were observed for microsatellite D19S884. In human PCO stroma/cortex (n = 4) and non-PCO ovarian stroma (n = 9), follicles (n = 3) and corpora lutea (n = 3) and in human ovarian cancer cell lines (KGN, SKOV-3, OVCAR-3, OVCAR-5), FBN1 mRNA levels were approximately 100 times greater than FBN2 and 200-1000-fold greater than FBN3. Expression of LTBP-1 mRNA was 3-fold greater than LTBP-2. We conclude that FBN3 appears to have little involvement in PCOS but cannot rule out that other markers in the region of chromosome 19p13.2 are associated with PCOS or that FBN3 expression occurs in other organs and that this may be influencing the PCOS phenotype.


Assuntos
Expressão Gênica , Proteínas dos Microfilamentos , Ovário/fisiologia , Síndrome do Ovário Policístico/genética , Isoformas de Proteínas , Linhagem Celular Tumoral , Células Cultivadas , Mapeamento Cromossômico , Cromossomos Humanos Par 19 , Feminino , Fibrilinas , Predisposição Genética para Doença , Genótipo , Haplótipos , Humanos , Masculino , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Repetições de Microssatélites , Dados de Sequência Molecular , Fenótipo , Síndrome do Ovário Policístico/metabolismo , Polimorfismo de Nucleotídeo Único , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo
15.
Hum Reprod ; 24(4): 936-44, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19095662

RESUMO

BACKGROUND: The ovarian follicular basal lamina underlies the epithelial membrana granulosa and maintains the avascular intra-follicular compartment. Additional layers of basal lamina occur in a number of pathologies, including pili annulati and diabetes. We previously found additional layers of follicular basal lamina in a significant percentage of healthy bovine follicles. We wished to determine if this phenomenon existed in humans, and if it was related to oocyte function in the bovine. METHODS AND RESULTS: We examined follicles from human ovaries (n = 18) by electron microscopy and found that many follicles had additional layers of basal lamina. Oocytes (n = 222) from bovine follicles with normal or unusual basal laminas were isolated and their ability to undergo in vitro maturation, fertilization and culture to blastocyst was compared. Healthy bovine follicles with a single layer of basal lamina had oocytes with significantly (P < 0.01) greater developmental competence than healthy follicles with additional layers of follicular basal lamina (65% versus 28%). CONCLUSIONS: These findings provide direct evidence that the phenotype of the follicular basal lamina is related to oocyte competence.


Assuntos
Oócitos/crescimento & desenvolvimento , Folículo Ovariano/ultraestrutura , Animais , Membrana Basal/ultraestrutura , Blastocisto/citologia , Bovinos , Contagem de Células , Feminino , Humanos , Técnicas In Vitro , Microscopia Eletrônica de Transmissão , Recuperação de Oócitos , Fenótipo , Técnicas de Reprodução Assistida , Especificidade da Espécie
16.
Med Hypotheses ; 124: 31-34, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30798911

RESUMO

Polycystic ovary syndrome (PCOS) affects around 10% of women of reproductive age and is most common in developed countries. The aetiology of PCOS is not completely understood. Current evidence suggests that the syndrome results from a genetic predisposition interacting with developmental events during fetal or perinatal life that together increase susceptibility in some individuals. This implies that environmental factors influence the initiation of PCOS in the fetus or infant, either directly or via the mother. PCOS is often considered to be an ancient disorder but there is no direct proof of this in the medical or historic record. One of the cardinal features, polycystic ovaries, was first described only in the early 1900s, despite reports of many thousands of autopsies recorded earlier. This conundrum could be explained by postulating that polycystic ovaries were rare before the 1900s and have become more common over the last 100 years. The hypothesis that PCOS is a syndrome of the 20th Century would eliminate the need to explain the paradox of why there exists a genetic predisposition to subfertility syndrome.


Assuntos
Síndrome do Ovário Policístico/epidemiologia , Síndrome do Ovário Policístico/história , Adulto , Androgênios , Comorbidade , Meio Ambiente , Feminino , Predisposição Genética para Doença , História do Século XX , História do Século XXI , Humanos , Infertilidade Feminina , Modelos Teóricos , Ovário/patologia , Síndrome do Ovário Policístico/etiologia , Prevalência
17.
PLoS One ; 14(3): e0213575, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30856218

RESUMO

Movement and expansion of mesonephric-derived stroma appears to be very important in the development of the ovary. Here, we examined the expression of 24 genes associated with stroma in fetal ovaries during gestation (n = 17; days 58-274) from Bos taurus cattle. RNA was isolated from ovaries for quantitative RT-PCR. Expression of the majority of genes in TGFß signalling, stromal transcription factors (NR2F2, AR), and some stromal matrix genes (COL1A1, COL3A1 and FBN1, but not FBN3) showed a positive linear increase with gestational age. Expression of genes associated with follicles (INSL3, CYP17A1, CYP11A1 and HSD3B1), was low until mid-gestation and then increased with gestational age. LHCGR showed an unusual bimodal pattern; high levels in the first and last trimesters. RARRES1 and IGFBP3 also increased with gestational age. To relate changes in gene expression in stromal cells with that in non stromal cells during development of the ovary we combined the data on the stromal genes with another 20 genes from non stromal cells published previously and then performed hierarchical clustering analysis. Three major clusters were identified. Cluster 1 genes (GATA4, FBN3, LHCGR, CYP19A1, ESR2, OCT4, DSG2, TGFB1, CCND2, LGR5, NR5A1) were characterised by high expression only in the first trimester. Cluster 2 genes (FSHR, INSL3, HSD3B1, CYP11A1, CYP17A1, AMH, IGFBP3, INHBA) were highly expressed in the third trimester and largely associated with follicle function. Cluster 3 (COL1A1, COL3A1, FBN1, TGFB2 TGFB3, TGFBR2, TGFBR3, LTBP2, LTBP3, LTBP4, TGFB1I1, ALDH1A1, AR, ESR1, NR2F2) had much low expression in the first trimester rising in the second trimester and remaining at that level during the third trimester. Cluster 3 contained members of two pathways, androgen and TGFß signalling, including a common member of both pathways namely the androgen receptor cofactor TGFß1 induced transcript 1 protein (TGFB1I1; hic5). GATA4, FBN3 and LHCGR, were highly correlated with each other and were expressed highly in the first trimester during stromal expansion before follicle formation, suggesting that this could be a critical phase in the development of the ovarian stroma.


Assuntos
Bovinos/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Ovário/embriologia , Animais , Bovinos/genética , Bovinos/metabolismo , Feminino , Redes Reguladoras de Genes , Idade Gestacional , Família Multigênica , Ovário/citologia , Ovário/metabolismo , Gravidez , Transdução de Sinais , Células Tecais/citologia , Células Tecais/metabolismo , Transcriptoma , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
18.
PLoS One ; 14(3): e0214130, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30901367

RESUMO

Cells on the surface of the mesonephros give rise to replicating Gonadal Ridge Epithelial-Like (GREL) cells, the first somatic cells of the gonadal ridge. Later germ cells associate with the GREL cells in the ovigerous cords, and the GREL cells subsequently give rise to the granulosa cells in follicles. To examine these events further, 27 bovine fetal ovaries of different gestational ages were collected and prepared for immunohistochemical localisation of collagen type I and Ki67 to identify regions of the ovary and cell proliferation, respectively. The non-stromal cortical areas (collagen-negative) containing GREL cells and germ cells and later in development, the follicles with oocytes and granulosa cells, were analysed morphometrically. Another set of ovaries (n = 17) were collected and the expression of genes associated with germ cell lineages and GREL/granulosa cells were quantitated by RT-PCR. The total volume of non-stromal areas in the cortex increased significantly and progressively with ovarian development, plateauing at the time the surface epithelium developed. However, the proportion of non-stromal areas in the cortex declined significantly and progressively throughout gestation, largely due to a cessation in growth of the non-stroma cells and the continued growth of stroma. The proliferation index in the non-stromal area was very high initially and then declined substantially at the time follicles formed. Thereafter, it remained low. The numerical density of the non-stromal cells was relatively constant throughout ovarian development. The expression levels of a number of genes across gestation either increased (AMH, FSHR, ESR1, INHBA), declined (CYP19A1, ESR2, ALDH1A1, DSG2, OCT4, LGR5) or showed no particular pattern (CCND2, CTNNB1, DAZL, FOXL2, GATA4, IGFBP3, KRT19, NR5A1, RARRES1, VASA, WNT2B). Many of the genes whose expression changed across gestation, were positively or negatively correlated with each other. The relationships between these genes may reflect their roles in the important events such as the transition of ovigerous cords to follicles, oogonia to oocytes or GREL cells to granulosa cells.


Assuntos
Bovinos/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Ovário/embriologia , Animais , Bovinos/genética , Feminino , Células Germinativas/citologia , Células Germinativas/metabolismo , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Mesonefro/citologia , Mesonefro/embriologia , Mesonefro/metabolismo , Ovário/citologia , Ovário/metabolismo
19.
Reprod Fertil Dev ; 19(3): 473-81, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17394796

RESUMO

Immunohistology has been used to examine the distribution of certain components of the basement membrane (BM) associated with bovine spermatogonial germ cells that are located within the seminiferous tubules. Histology was performed on testis tissue from Brahman cattle (Bos indicus) of three different age groups: pre-pubescent (4-6 months), juvenile (8-10 months) and adult (18-24 months) animals. There were no major changes in the BM composition apparent between these three age groups, except for certain lectin staining. These data suggest that the predominant collagen type IV component may have an alpha3 and alpha4 composition, although other chains, including the alpha5 and alpha6 chains, were also present. Possibly the main laminin type present was laminin 121 (alpha1beta2gamma1), although other variants were also present. Both nidogen-1 and perlecan, which are normal BM components, were also found as part of the seminiferous tubule BM. Interstitial collagens, such as type I, III and VI collagens, were found in the peritubular space, but were not part of the BM itself, although type VI collagen was most visible in the peritubular zone adjacent to the tubules. Examination of the BM with a range of lectins gave strong staining for (glcNAc)(2) entities, weak positive staining for alpha-l-fuc, but little or no staining for alpha-galNAc and (glcNAc)(3) at all ages, whereas staining for alpha-gal, beta-gal(1-->3)galNAc and alpha-man showed developmental changes.


Assuntos
Membrana Basal/química , Bovinos , Colágenos Fibrilares/análise , Laminina/análise , Glicoproteínas de Membrana/análise , Túbulos Seminíferos/química , Animais , Membrana Basal/citologia , Imuno-Histoquímica , Masculino , Túbulos Seminíferos/citologia
20.
PLoS One ; 12(3): e0173391, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28282394

RESUMO

In vitro culture of ovarian granulosa cells and theca cells has been very important for our understanding of their function and regulation. One of the most eagerly sought attributes of cell culture is the use of chemically-defined conditions. However, even under such in vitro conditions cell behaviour could differ from the in vivo situation because of differences in oxygen tension, nutrients, adhesion matrix and other factors. To examine this further we compared the transcriptomes of both granulosa cells and cells from the theca interna that were cultured in what are arguably the best in vitro conditions for maintaining the 'follicular' phenotypes of both tissue types, as displayed by their respective freshly-isolated counterparts. The array data analysed are from recently published data and use the same sizes of bovine follicles (small antral 3-6 mm) and the same Affymetrix arrays. We conducted analysis using Partek, Ingenuity Pathway Analysis and GOEAST. Principal Component Analysis (PCA) and hierarchical clustering clearly separated the in vivo from the in vitro groups for both cells types and transcriptomes were more homogeneous upon culture. In both cell cultures behaviours associated with cell adhesion, migration and interaction with matrix or substrate were more abundant. However, the pathways involved generally differed between the two cell types. With the thecal cultures a gene expression signature of an immune response was more abundant, probably by leukocytes amongst the cells cultured from the theca interna. These results indicate differences between in vivo and in vitro that should be considered when interpreting in vitro data.


Assuntos
Células da Granulosa/metabolismo , Células Tecais/metabolismo , Transcriptoma , Animais , Bovinos , Células Cultivadas , Análise por Conglomerados , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Células da Granulosa/citologia , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , Análise de Componente Principal , RNA/isolamento & purificação , RNA/metabolismo , Células Tecais/citologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa