1
|
Ru M, Liang H, Ruan J, Haji RA, Cui Y, Yin C, Wei Q, Huang J. Chicken ovarian follicular atresia: interaction network at organic, cellular, and molecular levels. Poult Sci 2024; 103:103893. [PMID: 38870615 PMCID: PMC11225904 DOI: 10.1016/j.psj.2024.103893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
Most of follicles undergo a degenerative process called follicular atresia. This process directly affects the egg production of laying hens and is regulated by external and internal factors. External factors primarily include nutrition and environmental factors. In follicular atresia, internal factors are predominantly regulated at 3 levels; organic, cellular and molecular levels. At the organic level, the hypothalamic-pituitary-ovary (HPO) axis plays an essential role in controlling follicular development. At the cellular level, gonadotropins and cytokines, as well as estrogens, bind to their receptors and activate different signaling pathways, thereby suppressing follicular atresia. By contrast, oxidative stress induces follicular atresia by increasing ROS levels. At the molecular level, granulosa cell (GC) apoptosis is not the only factor triggering follicular atresia. Autophagy is also known to give rise to atresia. Epigenetics also plays a pivotal role in regulating gene expression in processes that seem to be related to follicular atresia, such as apoptosis, autophagy, proliferation, and steroidogenesis. Among these processes, the miRNA regulation mechanism is well-studied. The current review focuses on factors that regulate follicular atresia at organic, cellular and molecular levels and evaluates the interaction network among these levels. Additionally, this review summarizes atretic follicle characteristics, in vitro modeling methods, and factors preventing follicular atresia in laying hens.
Collapse
Affiliation(s)
- Meng Ru
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Haiping Liang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Jiming Ruan
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Ramlat Ali Haji
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Yong Cui
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Chao Yin
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Qing Wei
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Jianzhen Huang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China.
| |
Collapse
|
2
|
Gan X, Wang Y, Gao S, Chen X, Hu S, Wang J, Hu J, Li L, Han C. Co-culture model reveals the characteristics of theca cells and the effect of granulosa cells on theca cells at different stages of follicular development. Reprod Domest Anim 2020; 56:58-73. [PMID: 33103290 DOI: 10.1111/rda.13849] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/21/2020] [Indexed: 12/27/2022]
Abstract
Theca cells (TCs) play an important role in follicular development, which cannot be separated from granulosa cells (GCs). However, compared with mammals, the TCs and the effects of GCs on TCs at different follicular development stages (FDSs) have specific characteristics in avian species, but none of them have been clearly defined. In this study, we established an in vitro co-culture (with GC at the corresponding stage) model of goose TCs at different FDSs (pre-hierarchical, hierarchical and F1) by using a transwell system. The properties of TCs in co-culture at the three FDSs, including cell morphology, activity and intracellular lipid content, as well as the expression of key genes involved in de novo lipogenesis, steroidogenesis, proliferation and apoptosis, were examined and defined. We further compared the mono-culture and co-culture groups. After co-culture, the activity of TCs showed significant (p < .01) increases in all stages; moreover, in pre-hierarchical TCs, the expression levels of FAS, SREBP, 3β-HSD and CCND1 were promoted, and PPARγ, CYP19, BCL2 and CAS3 were inhibited (p < .05); in the hierarchical TCs, the expression levels of PPARγ, FAS, CYP19, CCND1 and BCL2 were promoted, and SREBP, STAR, 3β-HSD and CAS3 were inhibited (p < .05), whereas in the F1 TCs, the expression levels of PPARγ, FAS, 3β-HSD, CYP19 and CCND1 were promoted, and STAR and CAS3 were inhibited (p < .05). These results suggested that GCs at the three FDSs have dynamic and complex influences on the physiological characteristics of TCs, and the influences on TCs at the three FDSs were varied.
Collapse
Affiliation(s)
- Xiang Gan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yushi Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Shanyan Gao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xi Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Chunchun Han
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
3
|
Sirotkin AV, Benčo A, Mlynček M, Harrath AH, Alwasel S, Kotwica J. The involvement of the phosphorylatable and nonphosphorylatable transcription factor CREB-1 in the control of human ovarian cell functions. C R Biol 2019; 342:90-96. [DOI: 10.1016/j.crvi.2019.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/20/2022]
|
4
|
Chen F, Wen X, Lin P, Chen H, Wang A, Jin Y. Activation of CREBZF Increases Cell Apoptosis in Mouse Ovarian Granulosa Cells by Regulating the ERK1/2 and mTOR Signaling Pathways. Int J Mol Sci 2018; 19:ijms19113517. [PMID: 30413092 PMCID: PMC6274897 DOI: 10.3390/ijms19113517] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/27/2018] [Accepted: 10/30/2018] [Indexed: 12/21/2022] Open
Abstract
CREBZF, a multifunction transcriptional regulator, participates in the regulation of numerous cellular functions. The aims of the present study were to detect the localization of CREBZF expression in the ovary and explore the role of CREBZF and related mechanisms in the apoptosis of ovarian granulosa cells. We found by immunohistochemistry that CREBZF was mainly located in granulosa cells and oocytes during the estrous cycle. Western blot analysis showed that SMILE was the main isoform of CREBZF in the ovary. The relationship between apoptosis and CREBZF was assessed via CREBZF overexpression and knockdown. Flow cytometry analysis showed that CREBZF induced cell apoptosis in granulosa cells. Western bolt analysis showed that overexpression of CREBZF upregulated BAX and cleaved Caspase-3, while it downregulated BCL-2. Furthermore, overexpression of CREBZF inhibited the ERK1/2 and mTOR signaling pathways through the phosphorylation of intracellular-regulated kinases 1/2 (ERK1/2) and p70 S6 kinase (S6K1). Moreover, we found that CREBZF also activated autophagy by increasing LC3-II. In summary, these results suggest that CREBZF might play a proapoptotic role in cell apoptosis in granulosa cells, possibly by regulating the ERK1/2 and mTOR signaling pathways.
Collapse
Affiliation(s)
- Fenglei Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China.
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100, Shaanxi, China.
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China.
| | - Xin Wen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China.
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Pengfei Lin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China.
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Huatao Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China.
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Aihua Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Yaping Jin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China.
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
5
|
SIRT1 induces resistance to apoptosis in human granulosa cells by activating the ERK pathway and inhibiting NF-κB signaling with anti-inflammatory functions. Apoptosis 2018; 22:1260-1272. [PMID: 28755171 DOI: 10.1007/s10495-017-1386-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SIRT1, a member of the sirtuin family, has recently emerged as a vital molecule in controlling ovarian function. The aims of the present study were to investigate SIRT1 expression and analyze SIRT1-mediated apoptosis in human granulosa cells (GCs). Human ovarian tissues were subjected to immunohistochemistry for localization of SIRT1 expression. SIRT1 knockdown in a human ovarian GC tumor line (COV434) was achieved by small interfering RNA, and the relationship between apoptosis and SIRT1 was assessed by quantitative reverse transcription polymerase chain reaction and western blotting. We further detected SIRT1 expression in human luteinized GCs. Associations among SIRT1 knockdown, SIRT1 stimulation (resveratrol) and expression of ERK1/2 and apoptotic regulatory proteins were analyzed in cell lines and luteinized GCs. Resveratrol downregulated the levels of nuclear factor (NF)-κB/p65, but this inhibitory effect was attenuated by suppressing SIRT1 activity. The NF-κB/p65 inhibitor pyrrolidine dithiocarbamate achieved similar anti-apoptosis effects. These results suggest that SIRT1 might play an anti-apoptotic role in apoptosis processes in GCs, possibly by sensing and regulating the ERK1/2 pathway, which has important clinical implications. Thus, our study provides a mechanistic link, whereby activation of SIRT1 function might help to sustain human reproduction by maintaining GCs as well as oocytes, offering a novel approach for developing a new class of therapeutic anti-inflammatory agents.
Collapse
|
6
|
Sirotkin AV, Benčo A, Tandlmajerová A, Lauková M, Vašíček D, Laurinčik J, Kornhauser J, Alwasel S, Harrath AH. cAMP response element-binding protein 1 controls porcine ovarian cell proliferation, apoptosis, and FSH and insulin-like growth factor 1 response. Reprod Fertil Dev 2018; 30:1145-1153. [DOI: 10.1071/rd17508] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 01/23/2018] [Indexed: 12/12/2022] Open
Abstract
The aim of the present study was to examine the role of cAMP response element-binding protein (CREB) and its phosphorylation in the regulation of ovarian cell proliferation and apoptosis, and of the response of proliferation and apoptosis to the upstream hormonal stimulators FSH and insulin-like growth factor (IGF) 1. In the first series of experiments, porcine ovarian granulosa cells, transfected or not with a gene construct encoding wild-type CREB1 (CREB1WT), were cultured with and without FSH (0, 1, 10 or 100 ng mL−1). In the second series of experiments, these cells were transfected or not with CREB1WT or non-phosphorylatable mutant CREB1 (CREB1M1) and cultured with and without FSH (0, 1, 10 or 100 ng mL−1) or IGF1 (0, 1, 10 and 100 ng mL−1). Levels of total and phosphorylated (p-) CREB1, proliferating cell nuclear antigen (PCNA), a marker of proliferation, and BAX, a marker of apoptosis, were evaluated by western immunoblotting and immunocytochemical analysis. Transfection of cells with CREB1WT promoted accumulation of total CREB1 within cells, but p-CREB1 was not detected in any cell group. Both CREB1WT and CREB1M1 reduced cell proliferation and apoptosis. Addition of 10 and 100 ng mL−1 FSH to non-transfected cells promoted CREB1 accumulation and apoptosis, whereas cell proliferation was promoted by all concentrations of FSH tested. FSH activity was not modified in cells transfected with either CREB1WT or CREB1M1. IGF1 at 100 ng mL−1 promoted cell proliferation, whereas all concentrations of IGF1 tested reduced apoptosis. Transfection with either CREB1WT or CREB1M1 did not modify the effects of either FSH or IGF1, although CREB1M1 reversed the effect of IGF1 on apoptosis from inhibitory to stimulatory. These observations suggest that CREB1 is involved in the downregulation of porcine ovarian cell proliferation and apoptosis. The absence of visible CREB1 phosphorylation and the similarity between the effects of CREB1WT and CREB1M1 transfection indicate that phosphorylation is not necessary for CREB1 action on these processes. Furthermore, the observations suggest that FSH promotes both ovarian cell proliferation and apoptosis, whereas IGF1 has proliferation-promoting and antiapoptotic properties. The effect of FSH on CREB1 accumulation and the ability of CREB1M1 to reverse the effects of IGF1 on apoptosis indicate that CREB1 is a mediator of hormonal activity, but the inability of either CREB1WT or CREBM1transfection to modify the primary effects of FSH and IGF1 suggest that CREB1 and its phosphorylation do not mediate the action of these hormones on ovarian cell proliferation and apoptosis.
Collapse
|
7
|
Sirotkin AV, Harrath AH, Grossmann R. Metabolic status and ghrelin regulate plasma levels and release of ovarian hormones in layer chicks. Physiol Res 2016; 66:85-92. [PMID: 27782746 DOI: 10.33549/physiolres.933306] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to examine the role of nutritional status, the metabolic hormone ghrelin and their interrelationships in the control of chicken hormones involved in the regulation of reproduction. For this purpose, we identified the effect of food deprivation, administration of ghrelin 1-18 and their combination on plasma levels of testosterone (T), estradiol (E), arginine-vasotocin (AVT) and growth hormone (GH) as well as the release of these hormones by isolated and cultured ovarian fragments. It was observed that food deprivation reduces plasma T and E and increases plasma AVT and GH levels. Food restriction also reduced the amount of E produced by isolated ovaries, but it did not affect the ovarian secretion of T and AVT. No ovarian GH secretion was detected. Ghrelin administered to ad libitum fed chickens did not affect plasma T and E levels, but it did increase plasma GH and AVT concentrations. Moreover, it partially prevented the effect of food deprivation on plasma E and AVT levels, but not on T or GH levels. Ghrelin administration to control birds promoted ovarian T, but not E or AVT release and reduced T and no other hormonal outputs in birds subjected to food restriction. Our results (1) confirmed the ovarian origin of the main plasma T and E and the extra-ovarian origin of the main blood AVT and GH; (2) showed that food deprivation-induced suppression of reproduction may be caused by suppression of T and E and the promotion of AVT and GH release; (3) suggest the involvement of ghrelin in control chicken E, AVT and GH output; and (4) indicates that ghrelin can either mimic or modify the effect of the intake of low calories on chicken plasma and ovarian hormones, i.e. it can mediate the effect of metabolic state on hormones involved in the control of reproduction.
Collapse
Affiliation(s)
- A V Sirotkin
- Department Zoology and Anthropology, Constantine the Philosopher University, Nitra, Slovakia.
| | | | | |
Collapse
|
8
|
Sirotkin AV, Harrath AH, Grossmann R. The role of metabolic state and obestatin in control of chicken ovarian hormone release. Poult Sci 2016; 95:1939-42. [PMID: 27030691 DOI: 10.3382/ps/pew108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2016] [Indexed: 11/20/2022] Open
Abstract
The aim of the present study was to examine the role and interrelationships between calorie restriction and obestatin in the control of hormone release by chicken ovarian tissue. For this purpose, we compared the release of progesterone (P), testosteron (T), estradiol (E), and arginine-vasotocin (AVT) by ovarian fragments isolated from chicken subjected and not subjested to food restriction, as well as the response of these ovarian fragments to obestatin additions.It was observed that food restriction promoted release of P, reduced output of T, but did not affect basal E and AVT release. Obestatin addition reduced E, promoted AVT, and did not alter P and T release by ovarian tissue isolated from ad libitum fed chicken. In ovarian fragments of fasted hens it reduced E, promoted T, and did not influence P and AVT release.The present observations demonstrate (1) that obestatin can directly control the release of avian ovarian hormones - regulators of reproduction, (2) that metabolic state can control the release of these hormones, and (3) metabolic state can alter the response of ovarian hormones to obestatin.
Collapse
Affiliation(s)
- Alexander V Sirotkin
- Dept. Zoology and Anthropology, Constantine the Philosopher University, 949 74 Nitra, Slovakia Dept. Genetics and Reproduction, Research Institute of Animal Production, 949 59 Lužianky
| | - Abdel Halim Harrath
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Roland Grossmann
- Dept. Functional Genomics & Bioregulation, Friedrich Loeffler Institute, Mariensee, Neustadt, Germany
| |
Collapse
|
9
|
Sirotkin AV, Harrath AH, Grossmann R. Comparison of the effects of human and chicken ghrelin on chicken ovarian hormone release. Comp Biochem Physiol B Biochem Mol Biol 2016; 201:59-63. [PMID: 27378405 DOI: 10.1016/j.cbpb.2016.06.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 06/20/2016] [Accepted: 06/27/2016] [Indexed: 11/26/2022]
Abstract
The aim of the present experiments was to examine the species-specific and cell-specific effects of ghrelin on chicken ovarian hormone release. For this purpose, we compared the effects of chicken and human ghrelin on the release of estradiol (E), testosterone (T), progesterone (P) and arginine-vasotocin (AVT) by cultured fragments of chicken ovarian follicles and on the release of T and AVT by cultured ovarian granulosa cells. In cultured chicken ovarian fragments, both human and chicken ghrelin promoted E release. T output was stimulated by chicken ghrelin but not by human ghrelin. No effect of either human or chicken ghrelin on P release was observed. Human ghrelin promoted but chicken ghrelin suppressed AVT release by chicken ovarian fragments. In cultured ovarian granulosa cells, human ghrelin inhibited while chicken ghrelin stimulated T release. Both human and chicken ghrelin suppressed AVT output by chicken granulosa cells. These data confirm the involvement of ghrelin in the control of ovarian secretory activity and demonstrate that the effect of ghrelin is species-specific. The similarity of avian ghrelin on avian ovarian granulosa cells and ovarian fragments (containing both granulosa and theca cells) suggests that ghrelin can influence chicken ovarian hormones primarily by acting on granulosa cells.
Collapse
Affiliation(s)
- Alexander V Sirotkin
- Dept. of Zoology and Anthropology, Constantine the Philosopher University, 949 74 Nitra, Slovakia; Dept. of Genetics and Reproduction, Research Institute of Animal Production, 949 59 Lužianky, Slovakia; King Saud University, Zoology Department, College of Science, Riyadh, Saudi Arabia.
| | - Abdel Halim Harrath
- King Saud University, Zoology Department, College of Science, Riyadh, Saudi Arabia.
| | - Roland Grossmann
- Dept. of Functional Genomics & Bioregulation, Friedrich Loeffler Institute, Mariensee, Neustadt, Germany.
| |
Collapse
|
10
|
Sirotkin AV, Grossmann R. Interrelationship between feeding level and the metabolic hormones leptin, ghrelin and obestatin in control of chicken egg laying and release of ovarian hormones. Comp Biochem Physiol A Mol Integr Physiol 2015; 184:1-5. [PMID: 25645297 DOI: 10.1016/j.cbpa.2015.01.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 12/16/2014] [Accepted: 01/23/2015] [Indexed: 12/18/2022]
Abstract
The aim of the present experiment is to examine the role of nutritional status, metabolic hormones and their interrelationships in the control of chicken ovarian ovulatory and secretory activity. For this purpose, we identified the effect of food restriction, administration of leptin, ghrelin 1-18, obestatin and combinations of food restriction with these hormones for 3days on chicken ovulation (egg laying) rate and ovarian hormone release. The release of progesterone (P), testosterone (T), estradiol (E) and arginine-vasotocin (AVT) by isolated and cultured ovarian fragments was determined by EIA. It was observed that food restriction significantly reduced the egg-laying rate, T, E and AVT release and promoted P output by ovarian fragments. Leptin, administrated to ad libitum-fed chickens, did not change these parameters besides promoting E release. Nevertheless, administration of leptin was able to prevent the effect of food restriction on ovulation, T and E (but not P or AVT) release. Ghrelin 1-18 administration to ad libitum-fed birds did not affect the measured parameters besides a reduction in P release. Ghrelin 1-18 administration prevented the food restriction-induced decrease in ovarian T, E and AVT, but it did not change P output or egg laying. Obestatin administrated to control chicken promoted their ovarian P, E and inhibited ovarian AVT release but did not affect egg laying. It was able to promote the effect of food restriction on P, T and AVT, but not E release or egg laying. Our results (1) confirm an inhibitory effect of food restriction on chicken ovulation rate; (2) shows that food restriction-induced reduction in egg laying is associated with a decrease in ovarian T, E and AVT and an increase in ovarian P release; (3) confirm the involvement of metabolic hormones leptin, ghrelin and obestatin in the control of chicken ovarian hormones output; and (4) the ability of metabolic hormones to mimic/antagonize or prevent/promote the effects of food restriction on both egg laying and ovarian hormones demonstrates that nutritional status can influence ovarian ovulatory and endocrine functions via changes in metabolic hormones.
Collapse
Affiliation(s)
- Alexander V Sirotkin
- Department of Zoology and Anthropology, Constantine the Philosopher University, 949 74 Nitra, Slovakia; Department of Genetics and Reproduction, Research Centre of Animal Production, Hlohovská 2, 949 59 Lužianky, Slovakia.
| | - Roland Grossmann
- Department of Functional Genomics and Bioregulation, Ïnstitute of Animal Genetics, Friedrich-Loeffler Institut (FLI), Mariensee, 31535 Neustadt, Germany.
| |
Collapse
|
11
|
Kolesarova A, Sirotkin AV, Mellen M, Roychoudhury S. Possible intracellular regulators of female sexual maturation. Physiol Res 2014; 64:379-86. [PMID: 25536325 DOI: 10.33549/physiolres.932838] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Protein kinases, transcription factors and other apoptosis- and proliferation-related proteins can regulate reproduction, but their involvement in sexual maturation remains to be elucidated. The general aim of the in vivo and in vitro experiments with porcine ovarian granulosa cells was to identify possible intracellular regulators of female sexual maturation. For this purpose, proliferation (expression of proliferating cell nuclear antigen - PCNA, mitogen-activated protein kinases - ERK 1,2 related MAPK and cyclin B1), apoptosis (expression of the apoptotic protein Bax and apoptosis regulator Bcl-2 protein), expression of some protein kinases (cAMP dependent protein kinase - PKA, cGMP-dependent protein kinase - PKG, tyrosine kinase - TK) and cAMP responsive element binding protein 1 (CREB-1) was examined in granulosa cells isolated from ovaries of immature and mature gilts. Expression of PCNA, ERK1,2 related MAPK, cyclin B1, Bcl-2, Bax, PKA, CREB-1, TK and PKG in porcine granulosa cells were detected by immunocytochemistry. Sexual maturation was associated with significant increase in the expression of Bcl-2, Bax, PKA, CREB-1 and TK and with decrease in the expression of ERK1,2 related MAPK, cyclin B1 and PKG in granulosa cells. No significant difference in PCNA expression was noted. The present data obtained from in vitro study indicate that sexual maturation in females is influenced by puberty-related changes in porcine ovarian signaling substances: increase in Bcl-2, Bax, PKA, CREB-1, TK and decrease in ERK1,2 related MAPK, cyclin B1 and PKG. It suggests that these signaling molecules could be potential regulators of porcine sexual maturation.
Collapse
Affiliation(s)
- A Kolesarova
- Department of Animal Physiology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture, Nitra, Slovak Republic.
| | | | | | | |
Collapse
|
12
|
Sirotkin AV, Makarevich AV, Grosmann R. Protein kinases and ovarian functions. J Cell Physiol 2010; 226:37-45. [DOI: 10.1002/jcp.22364] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
13
|
Lebedeva IY, Lebedev VA, Grossmann R, Parvizi N. Age-dependent role of steroids in the regulation of growth of the hen follicular wall. Reprod Biol Endocrinol 2010; 8:15. [PMID: 20156346 PMCID: PMC2833167 DOI: 10.1186/1477-7827-8-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 02/15/2010] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The ovaries are the primary targets of senescence effects in mammalian and avian species. In the present study, relationships between reproductive aging, sex steroids and the growth pattern of the pre-ovulatory follicle wall were investigated using young hens with long clutch (YLC), old hens with long clutch (OLC), old hens with short clutch (OSC), and old hens with interrupted long clutch (OILC). METHODS Experiment 1: Hens were sacrificed 1.5 and 14.5 h after ovulation. Experiment 2: YLC and OILC hens were sacrificed 3.5 h after treatments with LH and/or aminoglutethimide (AG), an inhibitor of steroid synthesis. Volumes of pre-ovulatory follicles (F1-F5) and plasma concentrations of ovarian steroids were determined. Experiment 3: Granulosa and theca cells from F3 follicles of OSC and/or YLC hens were exposed in vitro to estradiol-17beta (E2), testosterone (T) and LH and the proliferative activity of the cells was examined using CellTiter 96 Aqueous One Solution Assay. RESULTS In YLC and OLC groups, the total volume of F1-F5 follicles rose between 1.5 and 14.5 h after ovulation (P < 0.01), negatively correlating with the plasma level of E2 (P < 0.01). There was no growth of pre-ovulatory follicles in the middle of the ovulatory cycle in the OSC group, with a positive correlation being present between E2 and the follicular volume (P < 0.05). In young hens, AG caused a rise in the total follicular volume. This rise was associated with a fall in E2 (r = -0.54, P < 0.05). E2 enhanced proliferation of granulosa cells from YLC and OSC groups. The proliferative activity of granulosa and theca cells of YLC hens depended on the interaction between T and LH (P < 0.01). CONCLUSIONS These data indicate for the first time that the growth pattern of pre-ovulatory follicles during the ovulatory cycle changes in the course of reproductive aging. E2 seems to play a dual role in this adjustment; it stimulates the growth of the follicular wall in reproductive aged hens, whereas it may inhibit this process in young birds. T and LH are apparently involved in the growth regulation during the pre-ovulatory surge in young hens.
Collapse
Affiliation(s)
- Irina Y Lebedeva
- Department of Functional Genomics and Bioregulation, Institute of Animal Genetics, FLI, Mariensee, 31535 Neustadt, Germany
- Center of Biotechnology and Molecular Diagnostics, Russian Research Institute of Animal Breeding, Podolsk, 142132 Russia
| | - Vladimir A Lebedev
- Department of Functional Genomics and Bioregulation, Institute of Animal Genetics, FLI, Mariensee, 31535 Neustadt, Germany
- Department of Genetics and Biotechnology, Research Institute for Farm Animal Genetics and Breeding, Pushkin, St Petersburg, 196625 Russia
| | - Roland Grossmann
- Department of Functional Genomics and Bioregulation, Institute of Animal Genetics, FLI, Mariensee, 31535 Neustadt, Germany
| | - Nahid Parvizi
- Department of Functional Genomics and Bioregulation, Institute of Animal Genetics, FLI, Mariensee, 31535 Neustadt, Germany
| |
Collapse
|
14
|
Ou JT, Tang SQ, Sun DX, Zhang Y. Polymorphisms of three neuroendocrine-correlated genes associated with growth and reproductive traits in the chicken. Poult Sci 2009; 88:722-7. [PMID: 19276414 DOI: 10.3382/ps.2008-00497] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The identification and utilization of potential candidate genes for QTL with significant effects on economically important traits are becoming increasingly important in poultry breeding programs. Chicken insulin-like growth factor binding protein 1 and 3 and signal transducers and activators of transcription 5B (STAT5B) genes are 3 essential nodes for signaling pathways and gene networks of growth and reproduction. The pooled DNA sequencing approach was used for identification of 9 SNP of the 5' upstream region of the 3 genes. A total of 826 individuals from Beijing You chicken were genotyped for 5 SNP using a modified PCR-RFLP method and the association with chicken growth and reproductive traits was studied using the GLM procedure. The T56039403C (T-808C) SNP of the insulin-like growth factor binding protein 1 gene was associated with BW at 10 wk of age (P = 0.0061), and the C56072547T (C-968T) SNP of the insulin-like growth factor binding protein 3 gene was associated with BW at 8 and 10 wk of age (P = 0.0056 and P = 0.0016, respectively). The C4535156T (C-1591T), G4533815A (G-250A), and G4533675C (G-110C) SNP of the STAT5B gene were associated with age at first egg (P = 0.0143, P = 0.0088, and P = 0.0114, respectively). Moreover, Lewontin's D' (|D'|) and r(2) of C4535156T and G4533815A SNP, C4535156T and G4533675C SNP, and G4533815A and G4533675C SNP of the STAT5B gene were 0.939 and 0.852, 0.967 and 0.858, and 0.971 and 0.896, respectively. The 3 SNP were strong-linked with each other and lay within a haplotype block. Our results suggest that these SNP were significantly associated with early growth or with sexual maturation in chickens, or both, and may be potential molecular markers for MAS.
Collapse
Affiliation(s)
- J T Ou
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | | | | | | |
Collapse
|
15
|
Sirotkin AV, Ovcharenko D, Benčo A, Mlynček M. Protein kinases controlling PCNA and p53 expression in human ovarian cells. Funct Integr Genomics 2008; 9:185-95. [DOI: 10.1007/s10142-008-0102-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2008] [Revised: 11/25/2008] [Accepted: 11/25/2008] [Indexed: 11/30/2022]
|
16
|
Onagbesan O, Bruggeman V, Decuypere E. Intra-ovarian growth factors regulating ovarian function in avian species: a review. Anim Reprod Sci 2008; 111:121-40. [PMID: 19028031 DOI: 10.1016/j.anireprosci.2008.09.017] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Revised: 09/15/2008] [Accepted: 09/23/2008] [Indexed: 11/19/2022]
Abstract
There is now overwhelming evidence that the avian ovary is a site of production and action of several growth factors that have also been implicated in the functioning of the mammalian ovary. Several members of the Insulin-like growth factor family (IGF), the Epidermal growth factor family (EGF), the Transforming growth factor-beta family (TGF-beta), Fibroblast growth factors (FGF), the Tumour necrosis factor-alpha (TNF-alpha), and others, have been identified either in the granulosa and/or theca compartments of ovarian follicles and in the embryonic and juvenile ovary. Some have been specifically localized to the germinal disc area containing the oocyte. The mRNAs and proteins of the growth factors, receptor proteins and binding proteins of some of the members of each group have been reported in the chicken, turkey, quail and duck. The intra-ovarian roles reported for the different growth factors include regulation of cell proliferation, steroidogenesis, follicle selection, modulation of gonadotrophin action, control of ovulation rate, cell differentiation, production of growth factors, etc. The aim of this paper is to provide a review of the current knowledge of avian ovarian growth factors and their biological activity in the ovary. The review covers the detection of the growth factor proteins, the receptor proteins, binding proteins, their spatial and temporal distribution in embryonic, juvenile and adult ovaries and their regulation. The paper also discusses their roles in each follicular compartment during follicular development. Greater emphasis is given to the major growth factors that have been studied to greater detail and others are discussed very briefly.
Collapse
|
17
|
Hrabia A, Paczoska-Eliasiewicz HE, Berghman LR, Harvey S, Rzasa J. Expression and localization of growth hormone and its receptors in the chicken ovary during sexual maturation. Cell Tissue Res 2008; 332:317-28. [PMID: 18335240 DOI: 10.1007/s00441-008-0595-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2007] [Accepted: 01/28/2008] [Indexed: 11/30/2022]
Abstract
Roles of pituitary growth hormone (GH) in female reproduction are well established. Autocrine and/or paracrine actions of GH in the mammalian ovary have additionally been proposed, although whether the ovary is an extra-pituitary site of GH expression in the laying hen is uncertain. This possibility has therefore been assessed in the ovaries of Hy-Line hens before (between 10-16 weeks of age) and after (week 17) the onset of egg laying. Reverse transcription/polymerase chain reaction (RT-PCR) analysis has consistently detected a full-length (690 bp) pituitary GH cDNA in ovarian stroma from 10 weeks of age, although GH expression is far lower than that in the pituitary gland or hypothalamus. GH mRNA is also present in small (>1-4 mm diameter) follicles after their ontogenetic appearance at 14 weeks of age and in all other developing follicles after 16 weeks of age (>4-30 mm diameter). Immunoreactivity for GH is similarly present in the ovarian stroma from 10 weeks of age and in small (<4 mm diameter) and large (>4-30 mm) follicles from 14 and 16 weeks of age, respectively. The relative intensity of GH staining in the ovarian follicles is consistently greater in the granulosa cells than in the thecal cells and is comparable with that in the follicular epithelium. A 321-bp fragment of GH receptor (GHR) cDNA, coding for the intracellular domain of the receptor, has also been detected by RT-PCR in the ovary and is present in stromal tissue by 10 weeks of age, in small follicles (<4 mm diameter) by 14 weeks of age, and in larger follicles (>4-30 mm diameter) from 16 weeks. GHR immunoreactivity has similarly been detected, like GH, in the developing ovary and in all follicles and is more intense in granulosa cells than in the theca interna or externa. The expression and location of the GH gene therefore parallels that of the GHR gene during ovarian development in the laying hen, as does the appearance of GH and GHR immunoreactivity. These results support the possibility that GH has autocrine and/or paracrine actions in ovarian function prior to and after the onset of lay in hens.
Collapse
Affiliation(s)
- Anna Hrabia
- Department of Animal Physiology, Agricultural University in Krakow, Al. Mickiewicza 24/28, 30-059 Krakow, Poland.
| | | | | | | | | |
Collapse
|
18
|
Sirotkin AV, Grossmann R. Effects of ghrelin and its analogues on chicken ovarian granulosa cells. Domest Anim Endocrinol 2008; 34:125-34. [PMID: 17207955 DOI: 10.1016/j.domaniend.2006.11.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2006] [Revised: 11/06/2006] [Accepted: 11/29/2006] [Indexed: 11/23/2022]
Abstract
The aim of these in vitro experiments was (1) to examine the effects of ghrelin on the basic functions of ovarian cells (proliferation, apoptosis, secretory activity); (2) to determine the possible involvement of the GHS-R1a receptor and PKA- and MAPK-dependent post-receptor intracellular signalling cascades; (3) to identify the active part of the 28-amino acid molecule responsible for the effects of ghrelin on ovarian cells. We compared the effect of full-length ghrelin 1-28, a synthetic activator of GHS-R1a, GHRP6, and ghrelin molecular fragments 1-18 and 1-5 on cultured chicken ovarian cells. Indices of cell apoptosis (expression of the apoptotic peptide bax and the anti-apoptotic peptide bcl-2), proliferation (expression of proliferation-associated peptide PCNA), and expression of protein kinases (PKA and MAPK) within ovarian granulosa cells were analysed by immunocytochemistry. The secretion of progesterone (P(4)), testosterone (T), estradiol (E(2)) and arginine-vasotocin (AVT) by isolated ovarian follicular fragments was evaluated by RIA/EIA. It was observed that accumulation of bax was increased by ghrelin 1-28, GHRP6 and ghrelin 1-18, but not by ghrelin 1-5. Expression of bcl-2 was suppressed by addition of ghrelin 1-28, GHRP6 and ghrelin 1-5, but promoted by ghrelin 1-18. The occurrence of PCNA was reduced by ghrelin 1-28, GHRP6, ghrelin 1-18 and ghrelin 1-5. An increase in the expression of MAPK/ERK1, 2 was observed after addition of ghrelin 1-28, GHRP6 and ghrelin 1-18, but not ghrelin 1-5. The accumulation of PKA decreased after treatment with ghrelin 1-28 and increased after treatment with GHRP6 and ghrelin 1-18 but not ghrelin 1-5. Secretion of P(4) by ovarian follicular fragments was decreased after addition of ghrelin 1-28 or ghrelin 1-5 but stimulated by GHRP6 and ghrelin 1-18. Testosterone secretion was inhibited by ghrelins 1-28 and 1-18, but not by GHRP6 or ghrelin 1-5. Estradiol secretion was reduced after treatment with ghrelin 1-28 but stimulated by ghrelins 1-18 and 1-5; GHRP6 had no effect. AVT secretion was stimulated by ghrelin 1-28, GHRP6 and ghrelin 1-18, but inhibited by ghrelin 1-5. The comparison of the effects of the four ghrelin analogues on nine parameters of ovarian cells suggest (1) a direct effect of ghrelin on basic ovarian functions-apoptosis, proliferation, steroid and peptide hormone secretion; (2) that the majority of these effects can be mediated through GHS-R1a receptors; (3) an effect of ghrelin on MAPK- and PKA-dependent intracellular mechanisms, which can potentially mediate the action of ghrelin at the post-receptor level; (4) that ghrelin residues 5-18 may be responsible for the major effects of ghrelin on the avian ovary.
Collapse
Affiliation(s)
- A V Sirotkin
- Research Institute of Animal Production, Hlohovská 2, 949 92 Nitra, Slovakia.
| | | |
Collapse
|
19
|
Chen CF, Shiue YL, Yen CJ, Tang PC, Chang HC, Lee YP. Laying traits and underlying transcripts, expressed in the hypothalamus and pituitary gland, that were associated with egg production variability in chickens. Theriogenology 2007; 68:1305-15. [PMID: 17931698 DOI: 10.1016/j.theriogenology.2007.08.032] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Revised: 08/29/2007] [Accepted: 08/11/2007] [Indexed: 10/22/2022]
Abstract
The objective was to characterize the potential laying traits and underlying transcripts expressed in the hypothalamus and pituitary gland that were associated with egg production variability in five genetic stocks of chickens: two commercial lines, Red- (n=12) and Black-feather (n=14) Taiwan country chickens (TCCs); two selected lines of TCCs, B (high body weight/comb size; n=17) and L2 (high-egg production; n=14); and a commercial single comb White Leghorn (WL; n=17). Six laying traits, age at first egg, clutch length, pause length, oviposition lag within clutch, follicle rapid growth period, and rate of yolk accumulation were measured. The significance of differential values among five chicken stocks and correlation coefficients between laying traits and number of eggs to 50 weeks of age or laying rate after first egg, and the expression level of 33 transcripts were determined. Longer clutch length and shorter oviposition lag within clutch contributed to a higher number of eggs to 50 weeks of age or laying rate after first egg in L2 (P<0.05) and WL strains (P<0.05). However, their rate of yolk accumulation (P<0.05) and follicle rapid growth period (P<0.05) were different, indicating the accumulation of different alleles after long-term, independent selection. Across all five strains, numbers of eggs to 50 weeks of age were positive correlated with average clutch length (P<0.05) as well as the rate of yolk accumulation (P<0.05). Expressions of PLAG1, STMN2, PGDS, PARK7, ANP32A, PCDHA@, SCG2, BDH and SAR1A transcripts contributed to number of eggs to 50 weeks of age (P<0.05) or laying rate after first egg (P<0.05). Analysis of correlation coefficients indicated that PLAG1 additionally played roles in decreasing average pause length. Two transcripts, PRL and GARNL1, specifically contributed to number of eggs to 50 weeks of age or laying rate after first egg by reducing oviposition lag within clutch (P<0.05) and/or increasing average clutch length (P<0.05), respectively. Expression level of NCAM1, contributed to laying rate after first egg by association with a shorter oviposition lag within clutch (P<0.05). The current study attributed egg production phenotype in five strains into several laying traits; correlations between these traits and expression levels of underlying transcripts expressed in the hypothalamus and pituitary gland were also established.
Collapse
Affiliation(s)
- Chih-Feng Chen
- Department of Animal Science, National Chung-Hsing University, 402, Kuo-Kuang Road, Taichung, Taiwan
| | | | | | | | | | | |
Collapse
|
20
|
Sirotkin AV, Grossmann R. Leptin directly controls proliferation, apoptosis and secretory activity of cultured chicken ovarian cells. Comp Biochem Physiol A Mol Integr Physiol 2007; 148:422-9. [PMID: 17604668 DOI: 10.1016/j.cbpa.2007.06.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2007] [Revised: 05/31/2007] [Accepted: 06/01/2007] [Indexed: 11/24/2022]
Abstract
The aim of our in-vitro experiments was to examine, whether leptin can directly control functions of avian ovarian cells and to outline potential intracellular mediators of its effects. Granulosa cells or fragments of ovarian follicular wall were cultured with leptin (0, 1, 10 or 100 ng/mL medium). The expression of peptides involved in apoptosis (TdT, bax, its binding protein, bcl-2, ASK-1 and p53), cell cycle-related peptides (PCNA and cyclin B1), release of hormones (progesterone, testosterone, estradiol, arginine-vasotocin), as well as the expression of protein kinases (PKA, MAPK/ERK1,2 and CDK/p34) in the ovarian cells were examined by using immunocytochemistry, TUNEL, SDS-PAGE-Western immunoblotting, EIA and RIA. It was found that leptin inhibited expression of all markers of cytoplasmic apoptosis (bax, ASK-1 and p53), stimulated expression of anti-apoptotic peptide bcl-2, but did not affect nuclear DNA fragmentation (TdT). Furthermore, leptin inhibited expression of PCNA (marker of S-phase of mitosis), but not of cyclin B1 (marker of G phase of cell cycle). Moreover, it promoted release of progesterone and estradiol, suppressed release of testosterone, but did not affect arginine-vasotocin. Finally, leptin inhibited expression of MAPK/ERK1,2 and CDK/p34 and stimulated expression of PKA. The present observations demonstrate that leptin can directly control basic chicken ovarian functions - inhibit cytoplasmic apoptosis and proliferation (S-phase, but not G-phases of mitosis), regulate secretory activity (release of steroids, but not nonapeptide hormone) and expression of MAPK, PKA and CDC2, which might be potential intracellular mediators of leptin action.
Collapse
Affiliation(s)
- A V Sirotkin
- Research Institute of Animal Production, Slovak Centre of Agricultural Studies, Hlohovská 2, 949 92 Nitra, Slovakia.
| | | |
Collapse
|
21
|
Sirotkin AV, Grossmann R. The role of ghrelin and some intracellular mechanisms in controlling the secretory activity of chicken ovarian cells. Comp Biochem Physiol A Mol Integr Physiol 2007; 147:239-46. [PMID: 17293148 DOI: 10.1016/j.cbpa.2006.12.038] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Revised: 12/28/2006] [Accepted: 12/30/2006] [Indexed: 10/23/2022]
Abstract
The general aim of these in-vitro experiments was to determine whether ghrelin controls the secretory activity of chicken ovarian cells and whether its action is mediated by TK-, MAPK-, CDK- or PKA-dependent intracellular mechanisms. We postulated that particular protein kinases could be considered as mediators of ghrelin action (a) if they are controlled by ghrelin, and (b) if blockers of these kinases modify the action of ghrelin. In our in-vitro experiments we investigated whether ghrelin altered the accumulation of TK, MAPK, CDK and PKA in chicken ovarian cells and whether ghrelin, with or without blockers of MAPK, CDK and PKA, affected the secretion of progesterone (P4), testosterone (T), estradiol (E2) or arginine-vasotocin (AVT). In the first series of experiments, the influence of a ghrelin 1-18 analogue (1, 10 or 100 ng/mL) was studied on the expression of TK, MAPK and PKA in cultured chicken ovarian granulosa cells. The percentage of cells containing TK/phosphotyrosine MAPK/ERK1, 2 and PKA was determined using immunocytochemistry. Ghrelin increased the expression of both TK and MAPK. The low concentration of ghrelin (1 ng/mL) increased the accumulation of PKA in ovarian cells whilst the high concentration (100 ng/mL) decreased it. The 10 ng/mL concentration had no effect. In the second series of experiments, the effects of the ghrelin analogue combined with an MAPK blocker (PD98059; 100 ng/mL), a CDK blocker (olomoucine; 1 microg/mL), or a PKA blocker (KT5720; 100 ng/mL), were tested for their effects on the secretion of hormones by cultured fragments of chicken ovarian follicular wall. P4, T, E2 and AVT secretions were measured using RIA and EIA. Ghrelin increased T and decreased E2, but did not affect P4 or AVT secretion. The PKA blocker promoted P4 secretion and suppressed E2 and AVT, but did not affect T secretion. It prevented or even reversed the effect of ghrelin on T and E2, but did not modify its effect on AVT secretion. The MAPK blocker enhanced P4 and T and reduced AVT, but did not affect E2 secretion. It was able to prevent or reverse the effect of ghrelin on T and E, and it induced a stimulatory effect of ghrelin on AVT secretion. The CDK blocker reduced the secretion of AVT, but had no effect on steroid hormone secretion. It induced the stimulatory influence of ghrelin on the secretion of P4 and AVT, but did not modify the effect of ghrelin on other hormones. These observations clearly demonstrate that ghrelin is a potent regulator of the secretory activity of ovarian cells and of TK, MAPK and PKA. Furthermore, they suggest that MAPK-, CDK- and PKA-dependent intracellular mechanisms are involved in the control of ovarian secretion and that they mediate the effects of ghrelin on these processes.
Collapse
Affiliation(s)
- A V Sirotkin
- Research Institute of Animal Production, Hlohovská 2, 949 92 Nitra, Slovakia.
| | | |
Collapse
|
22
|
Sirotkin AV, Grossmann R, María-Peon MT, Roa J, Tena-Sempere M, Klein S. Novel expression and functional role of ghrelin in chicken ovary. Mol Cell Endocrinol 2006; 257-258:15-25. [PMID: 16891055 DOI: 10.1016/j.mce.2006.06.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 06/06/2006] [Accepted: 06/06/2006] [Indexed: 10/24/2022]
Abstract
Ghrelin has recently emerged as pleiotropic regulator of a wide array of endocrine and non-endocrine functions. The former likely includes the control of gonadal function, as expression of ghrelin and its putative receptor, the GH secretagogue receptor type 1a (GHS-R1a), has been described in mammalian gonads, and direct effects of ghrelin in the control of testicular secretion and cell proliferation have been reported. Yet, the expression and/or functional role of ghrelin in gonads from non-mammalian species remain to be analyzed. The present study aimed to evaluate the expression of ghrelin and GHS-R genes in the chicken ovary, and to assess the potential involvement of ghrelin in the direct control of chick ovarian function. To this end, RT-PCR assays for ghrelin and GHS-R1a mRNAs were performed in ovarian tissue, and cultures of chicken ovarian cells were conducted in the presence of increasing doses (1, 10 or 100 ng/ml) of the ghrelin analog, ghrelin 1-18. Our results demonstrate that both ghrelin and GHS-R1a mRNAs are expressed in chick ovarian tissue. Moreover, challenge of ovarian granulosa cells with ghrelin 1-18 was able to induce markers of proliferation (i.e. expression of both PCNA and cyclin), and to modulate markers of apoptosis (i.e. decreased expression of caspase-3, bax, bcl-2 and TUNEL-positive cells). Moreover, ghrelin 1-18 increased the expression of PCNA, cyclin, bax and p53 in cultures of ovarian follicular fragments, where it also stimulated the release of progesterone, estradiol, arginine-vasotocin (AVT) and IGF-I, but not of testosterone. In conclusion, our study provides novel evidence for the gonadal expression of the genes encoding ghrelin and its cognate receptor in a non-mammalian species, i.e. the chicken ovary, and unravels the potential involvement of this newly discovered molecule in the control of key gonadal functions in the chick, such as proliferation, apoptosis, and hormone release.
Collapse
Affiliation(s)
- A V Sirotkin
- Department of Animal Genetics & Reproduction, Research Institute of Animal Production, Hlohovská 2, 949 92 Nitra, Slovakia.
| | | | | | | | | | | |
Collapse
|
23
|
Sirotkin AV, Grossmann R. The role of protein kinase A and cyclin-dependent (CDC2) kinase in the control of basal and IGF-II-induced proliferation and secretory activity of chicken ovarian cells. Anim Reprod Sci 2005; 92:169-81. [PMID: 16029936 DOI: 10.1016/j.anireprosci.2005.05.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2004] [Revised: 05/11/2005] [Accepted: 05/11/2005] [Indexed: 11/19/2022]
Abstract
The aim of these experiments was to study the role of protein kinase A (PKA), cyclin-dependent kinase 2 (CDC2) and insulin-like growth factor II (IGF-II) in the control of ovarian function in domestic fowl, as well as the role of PKA and CDC2 in mediating the effects of IGF-II on the ovary. For this purpose, we studied the influence of an inhibitor of PKA (KT5720; 50 ng/ml), a CDC2 blocker (olomoucine; 1 microg/ml), IGF-II (0, 1, 10 or 100 ng/ml) and their combinations on cultured fragments of chicken ovarian follicular wall. Accumulation of PKA and CDC2 and secretion of progesterone (P4), testosterone (T), estradiol (E2) and arginine-vasotocin (AVT) were evaluated by using SDS-PAGE-Western blotting and RIA/EIA. IGF-II addition to culture medium stimulated T, E2 and AVT secretion and inhibited P4 secretion. These changes were associated with an increase in PKA and a decrease in CDC2 accumulation. The PKA blocker KT5720, when given alone, increased accumulation of PKA and secretion of T and E2, but not AVT and inhibited P4 secretion. The PKA blocker also prevented and even reversed the effects of IGF-II on PKA and steroid hormones secretion, but enhanced the action of IGF-II on AVT. The inhibitor of CDC2, olomoucine, when given alone, suppressed the expression of CDC2 and the secretion of P4 and AVT (but not T and E2). When given together with IGF-II, it augmented IGF-II-induced suppression of CDC2 and reversed the effects of IGF-II on P4 (but not on T, E2 or AVT). These observations demonstrate the involvement of PKA, CDC2 and IGF-II in regulating the secretory activity of avian ovarian cells. Our data also suggest the involvement of PKA in the mediation of IGF-II effects on P4, T, E2 and AVT secretion. CDC2 can mediate the effects of IGF-II on ovarian P4 secretion but not on other hormones.
Collapse
Affiliation(s)
- A V Sirotkin
- Research Institute of Animal Production, Hlohovská 2, 949 92 Nitra, Slovakia.
| | | |
Collapse
|