Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 132024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38771186

RESUMO

Current studies on cultured meat mainly focus on the muscle tissue reconstruction in vitro, but lack the formation of intramuscular fat, which is a crucial factor in determining taste, texture, and nutritional contents. Therefore, incorporating fat into cultured meat is of superior value. In this study, we employed the myogenic/lipogenic transdifferentiation of chicken fibroblasts in 3D to produce muscle mass and deposit fat into the same cells without the co-culture or mixture of different cells or fat substances. The immortalized chicken embryonic fibroblasts were implanted into the hydrogel scaffold, and the cell proliferation and myogenic transdifferentiation were conducted in 3D to produce the whole-cut meat mimics. Compared to 2D, cells grown in 3D matrix showed elevated myogenesis and collagen production. We further induced fat deposition in the transdifferentiated muscle cells and the triglyceride content could be manipulated to match and exceed the levels of chicken meat. The gene expression analysis indicated that both lineage-specific and multifunctional signalings could contribute to the generation of muscle/fat matrix. Overall, we were able to precisely modulate muscle, fat, and extracellular matrix contents according to balanced or specialized meat preferences. These findings provide new avenues for customized cultured meat production with desired intramuscular fat contents that can be tailored to meet the diverse demands of consumers.


Assuntos
Transdiferenciação Celular , Galinhas , Fibroblastos , Carne , Animais , Fibroblastos/metabolismo , Fibroblastos/citologia , Tecido Adiposo/citologia , Células Musculares/citologia , Desenvolvimento Muscular , Proliferação de Células , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Carne in vitro
2.
Reproduction ; 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38670156

RESUMO

Oogonial stem cells (OSCs) are a type of germ stem cell present in the adult ovary. They have the ability to self-renew through mitosis and differentiate into oocytes through meiosis. We have previously identified a population of OSCs in chicken ovary, but the underlying mechanism control their activation and proliferation were unclear. In this study, we observed that OSCs showed robust proliferation when cultured on a layer of chicken embryo fibroblasts (CEF), suggesting that CEF may secrete certain crucial factors that activate OSC proliferation. We further detected Transforming Growth Factor beta 1 (TGF-ß1) as a potent signaling molecule to promote OSC proliferation. Additionally, we revealed the signaling pathways that play important roles in the downstream of TGF-ß1-induced OSC proliferation. These findings provide insights into the mechanisms underlying OSC proliferation in chickens and offer a foundation for future research on in situ activation of OSC proliferation in ovary and improvement of egg-laying performance in chickens.

3.
PNAS Nexus ; 2(6): pgad202, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37388922

RESUMO

All female vertebrates develop a pair of ovaries except for birds, in which only the left gonad develops into an ovary, whereas the right gonad regresses. Previous studies found that the transcription factor Paired-Like Homeodomain 2 (PITX2), a key mediator for left/right morphogenesis in vertebrates, was also implicated in asymmetric gonadal development in chickens. In this study, we systematically screened and validated the signaling pathways that could be targeted by Pitx2 to control unilateral gonad development. Integrated chromatin immunoprecipitation sequencing (ChIP-seq) and RNA sequencing (RNA-seq) analyses indicated that Pitx2 directly binds to the promoters of genes encoding neurotransmitter receptors and leads to left-biased expression of both serotonin and dopamine receptors. Forcibly activating serotonin receptor 5-Hydroxytryptamine Receptor 1B (HTR1B) signaling could induce ovarian gene expression and cell proliferation to partially rescue the degeneration of the right gonad. In contrast, inhibiting serotonin signaling could block the development of the left gonad. These findings reveal a PITX2-HTR1B genetic pathway that guides the left-specific ovarian growth in chickens. We also provided new evidence showing neurotransmitters stimulate the growth of nonneuronal cells during the early development of reproductive organs well before innervation.

4.
Genes (Basel) ; 14(4)2023 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-37107539

RESUMO

Layer and broiler hens show a dramatic difference in the volume and frequency of egg production. However, it is unclear whether the intrinsic competency of oocyte generation is also different between the two types of chicken. All oocytes were derived from the primordial germ cells (PGC) in the developing embryo, and female PGC proliferation (mitosis) and the subsequent differentiation (meiosis) determine the ultimate ovarian pool of germ cells available for future ovulation. In this study, we systematically compared the cellular phenotype and gene expression patterns during PGC mitosis (embryonic day 10, E10) and meiosis (E14) between female layers and broilers to determine whether the early germ cell development is also subjected to the selective breeding of egg production traits. We found that PGCs from E10 showed much higher activity in cell propagation and were enriched in cell proliferation signaling pathways than PGCs from E14 in both types of chicken. A common set of genes, namely insulin-like growth factor 2 (IGF2) and E2F transcription factor 4 (E2F4), were identified as the major regulators of cell proliferation in E10 PGCs of both strains. In addition, we found that E14 PGCs from both strains showed an equal ability to initiate meiosis, which was associated with the upregulation of key genes for meiotic initiation. The intrinsic cellular dynamics during the transition from proliferation to differentiation of female germ cells were conserved between layers and broilers. Hence, we surmise that other non-cell autonomous mechanisms involved in germ-somatic cell interactions would contribute to the divergence of egg production performance between layers and broilers.


Assuntos
Galinhas , Células Germinativas , Animais , Feminino , Galinhas/genética , Células Germinativas/metabolismo , Meiose/genética , Oócitos , Mitose/genética
5.
Cell Prolif ; 56(3): e13371, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36526415

RESUMO

OBJECTIVES: Oogonial stem cells (OSCs) are germ cells that can sustain neo-oogenesis to replenish the pool of primary follicles in adult ovaries. In lower vertebrates, fresh oocytes are produced by numerous OSCs through mitosis and meiosis during each reproduction cycle, but the OSCs in adult mammals are rare. The birds have retained many conserved features and developed unique features of ovarian physiology during evolution, and the presence of OSCs within avian species remain unknown. MATERIALS AND METHODS: In this study, we investigated the existence and function of OSCs in adult chickens. The chicken OSCs were isolated and expanded in culture. We then used cell transplantation system to evaluate their potential for migration and differentiation in vivo. RESULTS: DDX4/SSEA1-positive OSCs were identified in both the cortex and medulla of the adult chicken ovary. These putative OSCs undergo meiosis in the reproductively active ovary. Furthermore, the isolated OSCs were expanded in vitro for months and found to express germline markers similar to those of primordial germ cells. When transplanted into the bloodstream of recipient embryos, these OSCs efficiently migrated into developing gonads, initiated meiosis, and then derived oocytes in postnatal ovaries. CONCLUSIONS: This study has confirmed the presence of functional OSCs in birds for the first time. The identification of chicken OSCs has great potential for improving egg laying and preserving endangered species.


Assuntos
Células-Tronco de Oogônios , Ovário , Feminino , Animais , Galinhas , Células-Tronco de Oogônios/fisiologia , Oócitos , Oogênese , Mamíferos
6.
Stem Cell Rev Rep ; 18(7): 2535-2546, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35397052

RESUMO

Primordial germ cells (PGCs) are the undifferentiated progenitors of the gametes. Unlike the poor maintenance of cultured mammalian PGCs, the avian PGCs can be expanded in vitro indefinitely while preserving pluripotency and germline competence. In mammals, the Oct4 is the master transcription factor that ensures the stemness of pluripotent cells such as PGCs, but the specific function of Oct4 in chicken PGCs remains unclear. As expected, the loss of Oct4 in chicken PGCs reduced the expression of key pluripotency factors and promoted the genes involved in endoderm and ectoderm differentiation. Furthermore, the global active chromatin was reduced as shown by the depletion of the H3K27ac upon Oct4 suppression. Interestingly, the de-activated chromatin caused the down-regulation of adjacent genes which are mostly known regulators of cell junction, chemotaxis and cell migration. Consequently, the Oct4-deficient PGCs show impaired cell migration and could not colonize the gonads when re-introduced into the bloodstream of the embryo. We propose that, in addition to maintaining pluripotency, the Oct4 mediated chromatin activation is dictating chicken PGC migration.


Assuntos
Galinhas , Cromatina , Animais , Movimento Celular/genética , Galinhas/genética , Galinhas/metabolismo , Cromatina/metabolismo , Células Germinativas , Mamíferos/genética , Mamíferos/metabolismo , Fatores de Transcrição/metabolismo
7.
Mol Reprod Dev ; 89(4): 214-226, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35118723

RESUMO

Primordial germ cells (PGCs), precursors to sperms and oocytes, are responsible for the transfer of genetic information to the next generation. The PGCs arise far away from the developing gonad and thus have to migrate across the embryo to reach their site of function. The migration of PGCs from extraembryonic regions to the genital ridges is accomplished through distinct routes among different species. In particular, the birds PGCs utilized the developing circulation system to travel long distance before settling within the gonad. This study screened the transcriptome profile of chicken PGCs isolated from the bloodstream and the genital ridges to identify the cell intrinsic signals that could guide the unique migration path through circulation. We found cell adhesion and extracellular matrix (ECM) associated pathways were highly enriched in the PGCs from blood but not gonads. The platelet-derived growth factor receptors (PDGFRA and PDGFRB) were downregulated during gonad colonization and knockdown of either PDGFRA or PDGFRB inhibit the proliferation of blood PGCs. Furthermore, the migration of blood PGCs was impaired by the suppression of PDGFRA but not PDGFRB. Hence, the chicken PGCs show dynamic transcriptional remodeling during the blood-to-gonad migration and colonization. The free-floating PGCs in the circulation already express genes associated with cell-cell and cell-ECM interactions and therefore prepare for gonadal colonization.


Assuntos
Galinhas , Receptor beta de Fator de Crescimento Derivado de Plaquetas , Animais , Adesão Celular/genética , Movimento Celular/genética , Proliferação de Células , Galinhas/genética , Células Germinativas , Gônadas , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo
8.
Biol Sex Differ ; 13(1): 6, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35135592

RESUMO

BACKGROUND: Birds exhibit a unique asymmetry in terms of gonad development. The female left gonad generates a functional ovary, whereas the right gonad regresses. In males, both left and right gonads would develop into testes. How is this left/right asymmetry established only in females but not in males remains unknown. The epigenetic regulation of gonadal developmental genes may contribute to this sex disparity. The modification of histone tails such as H3K27ac is tightly coupled to chromatin activation and gene expression. To explore whether H3K27ac marked chromatin activation is involved in the asymmetric development of avian gonads, we probed genome-wide H3K27ac occupancy in left and right gonads from both sexes and related chromatin activity profile to the expression of gonadal genes. Furthermore, we validated the effect of chromatin activity on asymmetric gonadal development by manipulating the chromatin histone acetylation levels. METHODS: The undifferentiated gonads from both sides of each sex were collected and subjected to RNA-Seq and H3K27ac ChIP-Seq experiments. Integrated analysis of gene expression and active chromatin regions were performed to identify the sex- and situs-specific regulation and expression of gonadal genes. The histone deacetylase inhibitor trichostatin A (TSA) was applied to the undifferentiated female right gonads to assess the effect of chromatin activation on gonadal gene expression and cell proliferation. RESULTS: Even before sex differentiation, the gonads already show divergent gene expression between different sexes and between left/right sides in females. The sex-specific H3K27ac chromatin distributions coincide with the higher expression of male/female specification genes in each sex. Unexpectedly, the H3K27ac marked chromatin activation show a dramatic difference between left and right gonads in both sexes, although the left/right asymmetric gonadal development was observed only in females but not in males. In females, the side-specific H3K27ac occupancy instructs the differential expression of developmental genes between the pair of gonads and contributes to the development of left but not right gonad. However, in males, the left/right discrepancy of H3K27ac chromatin distribution does not drive the side-biased gene expression or gonad development. The TSA-induced retention of chromatin acetylation causes up-regulation of ovarian developmental genes and increases cell proliferation in the female right gonad. CONCLUSIONS: We revealed that left/right asymmetry in H3K27ac marked chromatin activation exists in both sexes, but this discrepancy gives rise to asymmetric gonadal development only in females. Other mechanisms overriding the chromatin activation would control the symmetric development of male gonads in chicken.


Assuntos
Galinhas , Cromatina , Acetilação , Animais , Galinhas/genética , Galinhas/metabolismo , Cromatina/metabolismo , Epigênese Genética , Feminino , Expressão Gênica , Gônadas/metabolismo , Histonas/genética , Histonas/metabolismo , Masculino
9.
Poult Sci ; 98(11): 6117-6124, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31189182

RESUMO

Ovarian follicle selection is the critical step which determines the oocyte development and ovulation. In avian species, the somatic cells in the follicles decide the process of follicle selection but the precise molecular regulation is not well defined. N6-methyladenosine (m6A) is a ubiquitous reversible epigenetic RNA modification that plays an important role in the gene expression regulation and cell functions. In this study, we profiled transcriptome-wide m6A methylation in chicken follicles during follicular selection process in order to identify key factors involved in the follicle selection. The chicken follicle transcriptome was extensively methylated by m6A and a negative correlation was found between the m6A methylation enrichment and gene expression levels. Interestingly, both the m6A methylation peaks and the m6A modified transcripts increased during follicle selection, which lead to the dynamic expression of many folliculogenesis relevant genes. Functional enrichment analysis indicated that m6A modification of key factors in Wnt pathway could play a major role in regulating follicle selection. This study is the first to comprehensively characterize the m6A patterns in the chicken transcriptome, and provides deep insights into the m6A topology and relevant molecular mechanisms underlying follicle selection.


Assuntos
Adenosina/análogos & derivados , Galinhas/metabolismo , Folículo Ovariano/fisiologia , Adenosina/metabolismo , Animais , Feminino , Perfilação da Expressão Gênica , Metilação , RNA/metabolismo
10.
Biochem Biophys Res Commun ; 496(4): 1257-1262, 2018 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-29409950

RESUMO

Estrogen is the primary sex hormone responsible for the development and modulation of the female reproductive system in all vertebrates including avian species. The actions of estrogen are mediated by the estrogen receptor, which could be modulated by the selective estrogen receptor modulator tamoxifen (TAM). In this study, we administered TAM into the actively laying chicken to investigate the ovarian and hypothalamic responses to the estrogen action blockage. The laying was disrupted and the development of the pre-ovulatory hierarchical follicles was arrested. However, the TAM treatment caused an increase of estrogen level in both serum and ovary. Among the main estrogen targeted tissues, the hypothalamus showed specific dopaminergic activation as indicated by gene expression analysis. In the ovary, l-dopa, the precursor of dopamine, could stimulate the estrogen synthesis in undifferentiated follicles but not in the differentiated pre-ovulatory follicles. Thus, we established a feedback loop links ovarian estrogen production with hypothalamic l-dopa synthesis and we propose that the dopamine is involved in estrogen action to regulate the ovarian follicle development and ovulation.


Assuntos
Estrogênios/biossíntese , Hipotálamo/metabolismo , Levodopa/biossíntese , Ovário/efeitos dos fármacos , Ovário/metabolismo , Ovulação/fisiologia , Tamoxifeno/administração & dosagem , Animais , Galinhas , Relação Dose-Resposta a Droga , Feminino , Hipotálamo/efeitos dos fármacos , Ovulação/efeitos dos fármacos , Indução da Ovulação/métodos , Moduladores Seletivos de Receptor Estrogênico/administração & dosagem
11.
G3 (Bethesda) ; 6(5): 1305-12, 2016 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-26966259

RESUMO

Extracelluar matrix undergoes constant remodeling, cell-cell, and cell-matrix interactions during chicken ovarian follicle growth, which is coordinated by matrix metalloproteinases (MMPs), and their associated endogenous inhibitors (TIMPs). Transcriptome analysis revealed upregulation of MMP13 in sexually mature chicken ovaries. In this study, we found that the expression of MMP13 in chicken ovary was stably elevated from 60 d to 159 d, and was significantly higher at 159 d than at the other three developmental stages (P < 0.05). The expression of MMP13 mRNA increased from SW (small white follicles) to F5 (fifth largest follicles), then decreased to F1 (first largest follicles), and dramatically increased again in POF1 (newly postovulatory follicles) follicles (P < 0.05). The MMP13 protein was localized in stroma cells and primordial follicles of sexually immature chicken ovaries, in the theca cell layers of all sized follicles of sexually mature chicken ovaries. Furthermore, we identified a positive element (positions -1863 to -1036) controlling chicken MMP13 transcription, and, in this region, six single nucleotide polymorphisms were found and genotyped in chicken populations. In the White Recessive Rock population, hens with A(-1356)-C(-1079)/A(-1356)-C(-1079) genotype had earlier "age at first laying" than those with G(-1356)-T(-1079)/G(-1356)-T(-1079) genotype (P < 0.05), and exhibited significantly lower transcriptional activity (P < 0.01). Collectively, chicken MMP13 plays an important role in ovarian follicle growth and regression, and polymorphisms in its promoter region could be used as molecular markers for improving the trait "age at first laying" in chicken breeding.


Assuntos
Galinhas/genética , Expressão Gênica , Metaloproteinase 13 da Matriz/genética , Polimorfismo Genético , Regiões Promotoras Genéticas , Característica Quantitativa Herdável , Reprodução/genética , Animais , Galinhas/metabolismo , Ovos , Feminino , Imuno-Histoquímica , Desequilíbrio de Ligação , Metaloproteinase 13 da Matriz/metabolismo , Folículo Ovariano/metabolismo , Ovário/metabolismo , Deleção de Sequência
12.
J Poult Sci ; 53(2): 111-117, 2016 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-32908372

RESUMO

The chaperonin containing TCP-1 complex protein 1 subunit zeta (CCT6A) is the only cytosolic chaperonin in eukaryotes assisting in the folding of cytoplasmic proteins. Previous study revealed that the mRNA expression of chicken CCT6A gene was remarkably elevated in the sexually mature ovaries. However, the mechanism underlying chicken CCT6A expression changes remains largely unknown. In this study, haplotypes caused by two single nucleotide polymorphisms (SNPs) of chicken CCT6A gene promoter (g.-2215 T>C and g.-1959 T>C) were identified and their associations with egg production traits as well as effects on gene expression were analyzed. Altogether four haplotypes including A (C-2215-T-1959), B (C-2215-C-1959), C (T-2215-T-1959) and D (T-2215-C-1959) were detected in all of the five chicken populations. Diplotypes AA, AD and DD were predominant in Xinyang brown hens, among which diplotype AD was associated with higher egg number at the age of 28 weeks old (E28) (P<0.05). In addition, diplotype AD was also predominant in Xinyang brown and Hy-line brown chicken populations with high egg production; whereas in Wenchang and Shouguang chicken populations which are Chinese indigenous chicken breeds and relatively lower in egg production, diplotype AA was predominant. Compared with diplotypes AA and DD, the mRNA expression of CCT6A in diplotype AD birds is the highest in F1, F5, and POF1 follicles of Hy-line brown hens (P<0.05). These results suggest that the two SNPs in chicken CCT6A promoter region are potential DNA marker for improving egg production trait.

13.
PLoS One ; 10(12): e0146028, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26716441

RESUMO

The molecular mechanisms associated with follicle maturation and ovulation are not well defined in avian species. In this study, we used RNA-seq to study the gene expression profiles of the chicken follicles from different developmental stages (pre-hierarchical, pre-ovulatory and post-ovulatory). Transcriptomic analysis revealed a total of 1,277 and 2,310 genes were differentially expressed when follicles progressed through the pre-hierarchical to hierarchical and pre-ovulatory to post-ovulatory transitions, respectively. The differentially expressed genes (DEG) were involved in signaling pathways such as adherens junction, apoptosis and steroid biosynthesis. We further investigated the transcriptional regulation of follicular steroidogenesis by examining the follicle-specific methylation profiles of Star (steroidogenic acute regulatory protein), Cyp11a1 (cytochrome P450, family 11, subfamily a, polypeptide 1) and Hsd3b (hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 1), genes encoding the key enzymes for progesterone synthesis. The varied patterns of DNA methylation in proximal promoters of Star and Cyp11a1but not Hsd3b in different follicles could play a major role in controlling gene expression as well as follicular steroidogenic activity. Finally, the promoter-reporter analysis suggests that TGF-ß could be involved in the regulation of Hsd3b expression during ovulation. Together, current data not only provide novel insights into the molecular mechanisms of follicular physiology in chicken follicles, but also present the first evidence of epigenetic regulation of ovarian steroidogenesis in avian species.


Assuntos
Metilação de DNA/fisiologia , Folículo Ovariano/fisiologia , Ovulação/fisiologia , Progesterona/biossíntese , Regiões Promotoras Genéticas , Transcriptoma , Animais , Galinhas/metabolismo , Galinhas/fisiologia , Feminino , Regulação da Expressão Gênica/fisiologia , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/metabolismo , Ovulação/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transcriptoma/genética , Transcriptoma/fisiologia , Fator de Crescimento Transformador beta/fisiologia
14.
Anim Reprod Sci ; 161: 104-11, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26364124

RESUMO

Annexin A2 (ANXA2) is a calcium-dependent, phospholipid-binding protein found in various cells and tissues. ANXA2 plays multiple roles in regulating cellular functions and is often over-expressed in different types of tumors including ovarian cancer. Others and we previously found that ANXA2 was up-regulated in the ovaries of hens with higher laying rate, indicated that ANXA2 is involved in avian follicle development. In this study, we found that ANXA2 mRNA expression increased during chicken ovary maturation and follicle development. In the pre-ovulatory follicles, ANXA2 expression level was significantly higher in theca cells than granulosa cells. In theca cells, ANXA2 expression could be stimulated by follicle-stimulating hormone (FSH) and estrogen but not luteinizing hormone (LH) or progesterone. The core promoter regions control the basal and FSH-induced ANXA2 gene expression were identified. Forced expression of ANXA2 could induce the expression of angiogenic factors and receptors in theca cells. Furthermore, ANXA2 overexpression resulted increased vascular endothelial growth factor A (VEGFA) secretion and theca cell proliferation. Current study not only provides the first evidence of expression and regulation of ANXA2 in chicken ovary, but also suggests that ANXA2 is involved in follicular angiogenesis and contributes to successful follicle development and ovulation.


Assuntos
Anexina A2/fisiologia , Neovascularização Fisiológica/fisiologia , Folículo Ovariano/fisiologia , Animais , Galinhas/fisiologia , Estrogênios/farmacologia , Feminino , Hormônio Foliculoestimulante/farmacologia , Hormônio Luteinizante/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Folículo Ovariano/irrigação sanguínea , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/crescimento & desenvolvimento , Progesterona/farmacologia , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Células Tecais/efeitos dos fármacos , Células Tecais/fisiologia
15.
Asian-Australas J Anim Sci ; 27(11): 1526-31, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25358310

RESUMO

Matrix metalloproteinases (MMP) are key enzymes involved in cell and tissue remodeling during ovarian follicle development and ovulation. The control of MMP9 transcription in ovarian follicles occurs through a core promoter region (-2,400 to -1,700 bp). The aim of this study was to screen genetic variations in the core promoter region and examine MMP9 transcription regulation and reproduction performance. A single cytosine deletion/insertion polymorphism was found at -1954 C(+)/C(-). Genetic association analysis indicated significant correlation between the deletion genotype (C(-)) with total egg numbers at 28 weeks (p = 0.031). Furthermore, luciferase-reporter assay showed the deletion genotype (C(-)) had significantly lower promoter activity than the insertion genotype (C(+)) in primary granulosa cells (p<0.01). Therefore, the identified polymorphism could be used for marker-assisted selection to improve chicken laying performance.

16.
Biol Reprod ; 90(3): 57, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24451989

RESUMO

Matrix metalloproteinases (MMPs) are a specific class of proteolytic enzymes that play critical roles in follicular development and luteinization in mammals. However, the role of MMPs in avian ovary remains largely unknown. We found that three MMP genes (MMP1, MMP3, and MMP9) were significantly up-regulated in 23-wk-old (laying phase) chicken ovaries compared with 6-wk-old ovaries (prepubertal phase). In reproductively active chicken ovary, MMP1 expression (both mRNA and protein) remained low in prehierarchical and preovulatory follicles but increased in postovulatory follicles (POFs). Both MMP3 and MMP9 expression levels increased during follicular maturation. MMP3 reached maximal expression in the first largest follicle (F1), while MMP9 levels continued to rise in POF1 and POF2 after ovulation. Immunohistochemistry, Western blot analysis, and zymography experiments indicated that MMP1, MMP3, and MMP9 were synthesized and secreted by granulosa cells of different follicles in the chicken ovary. The mRNA expression of MMP1 and MMP3 in the granulosa cells was stimulated by follicle-stimulating hormone, luteinizing hormone, progesterone, and estrogen but not by transforming growth factor beta 1 (TGFB1). However, the mRNA of MMP9 was induced by TGFB1 but not follicle-stimulating hormone, luteinizing hormone, progesterone, or estrogen. Luciferase reporter and mutagenesis analysis indicated the AP1 and NFkappaB elements located in the promoter region from -1700 to -2400 bp were critical for both basal and TGFB1-induced MMP9 transcription. These data provide the first spatial-temporal expression analysis of MMP system in the chicken ovary.


Assuntos
Galinhas/fisiologia , Hormônios Esteroides Gonadais/farmacologia , Gonadotropinas/farmacologia , Metaloproteinase 1 da Matriz/biossíntese , Metaloproteinase 3 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Ovário/efeitos dos fármacos , Ovário/enzimologia , Fator de Crescimento Transformador beta1/farmacologia , Animais , Western Blotting , Células Cultivadas , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Imuno-Histoquímica , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 3 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Mutagênese , NF-kappa B/biossíntese , NF-kappa B/genética , Folículo Ovariano/efeitos dos fármacos , Plasmídeos/genética , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição AP-1/biossíntese , Fator de Transcrição AP-1/genética , Transfecção
17.
Gen Comp Endocrinol ; 189: 15-23, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23644154

RESUMO

CCT6A, the zeta subunit of the chaperonin containing TCP1 complex, is the only cytosolic chaperonin in eukaryotes and is estimated to assist in the folding of multiple proteins including actin, tubulin, cyclin E, myosin, transducin and the Von Hippel Lindau tumor suppressor. In this study, we examined the expression of CCT6A and progesterone receptor (PGR) mRNA in various tissues of chickens and the regulation of CCT6A and PGR mRNA in ovarian granulosa cells. Northern blot analysis revealed that CCT6A had one transcript and was highly expressed in the ovary tissues from chickens at both the sexually immature and mature stages. CCT6A mRNA expression was increased maximally from pre-hierarchy follicles to F5 follicles and subsequently declined in pre-ovulatory and post-ovulatory follicles. The expression of PGR mRNA exhibited the similar pattern to CCT6A. In granulosa cells isolated from pre-ovulatory follicles, follicle-stimulating hormone (FSH) inhibited the expression of CCT6A mRNA, whereas progesterone activated CCT6A and suppressed PGR expression in a time-dependent manner. We further investigated the regulation of CCT6A transcription by progesterone by constructing various progressive deletions and mutants and identified the core promoter element of CCT6A and the binding region of progesterone, which is located from -2056 to -2051. Taken together, our results indicate that CCT6A likely plays an important role in follicle growth, and in granulosa cells, progesterone activates CCT6A transcription via a progesterone response element (PRE) located in the distal promoter of CCT6A.


Assuntos
Chaperonina com TCP-1/genética , Células da Granulosa/metabolismo , Progesterona/farmacologia , Animais , Northern Blotting , Células Cultivadas , Embrião de Galinha , Feminino , Células da Granulosa/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Progesterona/genética
18.
Gen Comp Endocrinol ; 178(2): 314-22, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22732080

RESUMO

Connective tissue growth factor (CTGF) is a cysteine-rich, matrix-associated heparin-binding protein that is important in many cell types as a regulator of cell proliferation, angiogenesis, cell remodelling and other cellular processes. CTGF is necessary for normal follicle growth and luteinisation in mammals. The avian follicular hierarchy provides an excellent experimental model to study developmental events, particularly the role of cellular remodelling factors in the process of folliculogenesis. In this study, we examined CTGF expression and regulation in the hen ovary. CTGF expression was increased considerably as follicular development proceeds in pre-ovulatory follicles, peaking in expression at the time of ovulation. Immunohistochemistry revealed that CTGF protein was concentrated in the cytoplasm of follicular granulosa cells throughout the ovulation cycle. We isolated granulosa cells from the follicles at two key stages of the ovulation cycle (in terms of cellular alteration): during pre-ovulatory growth and during post-ovulatory regression. Follicle-stimulating hormone (FSH) and luteinising hormone (LH) inhibited CTGF expression in pre-ovulatory granulosa cells but stimulated CTGF expression in post-ovulatory granulosa cells. Moreover, TGFß1 stimulated CTGF expression in both pre- and post-ovulatory granulosa cells. Nevertheless, TGFß1 could rescue the inhibition of gonadotrophins on pre-ovulatory granulosa CTGF expression but could not further stimulate CTGF expression in gonadotrophin-treated post-ovulatory granulosa cells. The results of this study indicate that CTGF expression in avian granulosa cells is modulated by a combination of gonadotrophins and TGFß1 according to the different stages of follicle maturation and degradation. The results also suggest that the gonadotrophic action on post-ovulatory follicles in the avian ovary differs from the gonadotrophin-induced luteinisation in mammals.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/metabolismo , Gonadotropinas/farmacologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Ovário/citologia , Fator de Crescimento Transformador beta1/farmacologia , Animais , Western Blotting , Células Cultivadas , Galinhas , Fator de Crescimento do Tecido Conjuntivo/genética , Feminino , Imuno-Histoquímica
19.
In Vitro Cell Dev Biol Anim ; 44(8-9): 305-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18802739

RESUMO

Growth hormone (GH) plays important roles in oocyte development and facilitates the successful production of competent oocytes in many species both in vivo and in vitro. However, the mechanism of GH action on oocyte maturation is not well known. In this paper, the temporospatial messenger ribonucleic acid expression patterns of GH and several other GH-related factors were quantitatively analyzed in porcine cumulus-oocytes complex throughout in vitro maturation (IVM). GH expression was decreased in oocytes during IVM while absent in cumulus cells. GH receptor, insulin-like growth factor-1 (IGF-1), and IGF-1 receptor expressions were also downregulated in oocytes. In cumulus cells, the expression of IGF-1 decreased significantly while IGF-1 receptor expression remained constant. The transcripts of Janus kinase 2 increased in both oocytes and cumulus cells during IVM. The current precise gene expression information provides further evidence to explain the complex network of GH signaling involved in IVM of porcine oocyte.


Assuntos
Células do Cúmulo/metabolismo , Expressão Gênica , Hormônio do Crescimento/genética , Oócitos/metabolismo , Animais , Células do Cúmulo/citologia , Hormônio do Crescimento/metabolismo , Oócitos/citologia , Oogênese/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Somatomedinas/metabolismo , Suínos
20.
Anim Reprod Sci ; 104(2-4): 275-83, 2008 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-17368971

RESUMO

In vitro oocyte growth is the essential technology which enables oocytes to achieve maturation and acquire the competence for subsequent manipulation. There is increasing evidence that members of the transforming growth factor-beta (TGF-beta) superfamily are expressed in a variety of cell types within the ovary in a developmental stage-related manner and function as crucial factors in oocyte growth and follicular development. However, the expression of TGF-beta family members has been studied extensively in follicular compartment cells in the ovaries while poorly explored in the cumulus-oocytes complex (COC) within culture systems. Using semi-quantitative RT-PCR, we investigated the temporal and spatial expression patterns of several bone morphogenetic proteins (BMP-4, BMP-6, BMP-15 and GDF-9), as well as BMP receptors (BMPRIA, BMPRIB, BMPRII and ActRII), in porcine COCs throughout in vitro maturation (IVM). In oocytes, the transcription of BMP-6, BMP-15, GDF-9 and BMPRII were down-regulated, while BMP-4, BMPRIA and BMPRIB remained unchanged during IVM. In cumulus cells, BMP-4 mRNA expression increased significantly, while BMP-6 and ActRII was down-regulated during IVM. Nevertheless, mRNAs of BMPRIA, BMPRIB and BMPRII were constantly expressed in cumulus cells in the process. However, BMP-15 was absent in cumulus cells and ActRII was not detected in oocytes. In addition, hardly any transcription of BMP-2, BMP-5, BMP-7, ActRIA was found in porcine COCs throughout IVM. These data demonstrate a complex BMP-signaling system for member gene expression within porcine COCs during IVM and indicate the need for further functional characterization of these factors during oocyte maturation.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/biossíntese , Proteínas Morfogenéticas Ósseas/biossíntese , Células do Cúmulo/metabolismo , Oócitos/metabolismo , Suínos/fisiologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/genética , Técnicas de Cultura de Células/veterinária , Células do Cúmulo/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Oócitos/ultraestrutura , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Suínos/genética , Suínos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...