1
|
Chen L, Ruan Y, Ni L, Wang G, Gao Y, Zhang J, Li D, Xu H. KISS-1 knockdown inhibits cell growth, migration, and invasion in HTR-8/SVneo cells by regulating the GRP54-mediated PI3K/AKT signaling pathway. Autoimmunity 2024; 57:2297564. [PMID: 38155490 DOI: 10.1080/08916934.2023.2297564] [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: 05/30/2023] [Accepted: 12/14/2023] [Indexed: 12/30/2023]
Abstract
Recurrent spontaneous abortions (RSA) affect reproductive health and increase the risk of subsequent abortions. To investigate the role of KISS-1/GPR-54 signaling in RSA progression. Villus tissue was collected from RSA patients, and human trophoblastic HTR-8/SVneo cells were used. KISS-1 and GRP54 levels were detected using RT-qPCR and immunohistochemistry. Western blotting was performed to analyze ZO-1 and ZEB1 levels. Cell proliferation was determined via CCK-8 and cell clone formation assays. Transwell assays were performed to assess cell migration and invasion abilities. KISS-1 was down-regulated in the villus tissues of RSA patients. KISS-1 overexpression dramatically inhibited trophoblast proliferation, migration, and invasion. Mechanistically, ZEB1 expression was down-regulated, whereas ZO-1 expression was up-regulated, after KISS-1 overexpression. GPR54 silencing neutralized the effect of KISS-1 in HTR-8/SVneo cells. Additionally, KISS-1 overexpression inactivated the PI3K/AKT signaling pathway through GRP54. The KISS-1/GPR-54 signaling axis regulates RSA progression by regulating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
| | | | - Liping Ni
- Hangzhou Women's Hospital, Hangzhou, China
| | | | - Yajuan Gao
- Hangzhou Women's Hospital, Hangzhou, China
| | | | | | - Haiou Xu
- Hangzhou Women's Hospital, Hangzhou, China
| |
Collapse
|
2
|
Wang C, Xiong S, Hu S, Yang L, Huang Y, Chen H, Xu B, Xiao T, Liu Q. Genome-wide identification of Gα family in grass carp (Ctenopharyngodon idella) and reproductive regulation functional characteristics of Cignaq. BMC Genomics 2024; 25:800. [PMID: 39182029 PMCID: PMC11344465 DOI: 10.1186/s12864-024-10717-0] [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: 11/27/2023] [Accepted: 08/16/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND The Gα family plays a crucial role in the complex reproductive regulatory network of teleosts. However, the characterization and function of Gα family members, especially Gαq, remain poorly understood in teleosts. To analyze the characterization, expression, and function of grass carp (Ctenopharyngodon idella) Gαq, we identified the Gα family members in grass carp genome, and analyzed the expression, distribution, and signal transduction of Gαq/gnaq. We also explored the role of Gαq in the reproductive regulation of grass carp. RESULTS Our results showed that the grass carp genome contains 27 Gα genes with 46 isoforms, which are divided into four subfamilies: Gαs, Gαi/o, Gαq/11, and Gα12/13. The expression level of Cignaq in the testis was the highest and significantly higher than in other tissues, followed by the hypothalamus and brain. The luteinizing hormone receptor (LHR) was mainly localized to the nucleus in grass carp oocytes, with signals also present in follicular cells. In contrast, Gαq signal was mainly found in the cytoplasm of oocytes, with no signal in follicular cells. In the testis, Gαq and LHR were co-localized in the cytoplasm. Furthermore, the grass carp Gαq recombinant protein significantly promoted Cipgr expression. CONCLUSIONS These results provided preliminary evidence for understanding the role of Gαq in the reproductive regulation of teleosts.
Collapse
Affiliation(s)
- Chong Wang
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China
| | - Shuting Xiong
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China
| | - Shitao Hu
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China
| | - Le Yang
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China
| | - Yuhong Huang
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China
| | - Haitai Chen
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China
| | - Baohong Xu
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China
| | - Tiaoyi Xiao
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China.
| | - Qiaolin Liu
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China.
| |
Collapse
|
3
|
Meyer Z, Soukup ST, Lubs A, Ohde D, Walz C, Schoen J, Willenberg HS, Hoeflich A, Brenmoehl J. Impact of Dietary Isoflavones in Standard Chow on Reproductive Development in Juvenile and Adult Female Mice with Different Metabolic Phenotypes. Nutrients 2024; 16:2697. [PMID: 39203833 PMCID: PMC11357413 DOI: 10.3390/nu16162697] [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: 07/03/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
Two factors influencing female reproduction have been repeatedly studied in different animal species and humans, namely, 1. secondary plant compounds, especially phytoestrogens (mainly isoflavones (IFs)), and 2. the physical constitution/metabolic phenotype (e.g., obesity). So far, these research results have only been considered separately. In this study, we investigated the influence on reproduction of both phytochemicals, mainly dietary IFs, and the metabolic phenotype represented by three mouse models considered as three distinct genetic groups (a control group, a mouse model with high metabolic activity, and a mouse line with obese body weight). The IF content in different investigated standard chows with similar macronutrient profiles varied significantly (p < 0.005), leading to high mean total plasma IF levels of up to 5.8 µmol/L in juvenile and 6.7 µmol/L in adult female mice. Reproductive performance was only slightly affected; only an IF dose-dependent effect on gestation length was observed in all genetic groups, as well as an effect on pregnancy rate in obese mice. Dietary IF exposure, however, caused earlier onset of vaginal opening by 4-10 days in juvenile mice (p < 0.05), dependent on the genetic group, resulting in a slight acceleration of sexual maturation in the already precocious obese model and to a strong earlier maturation in the otherwise late-maturing sporty model, bred for high treadmill performance. Therefore, our results may help to draw the missing line between the effect of dietary secondary plant constituents, such as IFs, and metabolic phenotype on sexual development.
Collapse
Affiliation(s)
- Zianka Meyer
- Working Group Endocrinology of Farm Animals, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Sebastian T. Soukup
- Department of Safety and Quality of Fruit and Vegetables, Max Rubner-Institute, Federal Research Institute of Nutrition and Food, Haid-und-Neu-Straße 9, 76131 Karlsruhe, Germany
| | - Anna Lubs
- Working Group Cell Physiology & Reproduction, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Daniela Ohde
- Working Group Endocrinology of Farm Animals, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Christina Walz
- Working Group Endocrinology of Farm Animals, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Jennifer Schoen
- Working Group Cell Physiology & Reproduction, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
- Reproduction Biology Department, Leibniz Institute for Zoo and Wildlife Research IZW, Alfred-Kowalke-Straße 17, 10315 Berlin, Germany
| | - Holger S. Willenberg
- Center for Internal Medicine, Section of Endocrinology and Metabolic Diseases, University Medicine Rostock, Ernst-Heydemann-Str. 6, 18057 Rostock, Germany
| | - Andreas Hoeflich
- Working Group Endocrinology of Farm Animals, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Julia Brenmoehl
- Working Group Endocrinology of Farm Animals, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| |
Collapse
|
4
|
Hess RA, Park CJ, Soto S, Reinacher L, Oh JE, Bunnell M, Ko CJ. Male animal sterilization: history, current practices, and potential methods for replacing castration. Front Vet Sci 2024; 11:1409386. [PMID: 39027909 PMCID: PMC11255590 DOI: 10.3389/fvets.2024.1409386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
Sterilization and castration have been synonyms for thousands of years. Making an animal sterile meant to render them incapable of producing offspring. Castration or the physical removal of the testes was discovered to be the most simple but reliable method for managing reproduction and sexual behavior in the male. Today, there continues to be global utilization of castration in domestic animals. More than six hundred million pigs are castrated every year, and surgical removal of testes in dogs and cats is a routine practice in veterinary medicine. However, modern biological research has extended the meaning of sterilization to include methods that spare testis removal and involve a variety of options, from chemical castration and immunocastration to various methods of vasectomy. This review begins with the history of sterilization, showing a direct link between its practice in man and animals. Then, it traces the evolution of concepts for inducing sterility, where research has overlapped with basic studies of reproductive hormones and the discovery of testicular toxicants, some of which serve as sterilizing agents in rodent pests. Finally, the most recent efforts to use the immune system and gene editing to block hormonal stimulation of testis function are discussed. As we respond to the crisis of animal overpopulation and strive for better animal welfare, these novel methods provide optimism for replacing surgical castration in some species.
Collapse
Affiliation(s)
- Rex A. Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Epivara, Inc, Champaign, IL, United States
| | - Chan Jin Park
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Epivara, Inc, Champaign, IL, United States
| | | | | | - Ji-Eun Oh
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Mary Bunnell
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - CheMyong J. Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Epivara, Inc, Champaign, IL, United States
| |
Collapse
|
5
|
Samir H, Elfadadny A, Radwan F, El-Sherbiny HR, Swelum AA, Khalil WA, Watanabe G. Spatial local expressions of kisspeptin in the uterus and uterine tubes and its relationship to the reproductive potential in goats. Domest Anim Endocrinol 2024; 88:106850. [PMID: 38640803 DOI: 10.1016/j.domaniend.2024.106850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
Kisspeptins are neuropeptides encoded by the Kiss1 gene that was discovered as a metastasis suppressor gene in melanoma and breast cancer. Kisspeptin has pivotal functions for gonadotropin-releasing hormone secretion and plays integrated roles in the hypothalamic-pituitary-gonadal axis. However, little is known about the peripheral expression of kisspeptin in ruminants, especially in the female reproductive tract. Here, the objectives of the current study were to investigate the spatial localization of kisspeptin and mRNA expression of Kiss1 and its receptor (Kiss1r) in the fallopian tubes (FT) and uterus of goats at varied reproductive activity (cyclic versus true anoestrous goats, n=6, each). Specimens of the uterus and FT were collected and fixed using paraformaldehyde to investigate the localizations of kisspeptin in the selected tissues by immunohistochemistry. Another set of samples was snape-frozen to identify the expressions of mRNAs encoding Kiss1 and Kiss1r using real-time PCR. Results revealed immunolocalizations of kisspeptin in the uterus and the FT. The staining of kisspeptin was found mainly in the mucosal epithelium of the uterus the FT, and the endometrial glands. Very intense staining of kisspeptin was found in the uterine and FT specimens in the true anoestrous goats compared to that in cyclic ones. The expression of mRNA encoding Kiss1 gene was significantly higher in the uterine specimen of cyclic goats (1.00±0.09) compared to that in the true anoestrous goats (0.62±0.08) (P ˂0.05), while the expression of mRNA encoding Kiss1r was significantly (P ˂0.001) higher in the uterine tissues of true anoestrous goats (1.78±0.17) compared to that in cyclic ones (1.00±0.11). In conclusion, immunohistochemical localization of kisspeptin and the expression of mRNA encoding Kiss1/Kiss1r revealed spatial changes in the uterus and FT of goats according to the reproductive potential of goats (cyclic versus true anoestrous goats). However, the definitive local role of kisspeptin in the uterus and FT need further investigation.
Collapse
Affiliation(s)
- Haney Samir
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt; Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo 183-8509, Japan.
| | - Ahmed Elfadadny
- Department of Animal Medicine, Faculty of Veterinary Medicine, Damanhour University, Damanhour, El-Beheira 22511, Egypt
| | - Faten Radwan
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo 183-8509, Japan; Veterinarian graduated from the Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt
| | - Hossam R El-Sherbiny
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Ayman A Swelum
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Wael A Khalil
- Department of Animal Production, Faculty of Agriculture, Mansoura University, Mansoura 35516, Egypt
| | - Gen Watanabe
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo 183-8509, Japan
| |
Collapse
|
6
|
Kovács A, Szabó E, László K, Kertes E, Zagorácz O, Mintál K, Tóth A, Gálosi R, Berta B, Lénárd L, Hormay E, László B, Zelena D, Tóth ZE. Brain RFamide Neuropeptides in Stress-Related Psychopathologies. Cells 2024; 13:1097. [PMID: 38994950 PMCID: PMC11240450 DOI: 10.3390/cells13131097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/13/2024] Open
Abstract
The RFamide peptide family is a group of proteins that share a common C-terminal arginine-phenylalanine-amide motif. To date, the family comprises five groups in mammals: neuropeptide FF, LPXRFamides/RFamide-related peptides, prolactin releasing peptide, QRFP, and kisspeptins. Different RFamide peptides have their own cognate receptors and are produced by different cell populations, although they all can also bind to neuropeptide FF receptors with different affinities. RFamide peptides function in the brain as neuropeptides regulating key aspects of homeostasis such as energy balance, reproduction, and cardiovascular function. Furthermore, they are involved in the organization of the stress response including modulation of pain. Considering the interaction between stress and various parameters of homeostasis, the role of RFamide peptides may be critical in the development of stress-related neuropathologies. This review will therefore focus on the role of RFamide peptides as possible key hubs in stress and stress-related psychopathologies. The neurotransmitter coexpression profile of RFamide-producing cells is also discussed, highlighting its potential functional significance. The development of novel pharmaceutical agents for the treatment of stress-related disorders is an ongoing need. Thus, the importance of RFamide research is underlined by the emergence of peptidergic and G-protein coupled receptor-based therapeutic targets in the pharmaceutical industry.
Collapse
Affiliation(s)
- Anita Kovács
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Evelin Szabó
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Kristóf László
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Erika Kertes
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Olga Zagorácz
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Kitti Mintál
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Attila Tóth
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Rita Gálosi
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Bea Berta
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - László Lénárd
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Edina Hormay
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Bettina László
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Dóra Zelena
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Zsuzsanna E. Tóth
- Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary
| |
Collapse
|
7
|
Nakajo M, Kanda S, Oka Y. Involvement of the kisspeptin system in regulation of sexual behaviors in medaka. iScience 2024; 27:108971. [PMID: 38333699 PMCID: PMC10850746 DOI: 10.1016/j.isci.2024.108971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 11/09/2023] [Accepted: 01/16/2024] [Indexed: 02/10/2024] Open
Abstract
In mammals, kisspeptin (Kiss1) neurons are generally considered as a sex steroid-dependent key regulator of hypothalamic-pituitary-gonadal (HPG) axis. In contrast, previous studies in non-mammalian species, especially in teleosts, propose that Kiss1 is not directly involved in the HPG axis regulation, which suggests some sex-steroid-dependent functions of kisspeptin(s) other than the HPG axis regulation in non-mammals. Here, we used knockout (KO) medaka of kisspeptin receptor-coding genes (gpr54-1 and gpr54-2) and examined possible roles of kisspeptin in the regulation of sexual behaviors. We found that the KO pairs of gpr54-1, but not gpr54-2, spawned fewer eggs and exhibited delayed spawning than wild type pairs. Detailed behavior analysis suggested that the KO females are responsible for the delayed spawning and that the KO males showed hyper-motivation for courtship. Taken together, the present finding suggests that one of the reproductive-state-dependent functions of the Kiss1 may be the control of successful sexual behaviors.
Collapse
Affiliation(s)
- Mikoto Nakajo
- Department of Physiology, Division of Life Sciences, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Shinji Kanda
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Yoshitaka Oka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| |
Collapse
|
8
|
Li Z, Yang X, Fu R, Wu Z, Xu S, Jiao J, Qian M, Zhang L, Wu C, Xie T, Yao J, Wu Z, Li W, Ma G, You Y, Chen Y, Zhang HK, Cheng Y, Tang X, Wu P, Lian G, Wei H, Zhao J, Xu J, Ai L, Siwko S, Wang Y, Ding J, Song G, Luo J, Liu M, Xiao J. Kisspeptin-10 binding to Gpr54 in osteoclasts prevents bone loss by activating Dusp18-mediated dephosphorylation of Src. Nat Commun 2024; 15:1300. [PMID: 38346942 PMCID: PMC10861593 DOI: 10.1038/s41467-024-44852-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/05/2024] [Indexed: 02/15/2024] Open
Abstract
Osteoclasts are over-activated as we age, which results in bone loss. Src deficiency in mice leads to severe osteopetrosis due to a functional defect in osteoclasts, indicating that Src function is essential in osteoclasts. G-protein-coupled receptors (GPCRs) are the targets for ∼35% of approved drugs but it is still unclear how GPCRs regulate Src kinase activity. Here, we reveal that GPR54 activation by its natural ligand Kisspeptin-10 (Kp-10) causes Dusp18 to dephosphorylate Src at Tyr 416. Mechanistically, Gpr54 recruits both active Src and the Dusp18 phosphatase at its proline/arginine-rich motif in its C terminus. We show that Kp-10 binding to Gpr54 leads to the up-regulation of Dusp18. Kiss1, Gpr54 and Dusp18 knockout mice all exhibit osteoclast hyperactivation and bone loss, and Kp-10 abrogated bone loss by suppressing osteoclast activity in vivo. Therefore, Kp-10/Gpr54 is a promising therapeutic target to abrogate bone resorption by Dusp18-mediated Src dephosphorylation.
Collapse
Affiliation(s)
- Zhenxi Li
- Institute of Orthopedic Biomedical and Device Innovation, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China.
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| | - Xinghai Yang
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Ruifeng Fu
- Institute of Orthopedic Biomedical and Device Innovation, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Zhipeng Wu
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Shengzhao Xu
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jian Jiao
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Ming Qian
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Long Zhang
- Institute of Orthopedic Biomedical and Device Innovation, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Chunbiao Wu
- Institute of Orthopedic Biomedical and Device Innovation, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Tianying Xie
- Institute of Orthopedic Biomedical and Device Innovation, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Jiqiang Yao
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Zhixiang Wu
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Wenjun Li
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Guoli Ma
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yu You
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yihua Chen
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Han-Kun Zhang
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yiyun Cheng
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiaolong Tang
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Pengfei Wu
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Gewei Lian
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Haifeng Wei
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Jian Zhao
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Jianrong Xu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lianzhong Ai
- Institute of Orthopedic Biomedical and Device Innovation, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Stefan Siwko
- Department of Translational Medical Sciences, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, USA
| | - Yue Wang
- Shanghai Key Lab of Cell Engineering; Translational Medicine Research Center, Naval Medical University, Shanghai, 200433, China
| | - Jin Ding
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, 200433, China
| | - Gaojie Song
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, China
| | - Mingyao Liu
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jianru Xiao
- Institute of Orthopedic Biomedical and Device Innovation, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| |
Collapse
|
9
|
Szukiewicz D. Current Insights in Prolactin Signaling and Ovulatory Function. Int J Mol Sci 2024; 25:1976. [PMID: 38396659 PMCID: PMC10889014 DOI: 10.3390/ijms25041976] [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: 01/10/2024] [Revised: 01/31/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Prolactin (PRL) is a pleiotropic hormone released from lactotrophic cells of the anterior pituitary gland that also originates from extrapituitary sources and plays an important role in regulating lactation in mammals, as well as other actions. Acting in an endocrine and paracrine/autocrine manner, PRL regulates the hypothalamic-pituitary-ovarian axis, thus influencing the maturation of ovarian follicles and ovulation. This review provides a detailed discussion of the current knowledge on the role of PRL in the context of ovulation and ovulatory disorders, particularly with regard to hyperprolactinemia, which is one of the most common causes of infertility in women. Much attention has been given to the PRL structure and the PRL receptor (PRLR), as well as the diverse functions of PRLR signaling under normal and pathological conditions. The hormonal regulation of the menstrual cycle in connection with folliculogenesis and ovulation, as well as the current classifications of ovulation disorders, are also described. Finally, the state of knowledge regarding the importance of TIDA (tuberoinfundibular dopamine), KNDγ (kisspeptin/neurokinin B/dynorphin), and GnRH (gonadotropin-releasing hormone) neurons in PRL- and kisspeptin (KP)-dependent regulation of the hypothalamic-pituitary-gonadal (HPG) axis in women is reviewed. Based on this review, a rationale for influencing PRL signaling pathways in therapeutic activities accompanying ovulation disorders is presented.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
10
|
Sahin Z, Aktas O, Kalkan OF, Cuce G, Alver A, Sahin E, Erdem S, Saglam N, Solak Gormus ZI, Kutlu S. Intracerebroventricular injection of kisspeptin in male rats activates hypothalamo-pituitary-gonadal axis, but not hypothalamo-pituitary-adrenal axis. J Recept Signal Transduct Res 2024; 44:19-26. [PMID: 38647103 DOI: 10.1080/10799893.2024.2333470] [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: 05/15/2023] [Accepted: 03/17/2024] [Indexed: 04/25/2024]
Abstract
Kisspeptin is an important hormone involved in the stimulation of the hypothalamo-pituitary gonadal (HPG) axis. The HPG axis can be suppressed in certain conditions such as stress, which gives rise to the activation of the hypothalamo-pituitary-adrenal (HPA) axis. However, the physiological role of kisspeptin in the interaction of HPG and HPA axis is not fully understood yet. This study was conducted to investigate the possible effects of central kisspeptin injection on HPG axis as well as HPA axis activity. Adult male Wistar rats were randomly divided into seven groups as followed: sham (control), kisspeptin (50 pmol), P234 (1 nmol), kisspeptin + p234, kisspeptin + antalarmin (0.1 μg), kisspeptin + astressin 2B (1 μg), and kisspeptin + atosiban (300 ng/rat) (n = 10 each group). At the end of the experiments, the hypothalamus, pituitary, and serum samples of the rats were collected. There was no significant difference in corticotropic-releasing hormone immunoreactivity in the paraventricular nucleus of the hypothalamus, serum adrenocorticotropic hormone, and corticosterone levels among all groups. Moreover, no significant difference was detected in pituitary oxytocin level. Serum follicle-stimulating hormone and luteinizing hormone levels of the kisspeptin, kisspeptin + antalarmin, and kisspeptin + astressin 2B groups were significantly higher than the control group. Serum testosterone levels were significantly higher in the kisspeptin kisspeptin + antalarmin, kisspeptin + astressin 2B, and kisspeptin + atosiban groups compared to the control group. Our findings suggest that central kisspeptin injection causes activation in the HPG axis, but not the HPA axis in male rats.
Collapse
Affiliation(s)
- Zafer Sahin
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Osman Aktas
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Omer Faruk Kalkan
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Gokhan Cuce
- Department of Histology and Embryology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Ahmet Alver
- Department of Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Elif Sahin
- Department of Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Seniz Erdem
- Department of Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Neslihan Saglam
- Department of Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | | | - Selim Kutlu
- Department of Physiology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
11
|
Patel B, Koysombat K, Mills EG, Tsoutsouki J, Comninos AN, Abbara A, Dhillo WS. The Emerging Therapeutic Potential of Kisspeptin and Neurokinin B. Endocr Rev 2024; 45:30-68. [PMID: 37467734 PMCID: PMC10765167 DOI: 10.1210/endrev/bnad023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/13/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023]
Abstract
Kisspeptin (KP) and neurokinin B (NKB) are neuropeptides that govern the reproductive endocrine axis through regulating hypothalamic gonadotropin-releasing hormone (GnRH) neuronal activity and pulsatile GnRH secretion. Their critical role in reproductive health was first identified after inactivating variants in genes encoding for KP or NKB signaling were shown to result in congenital hypogonadotropic hypogonadism and a failure of pubertal development. Over the past 2 decades since their discovery, a wealth of evidence from both basic and translational research has laid the foundation for potential therapeutic applications. Beyond KP's function in the hypothalamus, it is also expressed in the placenta, liver, pancreas, adipose tissue, bone, and limbic regions, giving rise to several avenues of research for use in the diagnosis and treatment of pregnancy, metabolic, liver, bone, and behavioral disorders. The role played by NKB in stimulating the hypothalamic thermoregulatory center to mediate menopausal hot flashes has led to the development of medications that antagonize its action as a novel nonsteroidal therapeutic agent for this indication. Furthermore, the ability of NKB antagonism to partially suppress (but not abolish) the reproductive endocrine axis has supported its potential use for the treatment of various reproductive disorders including polycystic ovary syndrome, uterine fibroids, and endometriosis. This review will provide a comprehensive up-to-date overview of the preclinical and clinical data that have paved the way for the development of diagnostic and therapeutic applications of KP and NKB.
Collapse
Affiliation(s)
- Bijal Patel
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
| | - Kanyada Koysombat
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Edouard G Mills
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Jovanna Tsoutsouki
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
| | - Alexander N Comninos
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Ali Abbara
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Waljit S Dhillo
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| |
Collapse
|
12
|
Uenoyama Y, Tsukamura H. KNDy neurones and GnRH/LH pulse generation: Current understanding and future aspects. J Neuroendocrinol 2023; 35:e13285. [PMID: 37232103 DOI: 10.1111/jne.13285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/31/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023]
Abstract
Uncovering the central mechanism underlying mammalian reproduction is warranted to develop new therapeutic approaches for reproductive disorders in humans and domestic animals. The present study focused on the role of arcuate kisspeptin neurones (also known as KNDy neurones) as an intrinsic gonadotropin-releasing hormone (GnRH) pulse generator, which plays a fundamental role in mammalian reproduction via the stimulation of pituitary gonadotropin synthesis and release and thereby in gametogenesis and steroidogenesis in the gonads of mammals. We also discuss the mechanism that inhibits pulsatile GnRH/gonadotropin release under a negative energy balance, considering that reproductive disorders often occur during malnutrition in humans and livestock.
Collapse
Affiliation(s)
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Nagoya University, Nagoya, Japan
| |
Collapse
|
13
|
Venkata Ratna S, Prasada Rao T, Pathipati D, Chaitanya Kumar TV, Rambabu Naik B, Siva Kumar AVN. Kisspeptin promotes follicular development through its effects on modulation of P450 aromatase expression and steroidogenesis in sheep ovarian follicles. Reprod Domest Anim 2023; 58:1270-1278. [PMID: 37448136 DOI: 10.1111/rda.14430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/22/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023]
Abstract
The present study was conducted to ascertain whether the role of kisspeptin in promoting in vitro development of preantral follicles was through the regulation of P450 aromatase gene expression and steroidogenesis in sheep. Accordingly, the cumulus cells and oocytes were collected from different development stages of preantral follicles grown in vivo and cultured in vitro in TCM199B (Group I), TCM199B + KP (10 μg/mL) (Group II) and Standard medium + KP (10 μg/mL). To measure the steroid (Estradiol-17β; E2 and Progesterone; P4 ) synthesis through ELISA, spent culture medium was collected separately from the same in vitro groups. E2 synthesis in the spent medium collected from all the three groups showed an increasing trend from PFs' exposed to respective culture media for 3 min to 2-day culture stage but decreased thereafter till 6-day culture stage. This is followed by a sharp increase in E2 concentration in the spent medium collected after in vitro maturation. However, P4 synthesis in group III followed increased pattern as the development progressed from PFs' exposed to culture medium for 3 min to in vitro maturation stage. The steroid production was observed at all stages of in vitro development in altered supplemented conditions. The steroid synthesis in the spent medium was highest in the 6 day cultured PFs' in Standard medium + KP matured in vitro for 24 h. Therefore, supplementation of kisspeptin along with other growth factors promoted steroid production in cultured preantral follicles far better than in other media.
Collapse
Affiliation(s)
- S Venkata Ratna
- Department of Veterinary Biochemistry, College of Veterinary Science, Sri Venkateswara Veterinary University, Tirupati, Andhra Pradesh, India
| | - T Prasada Rao
- Department of Veterinary Biochemistry, College of Veterinary Science, Sri Venkateswara Veterinary University, Proddatur, Andhra Pradesh, India
| | - Deepa Pathipati
- Department of Veterinary Physiology, College of Veterinary Science, Sri Venkateswara Veterinary University, Tirupati, Andhra Pradesh, India
| | - T V Chaitanya Kumar
- Department of Veterinary Biochemistry, College of Veterinary Science, Sri Venkateswara Veterinary University, Tirupati, Andhra Pradesh, India
| | - B Rambabu Naik
- Department of Veterinary Physiology, College of Veterinary Science, Sri Venkateswara Veterinary University, Tirupati, Andhra Pradesh, India
| | - A V N Siva Kumar
- Department of Veterinary Physiology, College of Veterinary Science, Sri Venkateswara Veterinary University, Tirupati, Andhra Pradesh, India
| |
Collapse
|
14
|
Uenoyama Y, Inoue N, Tsukamura H. Kisspeptin and lactational anestrus: Current understanding and future prospects. Peptides 2023; 166:171026. [PMID: 37230188 DOI: 10.1016/j.peptides.2023.171026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 05/27/2023]
Abstract
Lactational anestrus, characterized by the suppression of pulsatile gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) release, would be a strategic adaptation to ensure survival by avoiding pregnancy during lactation in mammals. In the present article, we first provide a current understanding of the central regulation of reproduction in mammals, i.e., a fundamental role of arcuate kisspeptin neurons in mammalian reproduction by driving GnRH/LH pulses. Second, we discuss the central mechanism inhibiting arcuate Kiss1 (encoding kisspeptin) expression and GnRH/LH pulses during lactation with a focus on suckling stimulus, negative energy balance due to milk production, and the role of circulating estrogen in rats. We also discuss upper regulators that control arcuate kisspeptin neurons in rats during the early and late lactation periods based on the findings obtained by a lactating rat model. Finally, we discuss potential reproductive technology for the improvement of reproductive performance in milking cows.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan.
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
15
|
Park CJ, Minabe S, Hess RA, Lin PCP, Zhou S, Bashir ST, Barakat R, Gal A, Ko CJ. Single neonatal estrogen implant sterilizes female animals by decreasing hypothalamic KISS1 expression. Sci Rep 2023; 13:9627. [PMID: 37316510 DOI: 10.1038/s41598-023-36727-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/08/2023] [Indexed: 06/16/2023] Open
Abstract
Reproductive sterilization by surgical gonadectomy is strongly advocated to help manage animal populations, especially domesticated pets, and to prevent reproductive behaviors and diseases. This study explored the use of a single-injection method to induce sterility in female animals as an alternative to surgical ovariohysterectomy. The idea was based on our recent finding that repetitive daily injection of estrogen into neonatal rats disrupted hypothalamic expression of Kisspeptin (KISS1), the neuropeptide that triggers and regulates pulsatile secretion of GnRH. Neonatal female rats were dosed with estradiol benzoate (EB) either by daily injections for 11 days or by subcutaneous implantation of an EB-containing silicone capsule designed to release EB over 2-3 weeks. Rats treated by either method did not exhibit estrous cyclicity, were anovulatory, and became infertile. The EB-treated rats had fewer hypothalamic Kisspeptin neurons, but the GnRH-LH axis remained responsive to Kisspeptin stimulation. Because it would be desirable to use a biodegradable carrier that is also easier to handle, an injectable EB carrier was developed from PLGA microspheres to provide pharmacokinetics comparable to the EB-containing silicone capsule. A single neonatal injection of EB-microspheres at an equivalent dosage resulted in sterility in the female rat. In neonatal female Beagle dogs, implantation of an EB-containing silicone capsule also reduced ovarian follicle development and significantly inhibited KISS1 expression in the hypothalamus. None of the treatments produced any concerning health effects, other than infertility. Therefore, further development of this technology for sterilization in domestic female animals, such as dogs and cats is worthy of investigation.
Collapse
Affiliation(s)
- Chan Jin Park
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
- Epivara, Inc, Champaign, IL, 61820, USA
| | - Shiori Minabe
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, 028-3694, Japan
| | - Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
- Epivara, Inc, Champaign, IL, 61820, USA
| | - Po-Ching Patrick Lin
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | | | - Shah Tauseef Bashir
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | - Radwa Barakat
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
- Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Benha University, Qalyubia, 13518, Egypt
| | - Arnon Gal
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | - CheMyong Jay Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA.
- Epivara, Inc, Champaign, IL, 61820, USA.
| |
Collapse
|
16
|
Piet R. Circadian and kisspeptin regulation of the preovulatory surge. Peptides 2023; 163:170981. [PMID: 36842628 DOI: 10.1016/j.peptides.2023.170981] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 02/28/2023]
Abstract
Fertility in mammals is ultimately controlled by a small population of neurons - the gonadotropin-releasing hormone (GnRH) neurons - located in the ventral forebrain. GnRH neurons control gonadal function through the release of GnRH, which in turn stimulates the secretion of the anterior pituitary gonadotropins luteinizing hormone (LH) and follicle-stimulating hormone (FSH). In spontaneous ovulators, ovarian follicle maturation eventually stimulates, via sex steroid feedback, the mid-cycle surge in GnRH and LH secretion that causes ovulation. The GnRH/LH surge is initiated in many species just before the onset of activity through processes controlled by the central circadian clock, ensuring that the neuroendocrine control of ovulation and sex behavior are coordinated. This review aims to give an overview of anatomical and functional studies that collectively reveal some of the mechanisms through which the central circadian clock regulates GnRH neurons and their afferent circuits to drive the preovulatory surge.
Collapse
Affiliation(s)
- Richard Piet
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States.
| |
Collapse
|
17
|
Arslan M. Whole-genome sequencing and genomic analysis of Norduz goat (Capra hircus). Mamm Genome 2023:10.1007/s00335-023-09990-3. [PMID: 37004528 DOI: 10.1007/s00335-023-09990-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/21/2023] [Indexed: 04/04/2023]
Abstract
Artificial and natural selective breeding of goats has resulted in many different goat breeds all around the world. Norduz goat is one of these breeds, and it is a local goat breed of Turkey. The goats are favorable due to pre-weaning viability and reproduction values compared to the regional breeds. Development in sequencing technologies has let to understand huge genomic structures and complex phenotypes. Until now, such a comprehensive study has not been carried out to understand the genomic structure of the Norduz goats, yet. In the study, the next-generation sequencing was carried out to understand the genomic structure of Norduz goat. Real-time PCR was used to evaluate prominent CNVs in the Norduz goat individuals. Whole genome of the goat was constructed with an average of 33.1X coverage level. In the stringent filtering condition, 9,757,980 SNPs, 1,536,715 InDels, and 290 CNVs were detected in the Norduz goat genome. Functional analysis of high-impact SNP variations showed that the classical complement activation biological process was affected significantly in the goat. CNVs in the goat genome were found in genes related to defense against viruses, immune response, and cell membrane transporters. It was shown that GBP2, GBP5, and mammalian ortholog GBP1, which are INF-stimulated GTPases, were found to be high copy numbers in the goats. To conclude, genetic variations mainly in immunological response processes suggest that Norduz goat is an immunologically improved goat breed and natural selection could take an important role in the genetical improvements of the goats.
Collapse
Affiliation(s)
- Mevlüt Arslan
- Department of Genetics, Faculty of Veterinary Medicine, Van Yüzüncü Yıl University, Tuşba, 65080, Van, Turkey.
| |
Collapse
|
18
|
Gomes VCL, Beckers KF, Crissman KR, Landry CA, Flanagan JP, Awad RM, Piero FD, Liu CC, Sones JL. Sexually dimorphic pubertal development and adipose tissue kisspeptin dysregulation in the obese and preeclamptic-like BPH/5 mouse model offspring. Front Physiol 2023; 14:1070426. [PMID: 37035685 PMCID: PMC10076539 DOI: 10.3389/fphys.2023.1070426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/22/2023] [Indexed: 04/11/2023] Open
Abstract
Preeclampsia (PE) is a devastating hypertensive disorder of pregnancy closely linked to obesity. Long-term adverse outcomes may occur in offspring from preeclamptic pregnancies. Accordingly, sex-specific changes in pubertal development have been described in children from preeclamptic women, but the underlying mechanisms remain vastly unexplored. Features of PE are spontaneously recapitulated by the blood pressure high subline 5 (BPH/5) mouse model, including obesity and dyslipidemia in females before and throughout pregnancy, superimposed hypertension from late gestation to parturition and fetal growth restriction. A sexually dimorphic cardiometabolic phenotype has been described in BPH/5 offspring: while females are hyperphagic, hyperleptinemic, and overweight, with increased reproductive white adipose tissue (rWAT), males have similar food intake, serum leptin concentration, body weight and rWAT mass as controls. Herein, pubertal development and adiposity were further investigated in BPH/5 progeny. Precocious onset of puberty occurs in BPH/5 females, but not in male offspring. When reaching adulthood, the obese BPH/5 females display hypoestrogenism and hyperandrogenism. Kisspeptins, a family of peptides closely linked to reproduction and metabolism, have been previously shown to induce lipolysis and inhibit adipogenesis. Interestingly, expression of kisspeptins (Kiss1) and their cognate receptor (Kiss1r) in the adipose tissue seem to be modulated by the sex steroid hormone milieu. To further understand the metabolic-reproductive crosstalk in the BPH/5 offspring, Kiss1/Kiss1r expression in male and female rWAT were investigated. Downregulation of Kiss1/Kiss1r occurs in BPH/5 females when compared to males. Interestingly, dietary weight loss attenuated circulating testosterone concentration and rWAT Kiss1 downregulation in BPH/5 females. Altogether, the studies demonstrate reproductive abnormalities in offspring gestated in a PE-like uterus, which appear to be closely associated to the sexually dimorphic metabolic phenotype of the BPH/5 mouse model.
Collapse
Affiliation(s)
- Viviane C. L. Gomes
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Kalie F. Beckers
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Kassandra R. Crissman
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Camille A. Landry
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Juliet P. Flanagan
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Reham M. Awad
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Fabio Del Piero
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Chin-Chi Liu
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Jenny L. Sones
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
19
|
Prashar V, Arora T, Singh R, Sharma A, Parkash J. Hypothalamic Kisspeptin Neurons: Integral Elements of the GnRH System. Reprod Sci 2023; 30:802-822. [PMID: 35799018 DOI: 10.1007/s43032-022-01027-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/23/2022] [Indexed: 12/19/2022]
Abstract
Highly sophisticated and synchronized interactions of various cells and hormonal signals are required to make organisms competent for reproduction. GnRH neurons act as a common pathway for multiple cues for the onset of puberty and attaining reproductive function. GnRH is not directly receptive to most of the signals required for the GnRH secretion during the various phases of the ovarian cycle. Kisspeptin neurons of the hypothalamus convey these signals required for the synchronized release of the GnRH. The steroid-sensitive anteroventral periventricular nucleus (AVPV) kisspeptin and arcuate nucleus (ARC) KNDy neurons convey steroid feedback during the reproductive cycle necessary for GnRH surge and pulse, respectively. AVPV region kisspeptin neurons also communicate with nNOS synthesizing neurons and suprachiasmatic nucleus (SCN) neurons to coordinate the process of the ovarian cycle. Neurokinin B (NKB) and dynorphin play roles in the GnRH pulse stimulation and inhibition, respectively. The loss of NKB and kisspeptin function results in the development of neuroendocrine disorders such as hypogonadotropic hypogonadism (HH) and infertility. Ca2+ signaling is essential for GnRH pulse generation, which is propagated through gap junctions between astrocytes-KNDy and KNDy-KNDy neurons. Impaired functioning of KNDy neurons could develop the characteristics associated with polycystic ovarian syndrome (PCOS) in rodents. Kisspeptin-increased synthesis led to excessive secretion of the LH associated with PCOS. This review provides the latest insights and understanding into the role of the KNDy and AVPV/POA kisspeptin neurons in GnRH secretion and PCOS.
Collapse
Affiliation(s)
- Vikash Prashar
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Tania Arora
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Randeep Singh
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Arti Sharma
- Department of Computational Sciences, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Jyoti Parkash
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India.
| |
Collapse
|
20
|
Medeiros LDS, Rodrigues PDS, Santos DNL, Silva-Sampaio AC, Kirsten TB, Suffredini IB, Coque ADC, da Silva RA, Bernardi MM. Prenatal restraint stress downregulates the hypothalamic kisspeptidergic system transcripts genes, reduces the estrogen plasma levels, delayed the onset of puberty, and reduced the sexual behavior intensity in female rats. Physiol Behav 2023; 260:114055. [PMID: 36563733 DOI: 10.1016/j.physbeh.2022.114055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 10/28/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
AIMS This study investigated the possible relationships between the expression of the Kiss1 and Gpr54 gene expressions and the pituitary-gonadal hormones with the female onset of puberty and sexual behavior. The Kiss1 and Gpr54 gene expressions were examined because they are critical to controlling the hypothalamic activation of GnRH neurons and, in turn, the pituitary-gonadal hormones related to the early onset of puberty and sexual behavior. Further, it was evaluated that the pituitary and gonadal hormones involved in the vaginal opening and the expression of sexual behavior. METHODS Pregnant rats exposed to PRS from gestation days 17 to 20 were evaluated for maternal and open-field behaviors. The maternal behavior was analyzed because it may alter brain sexual organization affecting the pups development. It was observed in female pups the physical and development and, in adult age, the open-field behavior, the anxiety-like behavior, the estrous cycle, the sexual behavior, the serum FSH, LH, estrogen, progesterone, and testosterone levels, and the gene expression of kisspeptin protein (Kiss1) and Gpr54 in the hypothalamus. RESULTS the maternal and open-field behaviors were unaffected. In the F1 generation, PRS reduced weight at weaning, delayed the day of the vaginal opening and reduced the intensity of lordosis, the estrogen levels, and the Kiss1 and Gpr54 gene expression. These effects were attributed to hypothalamic kisspeptidergic system downregulation of transcripts genes and the reduced estrogen levels affected by the PRS.
Collapse
Affiliation(s)
- Loren da Silva Medeiros
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Paula da Silva Rodrigues
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Daniel Nascimento Lago Santos
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Ana Claudia Silva-Sampaio
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Thiago Berti Kirsten
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Ivana Barbosa Suffredini
- Núcleo de Pesquisas em Biodiversidade, Laboratório de Extração, Universidade Paulista - UNIP, Av. Paulista, 900, São Paulo, SP 01310-100, Brazil
| | - Alex de Camargo Coque
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Rodrigo Augusto da Silva
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil; School of Dentistry, Graduate Program in Health Sciences, University of Taubaté, Rua dos Operários, 9, Taubaté, SP 12020-340, Brazil
| | - Maria Martha Bernardi
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil.
| |
Collapse
|
21
|
Flórez JM, Martins K, Solin S, Bostrom JR, Rodríguez-Villamil P, Ongaratto F, Larson SA, Ganbaatar U, Coutts AW, Kern D, Murphy TW, Kim ES, Carlson DF, Huisman A, Sonstegard TS, Lents CA. CRISPR/Cas9-editing of KISS1 to generate pigs with hypogonadotropic hypogonadism as a castration free trait. Front Genet 2023; 13:1078991. [PMID: 36685939 PMCID: PMC9854396 DOI: 10.3389/fgene.2022.1078991] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/05/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction: Most male pigs are surgically castrated to avoid puberty-derived boar taint and aggressiveness. However, this surgical intervention represents a welfare concern in swine production. Disrupting porcine KISS1 is hypothesized to delay or abolish puberty by inducing variable hypogonadotropism and thus preventing the need for castration. Methods: To test this hypothesis, we generated the first KISS1-edited large animal using CRISPR/Cas9-ribonucleoproteins and single-stranded donor oligonucleotides. The targeted region preceded the sequence encoding a conserved core motif of kisspeptin. Genome editors were intracytoplasmically injected into 684 swine zygotes and transferred to 19 hormonally synchronized surrogate sows. In nine litters, 49 American Yorkshire and 20 Duroc liveborn piglets were naturally farrowed. Results: Thirty-five of these pigs bore KISS1-disruptive alleles ranging in frequency from 5% to 97% and did not phenotypically differ from their wild-type counterparts. In contrast, four KISS1-edited pigs (two boars and two gilts) with disruptive allele frequencies of 96% and 100% demonstrated full hypogonadotropism, infantile reproductive tracts, and failed to reach sexual maturity. Change in body weight during development was unaffected by editing KISS1. Founder pigs partially carrying KISS1-disruptive alleles were bred resulting in a total of 53 KISS1 +/+, 60 KISS1 +/-, and 34 KISS1 -/- F1 liveborn piglets, confirming germline transmission. Discussion: Results demonstrate that a high proportion of KISS1 alleles in pigs must be disrupted before variation in gonadotropin secretion is observed, suggesting that even a small amount of kisspeptin ligand is sufficient to confer proper sexual development and puberty in pigs. Follow-on studies will evaluate fertility restoration in KISS1 KO breeding stock to fully realize the potential of KISS1 gene edits to eliminate the need for surgical castration.
Collapse
Affiliation(s)
- Julio M. Flórez
- Acceligen Inc., Eagan, MN, United States,Department of Preventive Veterinary Medicine and Animal Reproduction, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal, Brazil
| | | | - Staci Solin
- Recombinetics Inc., Eagan, MN, United States
| | | | | | | | | | | | | | - Doug Kern
- Recombinetics Inc., Eagan, MN, United States
| | - Thomas W. Murphy
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE, United States
| | | | | | - Abe Huisman
- Hypor, Hendrix Genetics, Boxmeer, Netherlands
| | - Tad S. Sonstegard
- Acceligen Inc., Eagan, MN, United States,*Correspondence: Tad S. Sonstegard,
| | - Clay A. Lents
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE, United States
| |
Collapse
|
22
|
Bulut F, Kacar E, Bilgin B, Hekim MG, Keleştemur MM, Sahin Z, Ayar A, Ozcan M. Crosstalk between kisspeptin and gonadotropin-inhibitory hormone in the silence of puberty: preclinical evidence from a calcium signaling study. J Recept Signal Transduct Res 2022; 42:608-613. [PMID: 36137227 DOI: 10.1080/10799893.2022.2125014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Kisspeptin and gonadotropin-inhibitory hormone (GnIH) are among suggested neuroendocrine modulators of reproductive function. Intracellular calcium signaling is a critical component in the regulation of a variety of physiological and pathological processes including neurotransmitter release, and, therefore, can be used as signaling indicator for investigating the involvement of kisspeptin, GnIH, and gonadotropin-releasing hormone (GnRH) release. Hence, this study investigated the effects of kisspeptin and GnIH on calcium signaling using immortalized hypothalamic cells (rHypoE-8) as a model. Kisspeptin neurons were loaded with the ratiometric calcium dye (Fura-2 AM, 1 μmol) and intracellular free calcium ([Ca2+]i) responses were quantified using digital fluorescence imaging system. Kisspeptin-10 (100, 300, and 1000 nM) caused a significant increase in [Ca2+]i in rHypoE-8 cells (n = 58, n = 64, and n = 49, respectively, p < 0.001). The kisspeptin receptor antagonist, P234, inhibited the calcium responses to kisspeptin (p < 0.001, n = 32). GnIH (100 and 1000 nM), alone, did not cause any significant change in the mean basal [Ca2+]i levels in kisspeptin cells, but GnIH attenuated the kisspeptin-evoked [Ca2+]i transients (n = 47, p < 0.001). This novel findings of [Ca2+]i signaling in in vitro setting implicate that kisspeptin and GnIH may exert their effects on hypothalamus-pituitary-gonadal (HPG) axis by modulating kisspeptin neurons. These results also implicate that kisspeptin neurons may have an autocrine regulation.
Collapse
Affiliation(s)
- Ferah Bulut
- Department of Biophysics, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Emine Kacar
- Department of Physiology, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Batuhan Bilgin
- Department of Biophysics, Faculty of Medicine, Firat University, Elazig, Turkey
| | | | | | - Zafer Sahin
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Ahmet Ayar
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Mete Ozcan
- Department of Biophysics, Faculty of Medicine, Firat University, Elazig, Turkey
| |
Collapse
|
23
|
Coutinho EA, Esparza LA, Hudson AD, Rizo N, Steffen P, Kauffman AS. Conditional Deletion of KOR (Oprk1) in Kisspeptin Cells Does Not Alter LH Pulses, Puberty, or Fertility in Mice. Endocrinology 2022; 163:6763672. [PMID: 36260530 DOI: 10.1210/endocr/bqac175] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Indexed: 01/26/2023]
Abstract
Classic pharmacological studies suggested that endogenous dynorphin-KOR signaling is important for reproductive neuroendocrine regulation. With the seminal discovery of an interconnected network of hypothalamic arcuate neurons co-expressing kisspeptin, neurokinin B, and dynorphin (KNDy neurons), the KNDy hypothesis was developed to explain how gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) pulses are generated. Key to this hypothesis is dynorphin released from KNDy neurons acting in a paracrine manner on other KNDy neurons via kappa opioid receptor (KOR) signaling to terminate neural "pulse" events. While in vitro evidence supports this aspect of the KNDy hypothesis, a direct in vivo test of the necessity of KOR signaling in kisspeptin neurons for proper LH secretion has been lacking. We therefore conditionally knocked out KOR selectively from kisspeptin neurons of male and female mice and tested numerous reproductive measures, including in vivo LH pulse secretion. Surprisingly, despite validating successful knockout of KOR in kisspeptin neurons, we found no significant effect of kisspeptin cell-specific deletion of KOR on any measure of puberty, LH pulse parameters, LH surges, follicle-stimulating hormone (FSH) levels, estrous cycles, or fertility. These outcomes suggest that the KNDy hypothesis, while sufficient normally, may not be the only neural mechanism for sculpting GnRH and LH pulses, supported by recent findings in humans and mice. Thus, besides normally acting via KOR in KNDy neurons, endogenous dynorphin and other opioids may, under some conditions, regulate LH and FSH secretion via KOR in non-kisspeptin cells or perhaps via non-KOR pathways. The current models for GnRH and LH pulse generation should be expanded to consider such alternate mechanisms.
Collapse
Affiliation(s)
- Eulalia A Coutinho
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Lourdes A Esparza
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Alexandra D Hudson
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Nathanael Rizo
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Paige Steffen
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Alexander S Kauffman
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
24
|
Park JS, Cheon YP, Choi D, Lee SH. Expression of Kisspeptin in the Adult Hamster Testis. Dev Reprod 2022; 26:107-115. [PMID: 36285151 PMCID: PMC9578319 DOI: 10.12717/dr.2022.26.3.107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/13/2022] [Accepted: 08/29/2022] [Indexed: 11/04/2022]
Abstract
Kisspeptins, products of KISS1 gene, are ligands of the G-protein coupled receptor (GPR54), and the kisspeptin-GPR54 signaling has an important role as an upstream regulator of gonadotropin releasing hormone (GnRH) neurons. Interestingly, extrahypothalamic expressions of kisspeptin/GPR-54 in gonads have been found in primates and experimental rodents such as rats and mice. Hamsters, another potent experimental rodent, also have a kisspeptin-GPR54 system in their ovaries. The presence of testicular kisspeptin-GPR54 system, however, remains to be solved. The present study was undertaken to determine whether the kisspeptin is expressed in hamster testis. To do this, reverse transcription-polymerase chain reactions (RT-PCRs) and immunohistochemistry (IHC) were employed. After the nest PCR, two cDNA products (320 and 280 bp, respectively) were detected by 3% agarose gel electrophoresis, and sequencing analysis revealed that the 320 bp product was correctly amplified from hamster kisspeptin cDNA. Modest immunoreactive (IR) kisspeptins were detected in Leydig-interstitial cells, and the weak signals were detected in germ cells, mostly in round spermatids and residual bodies of elongated spermatids. In the present study, we found the kisspeptin expression in the testis of Syrian hamster. Further studies on the local role(s) of testicular kisspeptin are expected for a better understanding the physiology of hamster testis, including photoperiodic gonadal regression specifically occurred in hamster gonads.
Collapse
Affiliation(s)
- Jin-Soo Park
- Department of Biotechnology, Sangmyung
University, Seoul 03016, Korea
| | - Yong-Pil Cheon
- Division of Developmental Biology and
Physiology, School of Biological Sciences and Chemistry, Sungshin
University, Seoul 02844, Korea
| | - Donchan Choi
- Department of Life Science, College of
Environmental Sciences, Yong-In University, Yongin
17092, Korea
| | - Sung-Ho Lee
- Department of Biotechnology, Sangmyung
University, Seoul 03016, Korea,Corresponding author Sung-Ho
Lee, Department of Biotechnology, Sangmyung, University, Seoul 03016, Korea,
Tel: +82-2-2287-5139, Fax:
+82-2-2287-0070, E-mail:
| |
Collapse
|
25
|
Masumi S, Lee EB, Dilower I, Upadhyaya S, Chakravarthi VP, Fields PE, Rumi MAK. The role of Kisspeptin signaling in Oocyte maturation. Front Endocrinol (Lausanne) 2022; 13:917464. [PMID: 36072937 PMCID: PMC9441556 DOI: 10.3389/fendo.2022.917464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022] Open
Abstract
Kisspeptins (KPs) secreted from the hypothalamic KP neurons act on KP receptors (KPRs) in gonadotropin (GPN) releasing hormone (GnRH) neurons to produce GnRH. GnRH acts on pituitary gonadotrophs to induce secretion of GPNs, namely follicle stimulating hormone (FSH) and luteinizing hormone (LH), which are essential for ovarian follicle development, oocyte maturation and ovulation. Thus, hypothalamic KPs regulate oocyte maturation indirectly through GPNs. KPs and KPRs are also expressed in the ovarian follicles across species. Recent studies demonstrated that intraovarian KPs also act directly on the KPRs expressed in oocytes to promote oocyte maturation and ovulation. In this review article, we have summarized published reports on the role of hypothalamic and ovarian KP-signaling in oocyte maturation. Gonadal steroid hormones regulate KP secretion from hypothalamic KP neurons, which in turn induces GPN secretion from the hypothalamic-pituitary (HP) axis. On the other hand, GPNs secreted from the HP axis act on the granulosa cells (GCs) and upregulate the expression of ovarian KPs. While KPs are expressed predominantly in the GCs, the KPRs are in the oocytes. Expression of KPs in the ovaries increases with the progression of the estrous cycle and peaks during the preovulatory GPN surge. Intrafollicular KP levels in the ovaries rise with the advancement of developmental stages. Moreover, loss of KPRs in oocytes in mice leads to failure of oocyte maturation and ovulation similar to that of premature ovarian insufficiency (POI). These findings suggest that GC-derived KPs may act on the KPRs in oocytes during their preovulatory maturation. In addition to the intraovarian role of KP-signaling in oocyte maturation, in vivo, a direct role of KP has been identified during in vitro maturation of sheep, porcine, and rat oocytes. KP-stimulation of rat oocytes, in vitro, resulted in Ca2+ release and activation of the mitogen-activated protein kinase, extracellular signal-regulated kinase 1 and 2. In vitro treatment of rat or porcine oocytes with KPs upregulated messenger RNA levels of the factors that favor oocyte maturation. In clinical trials, human KP-54 has also been administered successfully to patients undergoing assisted reproductive technologies (ARTs) for increasing oocyte maturation. Exogenous KPs can induce GPN secretion from hypothalamus; however, the possibility of direct KP action on the oocytes cannot be excluded. Understanding the direct in vivo and in vitro roles of KP-signaling in oocyte maturation will help in developing novel KP-based ARTs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - M. A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
26
|
Bo T, Liu M, Tang L, Lv J, Wen J, Wang D. Effects of High-Fat Diet During Childhood on Precocious Puberty and Gut Microbiota in Mice. Front Microbiol 2022; 13:930747. [PMID: 35910597 PMCID: PMC9329965 DOI: 10.3389/fmicb.2022.930747] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/27/2022] [Indexed: 12/02/2022] Open
Abstract
Precocious puberty mostly stems from endocrine disorders. However, more and more studies show that a high-fat diet (HFD) is closely related to precocious puberty, but its mechanism is unknown. Since gut microbiota is associated with hormone secretion and obesity, it inspires us to detect the mechanism of gut microbiota in triggering precocious puberty. The model of precocious puberty was established by feeding female mice with an HFD from 21 days old. After puberty, the serum hormone levels, gut microbiome sequencing, and metabolomics were collected. DNA was extracted from feces, and the V3–V4 region of the bacterial 16S rRNA gene was amplified, followed by microbial composition analysis. Subsequently, associations between precocious puberty and the microbiota were determined. We found that (1) HFD after weaning caused precocious puberty, increased serum estradiol, leptin, deoxycholic acid (DCA), and gonadotropin-releasing hormone (GnRH) in the hypothalamus; (2) Through correlation analysis, we found that GnRH was positively correlated with Desulfovibrio, Lachnoclostridium, GCA-900066575, Streptococcus, Anaerotruncus, and Bifidobacterium, suggesting that these bacteria may have a role in promoting sexual development. (3) “HFD-microbiota” transplantation promoted the precocious puberty of mice. (4) Estrogen changes the composition and proportion of gut microbiota and promotes precocious puberty. Therefore, the effect of HFD on precocious puberty is regulated by the interaction of gut microbiota and hormones.
Collapse
Affiliation(s)
- Tingbei Bo
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Min Liu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Liqiu Tang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Jinzhen Lv
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Jing Wen
- College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Dehua Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, Shandong University, Qingdao, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Dehua Wang, ;
| |
Collapse
|
27
|
Ma Y, Awe O, Radovick S, Yang X, Divall S, Wolfe A, Wu S. Lower FSH With Normal Fertility in Male Mice Lacking Gonadotroph Kisspeptin Receptor. Front Physiol 2022; 13:868593. [PMID: 35557961 PMCID: PMC9089166 DOI: 10.3389/fphys.2022.868593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/31/2022] [Indexed: 12/03/2022] Open
Abstract
The kisspeptin receptor, crucial for hypothalamic control of puberty and reproduction, is also present in the pituitary gland. Its role in the pituitary gland is not defined. Kisspeptin signaling via the Kiss1r could potentially regulate reproductive function at the level of pituitary gonadotrope. Using Cre/Lox technology, we deleted the Kiss1r gene in pituitary gonadotropes (PKiRKO). PKiRKO males have normal genital development (anogenital distance WT: 19.1 ± 0.4 vs. PKiRKO: 18.5 ± 0.4 mm), puberty onset, testes cell structure on gross histology, normal testes size, and fertility. PKiRKO males showed significantly decreased serum FSH levels compared to WT males (5.6 ± 1.9 vs. 10.2 ± 1.8 ng/ml) with comparable LH (1.1 ± 0.2 vs. 1.8 ± 0.4 ng/ml) and testosterone levels (351.8 ± 213.0 vs. 342.2 ± 183.0 ng/dl). PKiRKO females have normal puberty onset, cyclicity, LH and FSH levels and fertility. Overall, these findings indicate that absence of pituitary Kiss1r reduces FSH levels in male mice without affecting testis function. PKiRKO mice have normal reproductive function in both males and females.
Collapse
Affiliation(s)
- Yaping Ma
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Olubusayo Awe
- Department of Cellular and Molecular Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sally Radovick
- Department of Pediatrics, Rutgers University Medical School, New Brunswick, NJ, United States
| | - Xiaofeng Yang
- Department of Cardiovascular Sciences, Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Sara Divall
- Department of Pediatrics, University of Washington, Seattle's Children's Hospital, Seattle, United States
| | - Andrew Wolfe
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sheng Wu
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Cellular and Molecular Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Cardiovascular Sciences, Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
28
|
Zhang S, Yu F, Che A, Tan B, Huang C, Chen Y, Liu X, Huang Q, Zhang W, Ma C, Qian M, Liu M, Qin J, Du B. Neuroendocrine Regulation of Stress-Induced T Cell Dysfunction during Lung Cancer Immunosurveillance via the Kisspeptin/GPR54 Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104132. [PMID: 35224894 PMCID: PMC9069377 DOI: 10.1002/advs.202104132] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 02/10/2022] [Indexed: 06/01/2023]
Abstract
Emerging evidence suggests that physiological distress is highly correlated with cancer incidence and mortality. However, the mechanisms underlying psychological challenges-mediated tumor immune evasion are not systematically explored. Here, it is demonstrated that acute restraint (AR) increases the level of the plasma neuropeptide hormones, kisspeptin, and the expression levels of its receptor, Gpr54, in the hypothalamus, splenic and tumor-infiltrating T cells, suggesting a correlation between the neuroendocrine system and tumor microenvironment. Accordingly, administration of kisspeptin-10 significantly impairs T cell function, whereas knockout of Gpr54 in T cells inhibits lung tumor progression by suppressing T cell dysfunction and exhaustion with or without AR. In addition, Gpr54 defective OT-1 T cells show superior antitumor activity against OVA peptide-positive tumors. Mechanistically, ERK5-mediated NR4A1 activation is found to be essential for kisspeptin/GPR54-facilitated T cell dysfunction. Meanwhile, pharmacological inhibition of ERK5 signaling by XMD8-92 significantly reduces the tumor growth by enhancing CD8+ T cell antitumor function. Furthermore, depletion of GPR54 or ERK5 by CRISPR/Cas9 in CAR T cells intensifies the antitumor responses to both PSMA+ and CD19+ tumor cells, while eliminating T cell exhaustion. Taken together, these results indicate that kisspeptin/GPR54 signaling plays a nonredundant role in the stress-induced tumor immune evasion.
Collapse
Affiliation(s)
- Su Zhang
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Fangfei Yu
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Anran Che
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | | | - Chenshen Huang
- Department of General SurgeryTongji HospitalSchool of MedicineTongji UniversityShanghai200065China
| | - Yuxue Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Xiaohong Liu
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Qi Huang
- Department of General SurgeryTongji HospitalSchool of MedicineTongji UniversityShanghai200065China
| | - Wenying Zhang
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Chengbin Ma
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Min Qian
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Mingyao Liu
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Juliang Qin
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Bing Du
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| |
Collapse
|
29
|
Lee EB, Dilower I, Marsh CA, Wolfe MW, Masumi S, Upadhyaya S, Rumi MAK. Sexual Dimorphism in Kisspeptin Signaling. Cells 2022; 11:1146. [PMID: 35406710 PMCID: PMC8997554 DOI: 10.3390/cells11071146] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 02/05/2023] Open
Abstract
Kisspeptin (KP) and kisspeptin receptor (KPR) are essential for the onset of puberty, development of gonads, and maintenance of gonadal function in both males and females. Hypothalamic KPs and KPR display a high degree of sexual dimorphism in expression and function. KPs act on KPR in gonadotropin releasing hormone (GnRH) neurons and induce distinct patterns of GnRH secretion in males and females. GnRH acts on the anterior pituitary to secrete gonadotropins, which are required for steroidogenesis and gametogenesis in testes and ovaries. Gonadal steroid hormones in turn regulate the KP neurons. Gonadal hormones inhibit the KP neurons within the arcuate nucleus and generate pulsatile GnRH mediated gonadotropin (GPN) secretion in both sexes. However, the numbers of KP neurons in the anteroventral periventricular nucleus and preoptic area are greater in females, which release a large amount of KPs in response to a high estrogen level and induce the preovulatory GPN surge. In addition to the hypothalamus, KPs and KPR are also expressed in various extrahypothalamic tissues including the liver, pancreas, fat, and gonads. There is a remarkable difference in circulating KP levels between males and females. An increased level of KPs in females can be linked to increased numbers of KP neurons in female hypothalamus and more KP production in the ovaries and adipose tissues. Although the sexually dimorphic features are well characterized for hypothalamic KPs, very little is known about the extrahypothalamic KPs. This review article summarizes current knowledge regarding the sexual dimorphism in hypothalamic as well as extrahypothalamic KP and KPR system in primates and rodents.
Collapse
Affiliation(s)
- Eun Bee Lee
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Iman Dilower
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Courtney A. Marsh
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Michael W. Wolfe
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Saeed Masumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| | - Sameer Upadhyaya
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| | - Mohammad A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| |
Collapse
|
30
|
Hudson AD, Kauffman AS. Metabolic actions of kisspeptin signaling: Effects on body weight, energy expenditure, and feeding. Pharmacol Ther 2022; 231:107974. [PMID: 34530008 PMCID: PMC8884343 DOI: 10.1016/j.pharmthera.2021.107974] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 12/18/2022]
Abstract
Kisspeptin (encoded by the Kiss1 gene) and its receptor, KISS1R (encoded by the Kiss1r gene), have well-established roles in stimulating reproduction via central actions on reproductive neural circuits, but recent evidence suggests that kisspeptin signaling also influences metabolism and energy balance. Indeed, both Kiss1 and Kiss1r are expressed in many metabolically-relevant peripheral tissues, including both white and brown adipose tissue, the liver, and the pancreas, suggesting possible actions on these tissues or involvement in their physiology. In addition, there may be central actions of kisspeptin signaling, or factors co-released from kisspeptin neurons, that modulate metabolic, feeding, or thermoregulatory processes. Accumulating data from animal models suggests that kisspeptin signaling regulates a wide variety of metabolic parameters, including body weight and energy expenditure, adiposity and adipose tissue function, food intake, glucose metabolism, respiratory rates, locomotor activity, and thermoregulation. Herein, the current evidence for the involvement of kisspeptin signaling in each of these physiological parameters is reviewed, gaps in knowledge identified, and future avenues of important research highlighted. Collectively, the discussed findings highlight emerging non-reproductive actions of kisspeptin signaling in metabolism and energy balance, in addition to previously documented roles in reproductive control, but also emphasize the need for more research to resolve current controversies and uncover underlying molecular and physiological mechanisms.
Collapse
Affiliation(s)
- Alexandra D Hudson
- Dept. of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Alexander S Kauffman
- Dept. of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, United States of America.
| |
Collapse
|
31
|
Mele E, D’Auria R, Scafuro M, Marino M, Fasano S, Viggiano A, Pierantoni R, Santoro A, Meccariello R. Differential Expression of Kisspeptin System and Kisspeptin Receptor Trafficking during Spermatozoa Transit in the Epididymis. Genes (Basel) 2022; 13:genes13020295. [PMID: 35205340 PMCID: PMC8871750 DOI: 10.3390/genes13020295] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/24/2022] [Accepted: 01/29/2022] [Indexed: 02/04/2023] Open
Abstract
The hypothalamus–pituitary–testis axis controls the production of spermatozoa, and the kisspeptin system, comprising Kiss1 and Kiss1 receptor (Kiss1R), is the main central gatekeeper. The activity of the kisspeptin system also occurs in testis and spermatozoa, but currently the need of peripheral kisspeptin to produce gametes is not fully understood. Hence, we characterized kisspeptin system in rat spermatozoa and epididymis caput and cauda and analyzed the possible presence of Kiss1 in the epididymal fluid. The presence of Kiss1 and Kiss1R in spermatozoa collected from epididymis caput and cauda was evaluated by Western blot; significant high Kiss1 levels in the caput (p < 0.001 vs. cauda) and constant levels of Kiss1R proteins were observed. Immunofluorescence analysis revealed that the localization of Kiss1R in sperm head shifts from the posterior region in the epididymis caput to perforatorium in the epididymis cauda. In spermatozoa-free epididymis, Western blot revealed higher expression of Kiss1 and Kiss1R in caput (p < 0.05 vs. cauda). Moreover, immunohistochemistry revealed that Kiss1 and Kiss1R proteins were mainly localized in the secretory epithelial cell types and in contractile myoid cells, respectively. Finally, both dot blot and Elisa revealed the presence of Kiss1 in the epididymal fluid collected from epididymis cauda and caput, indicating that rat epididymis and spermatozoa possess a complete kisspeptin system. In conclusion, we reported for the first time in rodents Kiss1R trafficking in spermatozoa during the epididymis transit and Kiss1 measure in the epididymal fluid, thus suggesting a possible role for the system in spermatozoa maturation and storage within the epididymis.
Collapse
Affiliation(s)
- Elena Mele
- Department of Movement Sciences and Wellness, University of Naples Parthenope, Via Medina 40, 80133 Naples, Italy;
| | - Raffaella D’Auria
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Via S. Allende, 84081 Baronissi, Italy; (R.D.); (M.M.); (A.V.)
| | - Marika Scafuro
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Costantinopoli 16, 80138 Naples, Italy;
| | - Marianna Marino
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Via S. Allende, 84081 Baronissi, Italy; (R.D.); (M.M.); (A.V.)
| | - Silvia Fasano
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via Costantinpoli 16, 80138 Naples, Italy; (S.F.); (R.P.)
| | - Andrea Viggiano
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Via S. Allende, 84081 Baronissi, Italy; (R.D.); (M.M.); (A.V.)
| | - Riccardo Pierantoni
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via Costantinpoli 16, 80138 Naples, Italy; (S.F.); (R.P.)
| | - Antonietta Santoro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Via S. Allende, 84081 Baronissi, Italy; (R.D.); (M.M.); (A.V.)
- Correspondence: (A.S.); (R.M.)
| | - Rosaria Meccariello
- Department of Movement Sciences and Wellness, University of Naples Parthenope, Via Medina 40, 80133 Naples, Italy;
- Correspondence: (A.S.); (R.M.)
| |
Collapse
|
32
|
Gołyszny M, Obuchowicz E, Zieliński M. Neuropeptides as regulators of the hypothalamus-pituitary-gonadal (HPG) axis activity and their putative roles in stress-induced fertility disorders. Neuropeptides 2022; 91:102216. [PMID: 34974357 DOI: 10.1016/j.npep.2021.102216] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/21/2021] [Accepted: 10/05/2021] [Indexed: 11/24/2022]
Abstract
Neuropeptides being regulators of the hypothalamus-pituitary-adrenal (HPA) axis activity, also affect the function of the hypothalamus-pituitary-gonadal (HPG) axis by regulating gonadotrophin-releasing hormone (GnRH) secretion from hypothalamic neurons. Here, we review the available data on how neuropeptides affect HPG axis activity directly or indirectly via their influence on the HPA axis. The putative role of neuropeptides in stress-induced infertility, such as polycystic ovary syndrome, is also described. This review discusses both well-known neuropeptides (i.e., kisspeptin, Kp; oxytocin, OT; arginine-vasopressin, AVP) and more recently discovered peptides (i.e., relaxin-3, RLN-3; nesfatin-1, NEFA; phoenixin, PNX; spexin, SPX). For the first time, we present an up-to-date review of all published data regarding interactions between the aforementioned neuropeptide systems. The reviewed literature suggest new pathophysiological mechanisms leading to fertility disturbances that are induced by stress.
Collapse
Affiliation(s)
- Miłosz Gołyszny
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18 Street, 40-752 Katowice, Poland.
| | - Ewa Obuchowicz
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18 Street, 40-752 Katowice, Poland.
| | - Michał Zieliński
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18 Street, 40-752 Katowice, Poland.
| |
Collapse
|
33
|
Jeet V, Magotra A, Bangar YC, Kumar S, Garg AR, Yadav AS, Bahurupi P. Evaluation of candidate point mutation of Kisspeptin 1 gene associated with litter size in Indian Goat breeds and its effect on transcription factor binding sites. Domest Anim Endocrinol 2022; 78:106676. [PMID: 34626930 DOI: 10.1016/j.domaniend.2021.106676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 12/30/2022]
Abstract
Kisspeptin gene (Kiss1) has a significant role in reproductive processes in mammals. However, only little information is available about the association of Kiss1 gene with litter size in Indian goat breeds. Thus, blood samples from 285 randomly selected animals were collected for DNA isolation and SNP profiling. The PCR product of 242 bp size harboring g.2540C>T mutation of Kiss1 gene was digested with the restriction enzyme Sac1. Least squares analysis revealed that Barbari goats showed significantly higher average litter size (2.86±0.08) compared to Beetal, Sirohi and Sojat breeds (P < 0.01). SNP locus g.2540C>T of Kiss1 gene also showed significant effect on litter size (P < 0.01). Goats with Genotype CT (2.66 ± 0.07) and TT (2.67 ± 0.26) had significantly higher (P < 0.01) litter size than CC (1.50 ± 0.05). From the transcription factor binding site analysis, it was predicted that due to g.2540C>T SNP, both native and mutant variant forms coded for putative binding sites for different transcription Factor. Allele T had putative binding sites for the androgen receptor which plays a significant role in the signaling pathway involved in increase in ovulation rate; which consequently can have a tremendous effect on average litter size.
Collapse
Affiliation(s)
- Vikram Jeet
- Department of Animal Genetics and Breeding, Lala Lajpat Rai University of Veterinary and Animal sciences (LUVAS), Hisar, Haryana, India
| | - Ankit Magotra
- Department of Animal Genetics and Breeding, Lala Lajpat Rai University of Veterinary and Animal sciences (LUVAS), Hisar, Haryana, India.
| | - Y C Bangar
- Department of Animal Genetics and Breeding, Lala Lajpat Rai University of Veterinary and Animal sciences (LUVAS), Hisar, Haryana, India
| | - S Kumar
- Department of Livestock Farm Complex, Lala Lajpat Rai University of Veterinary and Animal sciences (LUVAS), Hisar, Haryana, India
| | - A R Garg
- Department of Animal Genetics and Breeding, Lala Lajpat Rai University of Veterinary and Animal sciences (LUVAS), Hisar, Haryana, India
| | - A S Yadav
- Department of Animal Genetics and Breeding, Lala Lajpat Rai University of Veterinary and Animal sciences (LUVAS), Hisar, Haryana, India
| | - P Bahurupi
- Department of Animal Genetics and Breeding, Lala Lajpat Rai University of Veterinary and Animal sciences (LUVAS), Hisar, Haryana, India
| |
Collapse
|
34
|
Campo A, Dufour S, Rousseau K. Tachykinins, new players in the control of reproduction and food intake: A comparative review in mammals and teleosts. Front Endocrinol (Lausanne) 2022; 13:1056939. [PMID: 36589829 PMCID: PMC9800884 DOI: 10.3389/fendo.2022.1056939] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/07/2022] [Indexed: 12/23/2022] Open
Abstract
In vertebrates, the tachykinin system includes tachykinin genes, which encode one or two peptides each, and tachykinin receptors. The complexity of this system is reinforced by the massive conservation of gene duplicates after the whole-genome duplication events that occurred in vertebrates and furthermore in teleosts. Added to this, the expression of the tachykinin system is more widespread than first thought, being found beyond the brain and gut. The discovery of the co-expression of neurokinin B, encoded by the tachykinin 3 gene, and kisspeptin/dynorphin in neurons involved in the generation of GnRH pulse, in mammals, put a spotlight on the tachykinin system in vertebrate reproductive physiology. As food intake and reproduction are linked processes, and considering that hypothalamic hormones classically involved in the control of reproduction are reported to regulate also appetite and energy homeostasis, it is of interest to look at the potential involvement of tachykinins in these two major physiological functions. The purpose of this review is thus to provide first a general overview of the tachykinin system in mammals and teleosts, before giving a state of the art on the different levels of action of tachykinins in the control of reproduction and food intake. This work has been conducted with a comparative point of view, highlighting the major similarities and differences of tachykinin systems and actions between mammals and teleosts.
Collapse
Affiliation(s)
- Aurora Campo
- Muséum National d’Histoire Naturelle, Research Unit Unité Mixte de Recherche Biologie des Organsimes et Ecosystèmes Aquatiques (UMR BOREA), Biology of Aquatic Organisms and Ecosystems, Centre National pour la Recherche Scientifique (CNRS), Institut de Recherche pour le Développemen (IRD), Sorbonne Université, Paris, France
- Volcani Institute, Agricultural Research Organization, Rishon LeTsion, Israel
| | - Sylvie Dufour
- Muséum National d’Histoire Naturelle, Research Unit Unité Mixte de Recherche Biologie des Organsimes et Ecosystèmes Aquatiques (UMR BOREA), Biology of Aquatic Organisms and Ecosystems, Centre National pour la Recherche Scientifique (CNRS), Institut de Recherche pour le Développemen (IRD), Sorbonne Université, Paris, France
| | - Karine Rousseau
- Muséum National d’Histoire Naturelle, Research Unit Unité Mixte de Recherche Biologie des Organsimes et Ecosystèmes Aquatiques (UMR BOREA), Biology of Aquatic Organisms and Ecosystems, Centre National pour la Recherche Scientifique (CNRS), Institut de Recherche pour le Développemen (IRD), Sorbonne Université, Paris, France
- Muséum National d’Histoire Naturelle, Research Unit PhyMA Physiologie Moléculaire et Adaptation CNRS, Paris, France
- *Correspondence: Karine Rousseau,
| |
Collapse
|
35
|
Tsukamura H. Kobayashi Award 2019: The neuroendocrine regulation of the mammalian reproduction. Gen Comp Endocrinol 2022; 315:113755. [PMID: 33711315 DOI: 10.1016/j.ygcen.2021.113755] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/13/2021] [Accepted: 02/23/2021] [Indexed: 02/05/2023]
Abstract
Mammalian reproductive function is a complex system of many players orchestrated by the hypothalamus-pituitary-gonadal (HPG) axis. The hypothalamic gonadotropin-releasing hormone (GnRH) and the consequent pituitary gonadotropin release show two modes of secretory patterns, namely the surge and pulse modes. The surge mode is triggered by the positive feedback action of estrogen secreted from the mature ovarian follicle to induce ovulation in females of most mammalian species. The pulse mode of GnRH release is required for stimulating tonic gonadotropin secretion to drive folliculogenesis, spermatogenesis and steroidogenesis and is negatively fine-tuned by the sex steroids. Accumulating evidence suggests that hypothalamic kisspeptin neurons are the master regulator for animal reproduction to govern the HPG axis. Specifically, kisspeptin neurons located in the anterior hypothalamus, such as the anteroventral periventricular nucleus (AVPV) in rodents and preoptic nucleus (POA) in ruminants, primates and others, and the neurons located in the arcuate nucleus (ARC) in posterior hypothalamus in most mammals are considered to play a key role in generating the surge and pulse modes of GnRH release, respectively. The present article focuses on the role of AVPV (or POA) kisspeptin neurons as a center for GnRH surge generation and of the ARC kisspeptin neurons as a center for GnRH pulse generation to mediate estrogen positive and negative feedback mechanisms, respectively, and discusses how the estrogen epigenetically regulates kisspeptin gene expression in these two populations of neurons. This article also provides the mechanism how malnutrition and lactation suppress GnRH/gonadotropin pulses through an inhibition of the ARC kisspeptin neurons. Further, the article discusses the programming effect of estrogen on kisspeptin neurons in the developmental brain to uncover the mechanism underlying the sex difference in GnRH/gonadotropin release as well as an irreversible infertility induced by supra-physiological estrogen exposure in rodent models.
Collapse
Affiliation(s)
- Hiroko Tsukamura
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan.
| |
Collapse
|
36
|
Regulation of stress response on the hypothalamic-pituitary-gonadal axis via gonadotropin-inhibitory hormone. Front Neuroendocrinol 2022; 64:100953. [PMID: 34757094 DOI: 10.1016/j.yfrne.2021.100953] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/16/2021] [Accepted: 10/24/2021] [Indexed: 11/21/2022]
Abstract
Under stressful condition, reproductive function is impaired due to the activation of various components of the hypothalamic-pituitaryadrenal (HPA) axis, which can suppress the activity of the hypothalamic-pituitary-gonadal (HPG) axis at multiple levels. A hypothalamic neuropeptide, gonadotropin-inhibitory hormone (GnIH) is a key negative regulator of reproduction that governs the HPG axis. Converging lines of evidence have suggested that different stress types and their duration, such as physical or psychological, and acute or chronic, can modulate the GnIH system. To clarify the sensitivity and reactivity of the GnIH system in response to stress, we summarize and critically review the available studies that investigated the effects of various stressors, such as restraint, nutritional/metabolic and social stress, on GnIH expression and/or its neuronal activity leading to altered HPG action. In this review, we focus on GnIH as the potential novel mediator responsible for stress-induced reproductive dysfunction.
Collapse
|
37
|
Göcz B, Takács S, Skrapits K, Rumpler É, Solymosi N, Póliska S, Colledge WH, Hrabovszky E, Sárvári M. Estrogen differentially regulates transcriptional landscapes of preoptic and arcuate kisspeptin neuron populations. Front Endocrinol (Lausanne) 2022; 13:960769. [PMID: 36093104 PMCID: PMC9454256 DOI: 10.3389/fendo.2022.960769] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/28/2022] [Indexed: 11/17/2022] Open
Abstract
Kisspeptin neurons residing in the rostral periventricular area of the third ventricle (KPRP3V) and the arcuate nucleus (KPARC) mediate positive and negative estrogen feedback, respectively. Here, we aim to compare transcriptional responses of KPRP3V and KPARC neurons to estrogen. Transgenic mice were ovariectomized and supplemented with either 17β-estradiol (E2) or vehicle. Fluorescently tagged KPRP3V neurons collected by laser-capture microdissection were subjected to RNA-seq. Bioinformatics identified 222 E2-dependent genes. Four genes encoding neuropeptide precursors (Nmb, Kiss1, Nts, Penk) were robustly, and Cartpt was subsignificantly upregulated, suggesting putative contribution of multiple neuropeptides to estrogen feedback mechanisms. Using overrepresentation analysis, the most affected KEGG pathways were neuroactive ligand-receptor interaction and dopaminergic synapse. Next, we re-analyzed our previously obtained KPARC neuron RNA-seq data from the same animals using identical bioinformatic criteria. The identified 1583 E2-induced changes included suppression of many neuropeptide precursors, granins, protein processing enzymes, and other genes related to the secretory pathway. In addition to distinct regulatory responses, KPRP3V and KPARC neurons exhibited sixty-two common changes in genes encoding three hormone receptors (Ghsr, Pgr, Npr2), GAD-65 (Gad2), calmodulin and its regulator (Calm1, Pcp4), among others. Thirty-four oppositely regulated genes (Kiss1, Vgf, Chrna7, Tmem35a) were also identified. The strikingly different transcriptional responses in the two neuron populations prompted us to explore the transcriptional mechanism further. We identified ten E2-dependent transcription factors in KPRP3V and seventy in KPARC neurons. While none of the ten transcription factors interacted with estrogen receptor-α, eight of the seventy did. We propose that an intricate, multi-layered transcriptional mechanism exists in KPARC neurons and a less complex one in KPRP3V neurons. These results shed new light on the complexity of estrogen-dependent regulatory mechanisms acting in the two functionally distinct kisspeptin neuron populations and implicate additional neuropeptides and mechanisms in estrogen feedback.
Collapse
Affiliation(s)
- Balázs Göcz
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
- *Correspondence: Erik Hrabovszky, ; Miklós Sárvári, ; Balázs Göcz,
| | - Szabolcs Takács
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Éva Rumpler
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Norbert Solymosi
- Centre for Bioinformatics, University of Veterinary Medicine, Budapest, Hungary
| | - Szilárd Póliska
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - William H. Colledge
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
- *Correspondence: Erik Hrabovszky, ; Miklós Sárvári, ; Balázs Göcz,
| | - Miklós Sárvári
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
- *Correspondence: Erik Hrabovszky, ; Miklós Sárvári, ; Balázs Göcz,
| |
Collapse
|
38
|
Hypothalamic kisspeptin and kisspeptin receptors: Species variation in reproduction and reproductive behaviours. Front Neuroendocrinol 2022; 64:100951. [PMID: 34757093 DOI: 10.1016/j.yfrne.2021.100951] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/22/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023]
Abstract
Kisspeptin, encoded by the KISS1 gene, was first discovered as a potential metastasis suppressor gene. The prepro-kisspeptin precursor is cleaved into shorter mature bioactive peptides of varying sizes that bind to the G protein-coupled receptor GPR54 (=KISS1R). Over the last two decades, multiple types of Kiss and KissR genes have been discovered in mammalian and non-mammalian vertebrate species, but they are remarkably absent in birds. Kiss neuronal populations are distributed mainly in the hypothalamus. The KissRs are widely distributed in the brain, including the hypothalamic and non-hypothalamic regions, such as the hippocampus, amygdala, and habenula. The role of KISS1-KISS1R in humans and Kiss1-Kiss1R in rodents is associated with puberty, gonadal maturation, and the reproductive axis. However, recent gene deletion studies in zebrafish and medaka have provided controversial results, suggesting that the reproductive role of kiss is dispensable. This review highlights the evolutionary history, localisation, and significance of Kiss-KissR in reproduction and reproductive behaviours in mammalian and non-mammalian vertebrates.
Collapse
|
39
|
Zhao C, Wang B, Liu Y, Feng C, Xu S, Wang W, Liu Q, Li J. New Evidence for the Existence of Two Kiss/Kissr Systems in a Flatfish Species, the Turbot ( Scophthalmus maximus), and Stimulatory Effects on Gonadotropin Gene Expression. Front Endocrinol (Lausanne) 2022; 13:883608. [PMID: 35784551 PMCID: PMC9240279 DOI: 10.3389/fendo.2022.883608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
Seasonal reproduction is generally controlled by the hypothalamus-pituitary-gonadal (HPG) axis in fish. Previous studies have demonstrated that the kisspeptin (Kiss)/kisspeptin receptor (Kissr) system, a positive regulator of the HPG axis, mediates the responses to environmental cues. Turbot (Scophthalmus maximus), a representative species of Pleuronectiformes, is one of the most commercially important fish species cultured in Europe and North China. However, the mechanisms by which the Kiss/Kissr system regulates the reproductive axis of turbot according to seasonal changes, especially photoperiod, have not been clearly characterized. In the current study, the cDNA sequences of kiss2/kissr2, along with kiss1/kissr3 which was thought to be lost in flatfish species, were cloned and functionally characterized. The kiss1, kiss2, and kissr3 transcripts were highly detected in the brain and gonad, while kissr2 mRNA was only abundantly expressed in the brain. Moreover, kiss/kissr mRNAs were further examined in various brain areas of both sexes. The kiss1, kissr2, kissr3 mRNAs were highly expressed in the mesencephalon, while a substantial degree of kiss2 transcripts were observed in the hypothalamus. During annual reproductive cycle, both kiss and kissr transcript levels declined significantly from the immature to mature stages and increased at the degeneration stage in the brains of both sexes, especially in the mesencephalon and hypothalamus. The ovarian kiss1, kiss2, and kissr2 mRNA levels were highest at the vitellogenic stage (mature stage), while expression of kissr3 was highest at the immature stage. The testicular kiss and kissr transcripts were highest in the immature and degeneration stages, and lowest at the mature stage. In addition, intraperitoneal injection of Kiss1-10 and Kiss2-10 significantly stimulated mRNA levels of pituitary lhβ, fhsβ, and gthα. In summary, two Kiss/Kissr systems were firstly proven in a flatfish species of turbot, and it has a positive involvement in controlling the reproduction of the Kiss/Kissr system in turbot. The results will provide preliminary information regarding how the Kiss/Kissr system controls seasonal reproduction in turbot broodstock.
Collapse
Affiliation(s)
- Chunyan Zhao
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, China
- The Key Laboratory of Experimental Marine Biology, Centre for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Bin Wang
- Key Laboratory of Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Yifan Liu
- The Key Laboratory of Experimental Marine Biology, Centre for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan, China
| | - Chengcheng Feng
- The Key Laboratory of Experimental Marine Biology, Centre for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Shihong Xu
- The Key Laboratory of Experimental Marine Biology, Centre for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Wenqi Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, China
| | - Qinghua Liu
- The Key Laboratory of Experimental Marine Biology, Centre for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jun Li
- The Key Laboratory of Experimental Marine Biology, Centre for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
- *Correspondence: Jun Li,
| |
Collapse
|
40
|
Santos LC, Dos Anjos Cordeiro JM, Santana LDS, Barbosa EM, Santos BR, da Silva TQM, de Souza SS, Corrêa JMX, Lavor MSL, da Silva EB, Silva JF. Expression profile of the Kisspeptin/Kiss1r system and angiogenic and immunological mediators in the ovary of cyclic and pregnant cats. Domest Anim Endocrinol 2022; 78:106650. [PMID: 34399365 DOI: 10.1016/j.domaniend.2021.106650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 11/03/2022]
Abstract
The Kisspeptin/Kiss1r system has been studied in mammalian ovaries. However, there are still no studies on the modulation of this system and its relationship with angiogenic and immunological mediators in the ovary of domestic cats, especially during pregnancy. We evaluated the expression of Kisspeptin/Kiss1r and angiogenic and immunological mediators during folliculogenesis, luteogenesis and luteal regression of cyclic and pregnant cats. The ovary exhibited moderate to intense expression for Kiss1, VEGF, Flk-1, INFγ and MIF in oocytes and the follicular wall, while Kiss1r expression was low in granulosa cells. In these cells, there was also a greater expression of Kiss1, INFγ and MIF, mainly in secondary follicles, while tertiary and preovulatory follicles exhibited greater expression of VEGF and Flk-1 in this layer. In luteogenesis, Kiss1 immunostaining was higher in mature corpora lutea (MCL) of pregnant cats compared to vacuolated CL (VCL) and corpus albicans (CA). Pregnancy also increased the luteal gene expression of Kiss1 as well as Kiss1, Kiss1r, Flk-1, and MIF immunostaining in MCL, while reduced the area of VEGF expression in VCL and luteal mRNA expression of Mif when compared to non-pregnant animals. In addition, positive gene correlation between Kiss1r and Mif was observed in the CL. Kiss1, Kiss1r, Vegf and Mif expression were lower in the CA of cats in anestrus. These findings reveal that the expression of Kisspeptin/Kiss1r and angiogenic and immunological mediators, in the ovary of domestic cats, depend on the follicular and luteal stage, and the luteal expression of these mediators is influenced by pregnancy.
Collapse
Affiliation(s)
- Luciano Cardoso Santos
- Department of Biological Sciences, Centro de Microscopia Eletronica, Universidade Estadual de Santa Cruz, 45662-900, Ilheus, Brazil
| | | | - Larissa da Silva Santana
- Department of Biological Sciences, Centro de Microscopia Eletronica, Universidade Estadual de Santa Cruz, 45662-900, Ilheus, Brazil
| | - Erikles Macêdo Barbosa
- Department of Biological Sciences, Centro de Microscopia Eletronica, Universidade Estadual de Santa Cruz, 45662-900, Ilheus, Brazil
| | - Bianca Reis Santos
- Department of Biological Sciences, Centro de Microscopia Eletronica, Universidade Estadual de Santa Cruz, 45662-900, Ilheus, Brazil
| | - Thayná Queiroz Menezes da Silva
- Department of Biological Sciences, Centro de Microscopia Eletronica, Universidade Estadual de Santa Cruz, 45662-900, Ilheus, Brazil
| | - Sophia Saraiva de Souza
- Department of Agricultural Sciences, Hospital Veterinario, Universidade Estadual de Santa Cruz, 45662-900, Ilheus, Brazil
| | - Janaina Maria Xavier Corrêa
- Department of Agricultural Sciences, Hospital Veterinario, Universidade Estadual de Santa Cruz, 45662-900, Ilheus, Brazil
| | - Mário Sergio Lima Lavor
- Department of Agricultural Sciences, Hospital Veterinario, Universidade Estadual de Santa Cruz, 45662-900, Ilheus, Brazil
| | - Elisângela Barboza da Silva
- Department of Agricultural Sciences, Hospital Veterinario, Universidade Estadual de Santa Cruz, 45662-900, Ilheus, Brazil
| | - Juneo Freitas Silva
- Department of Biological Sciences, Centro de Microscopia Eletronica, Universidade Estadual de Santa Cruz, 45662-900, Ilheus, Brazil.
| |
Collapse
|
41
|
Cheng L, Yang S, Si L, Wei M, Guo S, Chen Z, Wang S, Qiao Y. Direct effect of RFRP-3 microinjection into the lateral ventricle on the hypothalamic kisspeptin neurons in ovariectomized estrogen-primed rats. Exp Ther Med 2021; 23:24. [PMID: 34815776 PMCID: PMC8593914 DOI: 10.3892/etm.2021.10946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 09/21/2021] [Indexed: 11/29/2022] Open
Abstract
RFamide-related peptide-3 (RFRP-3) may be involved in the inhibition of kisspeptin, but there is no direct evidence that RFRP-3 can directly act on kisspeptin neurons. The present study aimed to investigate the role and mechanism of RFRP-3 and kisspeptin in the hypothalamic-pituitary reproductive axis. In order to detect the expression and localization of RFRP-3 and kisspeptin in dorsomedial hypothalamic nucleus, double immunofluorescence method combined with confocal microscopy were performed. RFRP-3 was injected into the lateral ventricle of ovariectomized estrogen primed rats. Blood and brain tissues were collected at 60-, 120-, 240- and 360-min. Serum levels of gonadotropin-releasing hormone, luteinizing hormone and follicle-stimulating hormone were detected by ELISA. Kisspeptin expression in hypothalamus was detected by western blotting. Finally, surface plasmon resonance was used to verify whether RFRP-3 can directly interact with kisspeptin. Confocal images indicated that RFRP-3 and kisspeptin were co-expressed in the same neurons in the hypothalamus of ovariectomized estrogen-primed rats. Serum concentrations of gonadotropin-releasing hormone, luteinizing hormone and follicle-stimulating hormone were demonstrated to be significantly reduced following microinjection of RFRP-3 into the lateral ventricle for 60, 120, 240 and 360 min compared with the corresponding saline groups. The expression levels of kisspeptin in hypothalamus were gradually decreased following microinjection of RFRP-3 into the lateral ventricle. In addition, the affinity constant (KD) of RFRP-3 binding to kisspeptin was 6.005x10-5 M, indicating that RFRP-3 bound directly to kisspeptin in the range of protein-protein binding strength (KD, 10-3-10-6 M). In conclusion, RFRP-3 may regulate the hypothalamic-pituitary reproductive axis by inhibiting the expression of hypothalamic kisspeptin and direct binding.
Collapse
Affiliation(s)
- Luyang Cheng
- Department of Immunology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Songhe Yang
- Graduate School, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Lina Si
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Meng Wei
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Sen Guo
- Department of Immunology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Zhihong Chen
- Graduate School, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Shusong Wang
- Hebei Provincial Key Laboratory of Reproductive Medicine, Family Planning Science and Technology Research Institute of Hebei Province, Shijiazhuang, Hebei 050000, P.R. China
| | - Yuebing Qiao
- Graduate School, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| |
Collapse
|
42
|
Duittoz A, Cayla X, Fleurot R, Lehnert J, Khadra A. Gonadotrophin-releasing hormone and kisspeptin: It takes two to tango. J Neuroendocrinol 2021; 33:e13037. [PMID: 34533248 DOI: 10.1111/jne.13037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 01/06/2023]
Abstract
Kisspeptin (Kp), a family of peptides comprising products of the Kiss1 gene, was discovered 20 years ago; it is recognised as the major factor controlling the activity of the gonadotrophin-releasing hormone (GnRH) neurones and thus the activation of the reproductive axis in mammals. It has been widely documented that the effects of Kp on reproduction through its action on GnRH neurones are mediated by the GPR54 receptor. Kp controls the activation of the reproductive axis at puberty, maintains reproductive axis activity in adults and is involved in triggering ovulation in some species. Although there is ample evidence coming from both conditional knockout models and conditional-induced Kp neurone death implicating the Kp/GPR54 pathway in the control of reproduction, the mechanism(s) underlying this process may be more complex than a sole direct control of GnRH neuronal activity by Kp. In this review, we provide an overview of the recent advances made in elucidating the interplay between Kp- and GnRH- neuronal networks with respect to regulating the reproductive axis. We highlight the existence of a possible mutual regulation between GnRH and Kp neurones, as well as the implication of Kp-dependent volume transmission in this process. We also discuss the capacity of heterodimerisation between GPR54 and GnRH receptor (GnRH-R) and its consequences on signalling. Finally, we illustrate the role of mathematical modelling that accounts for the synergy between GnRH-R and GPR54 in explaining the role of these two receptors when defining GnRH neuronal activity and GnRH pulsatile release.
Collapse
Affiliation(s)
- Anne Duittoz
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRAe Val de Loire, Université de Tours, IFCE, Nouzilly, France
| | - Xavier Cayla
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRAe Val de Loire, Université de Tours, IFCE, Nouzilly, France
| | - Renaud Fleurot
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRAe Val de Loire, Université de Tours, IFCE, Nouzilly, France
| | - Jonas Lehnert
- Department of Quantitative Life Sciences, McGill University, Montreal, QC, Canada
| | - Anmar Khadra
- Department of Quantitative Life Sciences, McGill University, Montreal, QC, Canada
- Department of Physiology, McGill University, Montréal, QC, Canada
| |
Collapse
|
43
|
Aerts EG, Harlow K, Griesgraber MJ, Bowdridge EC, Hardy SL, Nestor CC, Hileman SM. Kisspeptin, Neurokinin B, and Dynorphin Expression during Pubertal Development in Female Sheep. BIOLOGY 2021; 10:biology10100988. [PMID: 34681086 PMCID: PMC8533601 DOI: 10.3390/biology10100988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/16/2021] [Accepted: 09/25/2021] [Indexed: 12/14/2022]
Abstract
The neural mechanisms underlying increases in gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) secretion that drive puberty onset are unknown. Neurons coexpressing kisspeptin, neurokinin B (NKB), and dynorphin, i.e., KNDy neurons, are important as kisspeptin and NKB are stimulatory, and dynorphin inhibitory, to GnRH secretion. Given this, we hypothesized that kisspeptin and NKB expression would increase, but that dynorphin expression would decrease, with puberty. We collected blood and hypothalamic tissue from ovariectomized lambs implanted with estradiol at five, six, seven, eight (puberty), and ten months of age. Mean LH values and LH pulse frequency were the lowest at five to seven months, intermediate at eight months, and highest at ten months. Kisspeptin and NKB immunopositive cell numbers did not change with age. Numbers of cells expressing mRNA for kisspeptin, NKB, or dynorphin were similar at five, eight, and ten months of age. Age did not affect mRNA expression per cell for kisspeptin or NKB, but dynorphin mRNA expression per cell was elevated at ten months versus five months. Thus, neither KNDy protein nor mRNA expression changed in a predictable manner during pubertal development. These data raise the possibility that KNDy neurons, while critical, may await other inputs for the initiation of puberty.
Collapse
Affiliation(s)
- Eliana G. Aerts
- Department of Physiology and Pharmacology, West Virginia University, P.O. Box 9229, Morgantown, WV 26506, USA; (E.G.A.); (M.J.G.); (E.C.B.); (S.L.H.)
| | - KaLynn Harlow
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA; (K.H.); (C.C.N.)
| | - Max J. Griesgraber
- Department of Physiology and Pharmacology, West Virginia University, P.O. Box 9229, Morgantown, WV 26506, USA; (E.G.A.); (M.J.G.); (E.C.B.); (S.L.H.)
| | - Elizabeth C. Bowdridge
- Department of Physiology and Pharmacology, West Virginia University, P.O. Box 9229, Morgantown, WV 26506, USA; (E.G.A.); (M.J.G.); (E.C.B.); (S.L.H.)
| | - Steven L. Hardy
- Department of Physiology and Pharmacology, West Virginia University, P.O. Box 9229, Morgantown, WV 26506, USA; (E.G.A.); (M.J.G.); (E.C.B.); (S.L.H.)
| | - Casey C Nestor
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA; (K.H.); (C.C.N.)
| | - Stanley M. Hileman
- Department of Physiology and Pharmacology, West Virginia University, P.O. Box 9229, Morgantown, WV 26506, USA; (E.G.A.); (M.J.G.); (E.C.B.); (S.L.H.)
- Department of Neuroscience, West Virginia University, Morgantown, WV 26506, USA
- Correspondence: ; Tel.: +1-304-293-1502; Fax: +1-304-293-3850
| |
Collapse
|
44
|
Rahawy MA, Al-Mutar HAK. Association of the KiSS1 gene with litter size in Cyprus and Iraqi black goats. Vet World 2021; 14:1995-2001. [PMID: 34566313 PMCID: PMC8448648 DOI: 10.14202/vetworld.2021.1995-2001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/17/2021] [Indexed: 11/17/2022] Open
Abstract
Aim: The study investigated the genetic polymorphism of the kisspeptin (KiSS1) gene and its relationship with litter size in Cyprus and Iraqi black goats. Materials and Methods: Blood samples (n=124) were collected from the two goat breeds reared at the Agricultural Research-Ruminant Research Station Breeding Station, Baghdad, Iraq. Genomic DNA was isolated using a DNA extraction kit. Polymerase chain reaction (PCR) was used to amplify the KiSS1 gene. All PCR products were sequenced and samples were used for further analysis using NCBI-Blast online on the exon 1 (595 bp) region of the KiSS1 gene. Results: The results of this study revealed a significantly (P<0.05) larger litter size of the Cyprus goat breed than in the Iraqi black goats in the first and second parity. Three (893G/C, 973C/A, and 979T/G) substitutions relative to the KiSS1 gene reference sequence (GenBank ID: J × 047312.1, KC989928.1) were identified. Only the mutation g893G>C was identified as a single nucleotide polymorphism (SNP) associated with litter size. Furthermore, the average alleles in KiSS1 gene of both types of goats 0.567 and 0.3715 GG, were recorded. The genotyping at locus g893C>G was demonstrating domination of fecundity quality litter size, Both genotypes SNP of GC were classified at this marked region of KiSS1 gene. Conclusion: The study concluded that the role of the KiSS1 gene in fecundity, revealing the status of this gene as an indicator in the assisted of caprine breeding selection.
Collapse
Affiliation(s)
- M A Rahawy
- Department of Surgery and Theriogenology, College of Veterinary Medicine, University of Mosul, Mosul, Iraq
| | | |
Collapse
|
45
|
Li Y, Zhao T, Liu Y, Lin H, Li S, Zhang Y. Knockout of tac3 genes in zebrafish shows no impairment of reproduction. Gen Comp Endocrinol 2021; 311:113839. [PMID: 34181932 DOI: 10.1016/j.ygcen.2021.113839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/07/2021] [Accepted: 06/16/2021] [Indexed: 01/28/2023]
Abstract
Neurokinin B (NKB) plays a pivotal role in the regulation of reproduction in vertebrates. However, whether this neuropeptide is dispensable for reproduction in teleosts remains unknown. In order to reveal its authentic functions in fish, in this study, two tachykinin 3 (tac3) genes encoding Nkbs were functional mutated in zebrafish using the Transcription Activator-like Effector Nucleases (TALEN) technology. We established tac3a-/-, tac3b-/- and tac3a-/-;tac3b-/- mutant lines, and investigated their reproductive performance and ontogeny. According to our study, spermatogenesis and folliculogenesis were not impaired in tac3a-/-, tac3b-/- and tac3a-/-;tac3b-/- mutant lines, but changes in the expression of genes related to reproductive axis were observed after loss of Tac3, suggesting that possible compensatory response was activated to maintained the reproductive function in zebrafish. In summary, our results indicate that mutation of tac3 genes do not disrupt the reproduction in zebrafish unlike in mammals, revealing the plasticity of reproductive neuroendocrine system in the brain of zebrafish.
Collapse
Affiliation(s)
- Yu Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - TingTing Zhao
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yun Liu
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Haoran Lin
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266373, China; Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Zhuhai 519000, China
| | - Shuisheng Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Yong Zhang
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266373, China; Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Zhuhai 519000, China.
| |
Collapse
|
46
|
Rzucidlo CL, Sperou ES, Holser RR, Khudyakov JI, Costa DP, Crocker DE. Changes in serum adipokines during natural extended fasts in female northern elephant seals. Gen Comp Endocrinol 2021; 308:113760. [PMID: 33781740 DOI: 10.1016/j.ygcen.2021.113760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 12/30/2020] [Accepted: 03/10/2021] [Indexed: 12/31/2022]
Abstract
Adipose tissue is essential to endotherms for thermoregulation and energy storage as well as functioning as an endocrine organ. Adipose derived hormones, or adipokines, regulate metabolism, energy expenditure, reproduction, and immune function in model systems but are less well studied in wildlife. Female northern elephant seals (NES) achieve high adiposity during foraging and then undergo natural fasts up to five weeks long during haul-outs associated with reproduction and molting, resulting in large changes in adipose reserves. We measured circulating levels of four adipokines: leptin, resistin, adiponectin, and kisspeptin-54, in 196 serum samples from female NES at the beginning and end of their breeding and molting fasts. We examined the relationships between these adipokines and life-history stage, adiposity, mass, cortisol, and an immune cytokine involved in the innate immune response interleukin 6 (IL-6). All four adipokines varied with life-history stage. Leptin concentrations were highest at the beginning of the breeding haul-out. Resistin concentrations were higher throughout the breeding haul-out compared to the molt haul-out. Adiponectin concentrations were highest at the beginning of both haul-outs. Kisspeptin-54 concentrations were highest at the end of the breeding haul-out. Leptin, resistin, and adiponectin were associated with measures of body condition, either adiposity, mass, or both. Resistin, adiponectin, and kisspeptin-54 were associated with circulating cortisol concentrations. Resistin was strongly associated with circulating IL-6, a multifunctional cytokine. Adiponectin was associated with glucose concentrations, suggesting a potential role in tissue-specific insulin sensitivity during life-history stages categorized by high adiposity. Increased cortisol concentrations late in lactation were associated with increased kisspeptin-54, suggesting a link to ovulation initiation in NES. This study suggests dramatic changes in circulating adipokines with life-history and body condition that may exert important regulatory roles in NES. The positive relationship between adiponectin and adiposity as well as the lack of a relationship between leptin and kisspeptin-54 differed from model systems. These differences from biomedical model systems suggest the potential for modifications of expression and function of adipose-derived hormones in species that undergo natural changes in adiposity as part of their life-history.
Collapse
Affiliation(s)
- Caroline L Rzucidlo
- Department of Biology, Sonoma State University, Rohnert Park, CA 94928, United States.
| | - Emily S Sperou
- Department of Biology, Sonoma State University, Rohnert Park, CA 94928, United States
| | - Rachel R Holser
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, United States
| | - Jane I Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, United States
| | - Daniel P Costa
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, United States
| | - Daniel E Crocker
- Department of Biology, Sonoma State University, Rohnert Park, CA 94928, United States
| |
Collapse
|
47
|
Terse PS, Peggins J, Seminara SB. Safety Evaluation of KP-10 (Metastin 45-54) Following once Daily Intravenous Administration for 14 Days in Dog. Int J Toxicol 2021; 40:337-343. [PMID: 34126799 DOI: 10.1177/10915818211023459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Kisspeptin-10 (previously referred as metastin 45-54), an active fragment of the endogenous full-length kisspeptin-145, is a potential therapeutic agent for reproductive disorders such as infertility, amenorrhea, and pubertal delay. A safety evaluation of KP-10 was conducted in dogs at the doses of 30, 100, and 1,000 μg/kg, given once daily intravenously for 14 days with a 14-day recovery period. There were no overt signs of drug-related toxicity observed in clinical signs, body weights, food consumption, clinical pathology, histopathology, urinalysis, electrocardiogram, or respiratory rate. Due to very rapid clearance of the peptide, luteinizing hormone (LH) levels were measured as a surrogate marker to demonstrate KP-10 exposure. The LH response reached a maximum concentration at 5 minutes post-dose and remained relatively unchanged for at least 30 minutes after dosing with no gender effect. LH concentrations on Day 1 were generally greater than on day 14. Vaginal cytology results indicated all dogs were in anestrous throughout the dosing period. There were also no KP-10-related findings observed in recovery animals on Day 29. In conclusion, KP-10 demonstrated favorable safety profile in dog where 1,000 μg/kg dose was considered as a no-observed-adverse-effect level dose when administered IV once daily for 14 days.
Collapse
Affiliation(s)
- Pramod S Terse
- National Center for Advancing Translational Sciences, NIH, Bethesda, MD, USA
| | | | | |
Collapse
|
48
|
Nitric oxide resets kisspeptin-excited GnRH neurons via PIP2 replenishment. Proc Natl Acad Sci U S A 2021; 118:2012339118. [PMID: 33443156 DOI: 10.1073/pnas.2012339118] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fertility relies upon pulsatile release of gonadotropin-releasing hormone (GnRH) that drives pulsatile luteinizing hormone secretion. Kisspeptin (KP) neurons in the arcuate nucleus are at the center of the GnRH pulse generation and the steroid feedback control of GnRH secretion. However, KP evokes a long-lasting response in GnRH neurons that is hard to reconcile with periodic GnRH activity required to drive GnRH pulses. Using calcium imaging, we show that 1) the tetrodotoxin-insensitive calcium response evoked by KP relies upon the ongoing activity of canonical transient receptor potential channels maintaining voltage-gated calcium channels in an activated state, 2) the duration of the calcium response is determined by the rate of resynthesis of phosphatidylinositol 4,5-bisphosphate (PIP2), and 3) nitric oxide terminates the calcium response by facilitating the resynthesis of PIP2 via the canonical pathway guanylyl cyclase/3',5'-cyclic guanosine monophosphate/protein kinase G. In addition, our data indicate that exposure to nitric oxide after KP facilitates the calcium response to a subsequent KP application. This effect was replicated using electrophysiology on GnRH neurons in acute brain slices. The interplay between KP and nitric oxide signaling provides a mechanism for modulation of the refractory period of GnRH neurons after KP exposure and places nitric oxide as an important component for tonic GnRH neuronal pulses.
Collapse
|
49
|
Moustafa A. Hindlimb unloading-induced reproductive suppression via Downregulation of hypothalamic Kiss-1 expression in adult male rats. Reprod Biol Endocrinol 2021; 19:37. [PMID: 33663539 PMCID: PMC7931529 DOI: 10.1186/s12958-021-00694-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 01/08/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Spaceflights-induced microgravity can alter various physiological processes in human's body including the functional status of the reproductive system. Rodent model of tail-suspension hindlimb unloading is extensively used to stimulate the organs responses to the microgravity condition. This study explores the potential effects of hindlimb unloading on testicular functions and spermatogenesis in adult male rats and the underlying mechanism/s. METHODS Twenty Sprague-Dawley rats were allotted into two groups: normally loaded group (control; all arms were in touch with the grid floor) and hindlimb unloaded group (HU; only the forearms were in contact with the grid floor). RESULTS Following 30 days of exposure, the HU group saw a decline in body weight, testicular and epidydimal weights, and all semen parameters. The circulating concentrations of gonadotropin-releasing hormone (GnRH), follicle stimulating hormone (FSH), luteinizing hormone (LH) and testosterone significantly decreased, while levels of kisspeptin, corticosterone, inhibin, prolactin and estradiol (E2) increased in the HU group. Intratesticular levels of 5α-reductase enzyme and dihydrotestosterone (DHT) were suppressed, while the levels of aromatase and kisspeptin were significantly elevated in the HU group. Hypothalamic kisspeptin (Kiss1) mRNA expression levels were downregulated while its receptors (Kiss1R) were upregulated in the HU group. On the contrary, the mRNA expression levels of testicular Kiss1 were upregulated while Kiss1R were downregulated. The pituitary mRNA expression levels of FSHβ and LHβ decreased in the HU group. The levels of the antioxidant enzymes, superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) and nitric oxide (NO) concentrations, and total antioxidant capacity (TAC) were elevated while malondialdehyde (MDA) concentrations declined in the testes of HU group. The testes of the HU rats showed positive immunostaining of caspase-3, heat shock protein 70 (HSP70) and Bcl2. CONCLUSIONS Altogether, these results revealed an inhibitory effect of hindlimb unloading on kisspeptin signaling in the hypothalamic-pituitary-testicular axis with impaired spermatogenesis and steroidogenesis.
Collapse
Affiliation(s)
- Amira Moustafa
- Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
50
|
Macedo-Garzón B, Loredo-Ranjel R, Chávez-Maldonado M, Jiménez-Flores JR, Villamar-Duque TE, Cárdenas R. Distribution and expression of GnRH 1, kiss receptor 2, and estradiol α and ß receptors in the anterior brain of females of Chirostoma humboldtianum. FISH PHYSIOLOGY AND BIOCHEMISTRY 2021; 47:33-47. [PMID: 33118089 DOI: 10.1007/s10695-020-00891-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/15/2020] [Indexed: 06/11/2023]
Abstract
Reproduction in vertebrates is a complex process regulated by many hormones, and by paracrine factors and their receptors. This study aimed to examine the expression of pjGonadotropin-releasing hormone (GnRH 1), the kisspeptin receptor 2 (kissr2), and estradiol receptors α and β (ER α and ER β) during different stages of the sexual cycle and their distribution within the anterior brain of females of Chirostoma humboldtianum. Among these molecules, the kissr2 showed the maximal variation in expression, while GnRH 1 showed minimal variation of expression, and ERβ and ERα had intermediate variation of expression. The distribution of these molecules in the anterior brain was consistent with their levels of expression; kissr2 was widely distributed throughout the telencephalon and diencephalon, while ER and GnRH 1 showed more restricted distributions. No coexpression of kissr2 and ER in GnRH 1ergic neurons, suggesting that regulation of this GnRH variant is indirectly mediated by kisspeptin and estradiol.
Collapse
Affiliation(s)
- Beatriz Macedo-Garzón
- Laboratorio de Endocrinología de peces, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios # 1, Los Reyes Iztacacala, 54090, Tlalnepantla, Edo. de México, México
| | - Rosaura Loredo-Ranjel
- Laboratorio de Endocrinología de peces, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios # 1, Los Reyes Iztacacala, 54090, Tlalnepantla, Edo. de México, México
| | - Mónica Chávez-Maldonado
- Laboratorio de Endocrinología de peces, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios # 1, Los Reyes Iztacacala, 54090, Tlalnepantla, Edo. de México, México
| | - J Rafael Jiménez-Flores
- Laboratorio de Inmunología, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de lo Barrios # 1, Los Reyes Iztacala, 54090, Tlalnepantla, Edo. de México, México
| | - Tomás E Villamar-Duque
- Bioterio General, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios # 1, Los Reyes Iztacala, 54090, Tlalnepantla, Edo. de México, México
| | - Rodolfo Cárdenas
- Laboratorio de Endocrinología de peces, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios # 1, Los Reyes Iztacacala, 54090, Tlalnepantla, Edo. de México, México.
| |
Collapse
|