1
|
Velmurugan H, Mannava AS, Thangaraju P, Neelambaran K. Kisspeptin and its Current Clinical Status-A Systematic Review. Curr Med Chem 2025; 32:1313-1322. [PMID: 38265397 DOI: 10.2174/0109298673251224230919093656] [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: 02/28/2023] [Revised: 06/25/2023] [Accepted: 07/31/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND Kisspeptin was initially known as metastin for its role in suppressing metastasis in melanoma and breast cancer. Later, based on its ability to stimulate GPR54, its importance in maintaining an intact hypothalamic-pituitary-ovarian axis was recognised, which is the basis for the widespread application of the drug in several conditions such as secondary amenorrhea, regulation of puberty onset, ovarian function, trophoblast invasion, fertility regulation, parturition, and lactation. This systematic study aims to evaluate the current status of kisspentin in clinical trials. METHODS The keywords 'kisspeptin' or 'metastin' were used in the clinicaltrials.gov website and Clinical Trial Registry of India (CTRI) to find eligible clinical trials or records carried out without time constraints until February 26, 2023. RESULTS A total of 33 records were identified through clinical trial databases. All records were screened, and four trials were rejected as they failed to meet the inclusion criteria. Finally, 29 (87.9%) reports of interventional clinical trials with kisspeptin were reviewed. CONCLUSION Kisspeptin can be viewed as a multipurpose drug with considerably fewer side effects due to its effects simulating normal physiological processes in our body.
Collapse
Affiliation(s)
- Hemasri Velmurugan
- Department of Pharmacology, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| | - Anjali Srikanth Mannava
- Department of Pharmacology, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| | - Pugazhenthan Thangaraju
- Department of Pharmacology, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| | - Krishnapriya Neelambaran
- Department of Pharmacology, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| |
Collapse
|
2
|
Joy KP, Chaube R. Kisspeptin control of hypothalamus-pituitary-ovarian functions. VITAMINS AND HORMONES 2024; 127:153-206. [PMID: 39864941 DOI: 10.1016/bs.vh.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The discovery of Kisspeptin (Kiss) has opened a new direction in research on neuroendocrine control of reproduction in vertebrates. Belonging to the RF amide family of peptides, Kiss and its cognate receptor Gpr54 (Kissr) have a long and complex evolutionary history. Multiple forms of Kiss and Kissr are identified in non-mammalian vertebrates, with the exception of birds, and monotreme mammals. However, only a single form of the ligand (KISS1/Kiss1) and receptor (KISS1R/Kiss1r) is retained in higher mammals. Kiss1 is distributed in the hypothalamus-pituitary-gonadal (HPG) axis and its primary function is to stimulate gonadotropin-releasing hormone (GnRH) secretion. Kiss1 neurons are distributed in the rostral periventricular area of the third ventricle (RP3V) and arcuate/infundibular nucleus (ARN/IFN). The ARN/IFN is considered the GnRH pulse generator controlled by steroid negative feedback, and the RP3V neurons is concerned with GnRH surge induced by steroid positive feedback in females. The Kiss1-Kiss1r signaling is important in all aspects of reproduction: puberty onset, maintenance of adult gonadal functions and reproductive aging, and hence assumes therapeutic potentials in the treatment of reproductive dysfunctions and induction of artificial reproduction. This chapter reviews involvement of Kiss1 in the control of the HPG axis functions in female mammals.
Collapse
Affiliation(s)
- K P Joy
- Retired Professor, Department of Zoology, Banaras Hindu University, Varanasi, Uttar pradesh, India.
| | - R Chaube
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar pradesh, India
| |
Collapse
|
3
|
Tang DD. Dissecting a Brake for Airway Smooth Muscle Cell Movement. Am J Respir Cell Mol Biol 2024; 70:435-436. [PMID: 38568237 PMCID: PMC11160410 DOI: 10.1165/rcmb.2024-0120ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024] Open
Affiliation(s)
- Dale D Tang
- Department of Molecular and Cellular Physiology Albany Medical College Albany, New York
| |
Collapse
|
4
|
Balraj P, Ambhore NS, Ramakrishnan YS, Borkar NA, Banerjee P, Reza MI, Varadharajan S, Kumar A, Pabelick CM, Prakash YS, Sathish V. Kisspeptin/KISS1R Signaling Modulates Human Airway Smooth Muscle Cell Migration. Am J Respir Cell Mol Biol 2024; 70:507-518. [PMID: 38512807 PMCID: PMC11160419 DOI: 10.1165/rcmb.2023-0469oc] [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: 12/28/2023] [Accepted: 03/20/2024] [Indexed: 03/23/2024] Open
Abstract
Airway remodeling is a cardinal feature of asthma, associated with increased airway smooth muscle (ASM) cell mass and upregulation of extracellular matrix deposition. Exaggerated ASM cell migration contributes to excessive ASM mass. Previously, we demonstrated the alleviating role of Kp (kisspeptin) receptor (KISS1R) activation by Kp-10 in mitogen (PDGF [platelet-derived growth factor])-induced human ASM cell proliferation in vitro and airway remodeling in vivo in a mouse model of asthma. Here, we examined the mechanisms by which KISS1R activation regulates mitogen-induced ASM cell migration. KISS1R activation using Kp-10 significantly inhibited PDGF-induced ASM cell migration, further confirmed using KISS1R shRNA. Furthermore, KISS1R activation modulated F/G actin dynamics and the expression of promigration proteins like CDC42 (cell division control protein 42) and cofilin. Mechanistically, we observed reduced ASM RhoA-GTPAse with KISS1R activation. The antimigratory effect of KISS1R was abolished by PKA (protein kinase A)-inhibitory peptide. Conversely, KISS1R activation significantly increased cAMP and phosphorylation of CREB (cAMP-response element binding protein) in PDGF-exposed ASM cells. Overall, these results highlight the alleviating properties of Kp-10 in the context of airway remodeling.
Collapse
Affiliation(s)
- Premanand Balraj
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| | | | | | | | - Priyanka Banerjee
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| | - Mohammad Irshad Reza
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| | - Subashini Varadharajan
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| | - Ashish Kumar
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| | - Christina M. Pabelick
- Department of Anesthesiology and Perioperative Medicine, and
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Y. S. Prakash
- Department of Anesthesiology and Perioperative Medicine, and
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| |
Collapse
|
5
|
Salmeri N, Viganò P, Cavoretto P, Marci R, Candiani M. The kisspeptin system in and beyond reproduction: exploring intricate pathways and potential links between endometriosis and polycystic ovary syndrome. Rev Endocr Metab Disord 2024; 25:239-257. [PMID: 37505370 DOI: 10.1007/s11154-023-09826-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/18/2023] [Indexed: 07/29/2023]
Abstract
Endometriosis and polycystic ovary syndrome (PCOS) are two common female reproductive disorders with a significant impact on the health and quality of life of women affected. A novel hypothesis by evolutionary biologists suggested that these two diseases are inversely related to one another, representing a pair of diametrical diseases in terms of opposite alterations in reproductive physiological processes but also contrasting phenotypic traits. However, to fully explain the phenotypic features observed in women with these conditions, we need to establish a potential nexus system between the reproductive system and general biological functions. The recent discovery of kisspeptin as pivotal mediator of internal and external inputs on the hypothalamic-pituitary-gonadal axis has led to a new understanding of the neuroendocrine upstream regulation of the human reproductive system. In this review, we summarize the current knowledge on the physiological roles of kisspeptin in human reproduction, as well as its involvement in complex biological functions such as metabolism, inflammation and pain sensitivity. Importantly, these functions are known to be dysregulated in both PCOS and endometriosis. Within the evolving scientific field of "kisspeptinology", we critically discuss the clinical relevance of these discoveries and their potential translational applications in endometriosis and PCOS. By exploring the possibilities of manipulating this complex signaling system, we aim to pave the way for novel targeted therapies in these reproductive diseases.
Collapse
Affiliation(s)
- Noemi Salmeri
- Gynecology and Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Paola Viganò
- Infertility Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via M. Fanti 6, 20122, Milan, Italy.
| | - Paolo Cavoretto
- Gynecology and Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Roberto Marci
- Gynecology & Obstetrics, University of Ferrara, 44121, Ferrara, Italy
| | - Massimo Candiani
- Gynecology and Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| |
Collapse
|
6
|
Gu X, Xiong W, Yang Y, Li H, Xiong C. A comprehensive meta-analysis to identify susceptibility genetic variants for precocious puberty. Ann Hum Genet 2024; 88:138-153. [PMID: 37933223 DOI: 10.1111/ahg.12525] [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: 02/28/2023] [Revised: 07/25/2023] [Accepted: 08/04/2023] [Indexed: 11/08/2023]
Abstract
PURPOSE Currently, several genetic variants in ERα gene (rs2234693 and rs9340799), ERβ gene (rs1256049 and rs4986938), KISS1 gene (rs4889, rs1132506 and rs5780218), LIN28B gene (rs314263, rs314276 and rs314280), and MKRN3 gene (rs2239669) have been repeatedly explored for their contribution to precocious puberty (PP) susceptibility. However, the results remain conflicting rather than conclusive. We here performed a meta-analysis to identify the real susceptibility genetic variants for PP. METHODS After screening by inclusion criteria, 20 related studies were finally included in this meta-analysis. The odds ratios and 95% confidence intervals were calculated to assess the strength of association. Sensitive analysis, publication bias, and trial sequential analysis (TSA) were performed to evaluate the stability and reliability of results. RESULTS Rs2234693, rs9340799, and rs1256049 were significantly associated with PP susceptibility (p < 0.0084). Stratified analysis according to ethnicity showed that rs2234693 and rs9340799 were significantly associated with PP susceptibility in Asian and Chinese populations. Stratified analysis according to PP subtype showed that rs2234693 and rs9340799 were significantly associated with idiopathic central PP susceptibility in Asian and Chinese populations (p < 0.0084). The results of publication bias, sensitivity analysis, and TSA provided solid evidence for the association between these three variants and PP susceptibility. CONCLUSIONS Rs2234693 and rs9340799 in ERα gene and rs1256049 in ERβ gene may serve as susceptive factors for PP development. The present finding should be confirmed in replication studies and reinforced in functional studies, which will ultimately improve the feasibility of the application of these three PP-susceptible loci in clinical practice.
Collapse
Affiliation(s)
- Xiuli Gu
- Wuhan Huake Reproductive Hospital, Wuhan, China
| | - Weining Xiong
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Yan Yang
- Wuhan Huake Reproductive Hospital, Wuhan, China
| | - Honggang Li
- Institute of reproductive health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | |
Collapse
|
7
|
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
|
8
|
Kleimenova T, Polyakova V, Linkova N, Drobintseva A, Medvedev D, Krasichkov A. The Expression of Kisspeptins and Matrix Metalloproteinases in Extragenital Endometriosis. Biomedicines 2024; 12:94. [PMID: 38255200 PMCID: PMC10813454 DOI: 10.3390/biomedicines12010094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/19/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
Endometriosis is characterized by a condition where endometrial tissue grows outside the uterine cavity. The mechanisms of endometrium growth during endometriosis might be similar to the development of a tumor. The kisspeptin (KISS1) gene was initially discovered as a suppressor of metastasis. Matrix metalloproteinases (MMPs) and their inhibitors are described as factors in the early stages of endometriosis and tumor growth progression. We applied the quantitative polymerase chain reaction and the immunofluorescence method to investigate KISS1, its receptor (KISS1R), MMP-2, and MMP-9 in the eutopic and ectopic endometrium in women with and without endometriosis. We presume that the dysregulation of KISS1 and MMPs might contribute to endometriosis pathogenesis. Samples for the immunofluorescence study were collected from patients with a confirmed diagnosis of endometriosis in stages I-IV, aged 23 to 38 years old (n = 40). The cell line was derived from the endometrium of patients with extragenital endometriosis (n = 7). KISS1 and KISS1R expression are present in the ectopic endometrium of patients with extragenital endometriosis, as opposed to the control group where these proteins were not expressed. There is a decrease in KISS1 and KISS1R values at all stages of endometriosis. MMP-2 and MMP-9 genes express statistically significant increases in stages II, III, and IV of extragenital endometriosis. MMP synthesis increased in the last stages of endometriosis. We suppose that the KISS1/KISS1R system can be used in the future as a suppressive complex to reduce MMP-2 and MMP-9 expression and prevent endometrial cells from invading.
Collapse
Affiliation(s)
- Tatiana Kleimenova
- Department of Medical Biology, Federal State Budgetary Educational Institution of Higher Education, St. Petersburg State Pediatric Medical University, Ministry of Healthcare of the Russian Federation, 194100 St. Petersburg, Russia
| | - Victoria Polyakova
- Research Laboratory for the Development of Drug Delivery Systems, St. Petersburg State Research Institute of Phthisiopulmonology, Ministry of Healthcare of the Russian Federation, 2-4, Ligovskiy pr., 191036 St. Petersburg, Russia
| | - Natalia Linkova
- Research Laboratory for the Development of Drug Delivery Systems, St. Petersburg State Research Institute of Phthisiopulmonology, Ministry of Healthcare of the Russian Federation, 2-4, Ligovskiy pr., 191036 St. Petersburg, Russia
- Department of Biogerontology, St. Petersburg Institute of Bioregulation and Gerontology, Dynamo pr., 3, 197110 St. Petersburg, Russia
| | - Anna Drobintseva
- Department of Medical Biology, Federal State Budgetary Educational Institution of Higher Education, St. Petersburg State Pediatric Medical University, Ministry of Healthcare of the Russian Federation, 194100 St. Petersburg, Russia
| | - Dmitriy Medvedev
- Department of Biogerontology, St. Petersburg Institute of Bioregulation and Gerontology, Dynamo pr., 3, 197110 St. Petersburg, Russia
| | - Alexander Krasichkov
- Department of Radio Engineering Systems, Saint Petersburg Electrotechnical University ‘LETI’, 197376 St. Petersburg, Russia
| |
Collapse
|
9
|
Kumar V, Doshi G. Revolutionizing Infertility Management through Novel Peptide-based Targets. Curr Protein Pept Sci 2024; 25:738-752. [PMID: 38778605 DOI: 10.2174/0113892037304433240430144106] [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: 01/15/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024]
Abstract
Around 48 million couples and 186 million people worldwide have infertility; of these, approximately 85% have an identifiable cause, the most common being ovulatory dysfunctions, male infertility, polycystic ovary syndrome, and tubule disease. The remaining 15% have infertility for unknown reasons, including lifestyle and environmental factors. The regulation of the hypothalamic- pituitary-adrenal axis (HPA) is crucial for the secretion of gonadotropin-releasing hormone (GnRH), luteinizing hormone (LH), and follicle-stimulating hormone (FSH), which are essential for female reproductive functions. GnRH is the primary reproductive axis regulator. The pattern of GnRH, FSH, and LH release is determined by its pulsatile secretion, which in turn controls endocrine function and gamete maturation in the gonads. Peptides called Kisspeptin (KP), Neurokinin-B (NKB), and Orexin influence both positive and negative feedback modulation of GnRH, FSH, and LH secretion in reproduction. This review article mainly focuses on the historical perspective, isoform, and signaling pathways of KP, NKB, and Orexin novel peptide-based targets including clinical and preclinical studies and having a promising effect in the management of infertility.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400 056, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400 056, India
| |
Collapse
|
10
|
Zhang J, Jin L, Kong L, Nie L, Yuan D. Physiological and pathological roles of locally expressed kisspeptin and KISS1R in the endometrium. Hum Reprod 2023:7146150. [PMID: 37105233 DOI: 10.1093/humrep/dead080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/25/2023] [Indexed: 04/29/2023] Open
Abstract
Kisspeptins, encoded by the KISS1 gene, are a family of polypeptides that bind the kisspeptin receptor (KISS1R) to perform biological functions. Produced mainly in the hypothalamus, these neuropeptides regulate the pulsatile secretion of GnRH and trigger the hypothalamus-pituitary-gonadal axis. Other peripheral organs also express kisspeptin, which inhibits metastasis. Kisspeptin and KISS1R are reportedly present in the endometrium and may play roles in limiting the migration and invasion of trophoblasts into the endometrium during pregnancy (decidua) to maintain endometrial homeostasis. A deficiency of kisspeptin and KISS1R in the endometrium can lead to pathological conditions such as endometriosis and endometrial carcinoma. Kisspeptin and KISS1R in the endometrium can also promote endometrial receptivity and decidualization. Overall, kisspeptin and KISS1R are important for maintaining the normal physiological functions of the endometrium. By summarizing the roles of kisspeptin and KISS1R in the endometrium, our review explores the regulatory roles in the peripheral reproductive system of this peptide family that plays broad and profound roles in many physiological processes.
Collapse
Affiliation(s)
- Jieyu Zhang
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Lei Jin
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Lingnan Kong
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Li Nie
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Dongzhi Yuan
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Aasif A, Alam R, Ahsan H, Khan MM, Khan A, Khan S. The role of kisspeptin in the pathogenesis of a polycystic ovary syndrome. Endocr Regul 2023; 57:292-303. [PMID: 38127687 DOI: 10.2478/enr-2023-0032] [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] [Indexed: 12/23/2023] Open
Abstract
Hypothalamic-pituitary gonadal (HPG) axis is responsible for the development and regulation of the female reproductive system. In polycystic ovary syndrome (PCOS), there is a disturbance in the HPG axis. Kisspeptin, a neuropeptide produced by the KISS1 gene, plays a vital role in the regulation of HPG axis by binding with its receptors KISS1R/GPR54, and stimulates gonadotropin secretion from the hypothalamus into pituitary to release luteinizing hormone (LH) and follicle stimulating hormone (FSH). Polymorphisms or mutations in the KISS1 gene can cause disturbance in the kisspeptin signaling pathway and is thought to disrupt HPG axis. Altered signaling of kisspeptin can cause abnormal secretion of GnRH pulse, which leads to increased LH/FSH ratio, thereby affecting androgen levels and ovulation. The increased levels of androgen worsen the symptoms of PCOS. In the present article, we review the molecular physiology and pathology of kisspeptin and how it is responsible for the development of PCOS. The goal of this review article is to provide an overview and metabolic profile of kisspeptin in PCOS patients and the expression of kisspeptin in PCOS animal models. In the present article, we also review the molecular physiology and pathology of kisspeptin and how it is responsible for the development of PCOS.
Collapse
Affiliation(s)
- Adiba Aasif
- 1Department of Biochemistry, Integral Institute of Medical Sciences and Research, Lucknow, India
| | - Roshan Alam
- 1Department of Biochemistry, Integral Institute of Medical Sciences and Research, Lucknow, India
| | - Haseeb Ahsan
- 2Department of Biochemistry, Faculty of Dentistry, Jamia Millia Islamia, New Delhi, India
| | - Mohammad Mustufa Khan
- 3Department of Basic Medical Sciences, Integral Institute of Allied Health Sciences and Research, Integral University, Lucknow, India
| | - Arshiya Khan
- 4Department of Obstetrics and Gynecology, Integral Institute of Medical Sciences and Research, Lucknow, India
| | - Saba Khan
- 1Department of Biochemistry, Integral Institute of Medical Sciences and Research, Lucknow, India
| |
Collapse
|
12
|
Hu KL, Chen Z, Deng W, Li X, Ju L, Yang H, Zhang H, Mu L. Diagnostic Value of Kisspeptin Levels on Early Pregnancy Outcome: a Systematic Review and Meta-analysis. Reprod Sci 2022; 29:3365-3372. [PMID: 35212930 DOI: 10.1007/s43032-022-00856-8] [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: 11/09/2021] [Accepted: 01/09/2022] [Indexed: 12/14/2022]
Abstract
The objective of this study is to investigate whether kisspeptin levels in early pregnancy have a better diagnostic value on early pregnancy outcome as compared with human chorionic gonadotropin (hCG). This study was a systematic review and meta-analysis aiming to investigate the diagnostic value of kisspeptin levels on early pregnancy outcome. The primary outcome was miscarriage or viable intrauterine pregnancy. Five studies were included for systematic review, and three studies were included for meta-analysis. Meta-analysis showed kisspeptin levels had a good diagnostic value with the area under the curve (AUC) 0.902 (0.866, 0.937) when kisspeptin was measured after 6 weeks of gestation. Sensitivity analysis demonstrated kisspeptin levels had a diagnostic value with AUC = 0.881 (0.855, 0.906). hCG levels had a diagnostic value with AUC = 0.834 (0.785, 0.883), which was inferior to the diagnostic value of kisspeptin (mean difference = 0.09 (0.02, 0.16)). Kisspeptin measurement has a potential for comparable or even higher accuracy than hCG in differentiating between miscarriage and viable intrauterine pregnancy after 6 weeks of gestation.
Collapse
Affiliation(s)
- Kai-Lun Hu
- Center for Reproductive Medicine, Peking University Third Hospital, No.49 Huayuan North Road, Haidian District, Beijing, People's Republic of China, 100191
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Zimiao Chen
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Wenhai Deng
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Xiaoxue Li
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Liping Ju
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Haiyan Yang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Hong Zhang
- Medical Reproductive Center, The Second Affiliated Hospital of Soochow University, No.1055 Sanxiang Road, Gusu District, Suzhou, People's Republic of China, 215026.
| | - Liangshan Mu
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200.
| |
Collapse
|
13
|
Önal M, Karli P, Özdemir AZ, Kocaman A, Katirci Y, Çoban G, Nakişli GK, Civil Y, Avci B. Serum kisspeptin levels in deep-infiltrating, ovarian, and superficial endometriosis: A prospective observational study. Medicine (Baltimore) 2022; 101:e31529. [PMID: 36397399 PMCID: PMC9666188 DOI: 10.1097/md.0000000000031529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The diagnosis of endometriosis may delay for many years due to non-deterministic symptoms and avoiding surgical interventions. Kisspeptins are hormones that interact with endometrial tissue to limit invasions during placentation and various cancers and are suggested to be also associated with endometriosis. This study evaluated if serum kisspeptin levels are associated with the invasion depth in endometriosis. Forty patients between 18 and 45 years of age and admitted to a tertiary-care Obstetrics and Gynecology Department between 2020 and 2021 with a diagnosis of endometriosis, and 40 patients without endometrioma were included in the study. Demographic, obstetric, clinical, and biochemical characteristics were evaluated in patients with superficial (SE) and deep infiltrating (DIE) endometriosis and healthy controls. Twenty patients (50%) had SE, 14 (35%) had DIE, and 22 (55%) had endometrioma in the patient group. Fertility rates were higher among controls, but similar between patients with SE and DIE. CA125 levels were significantly higher in the DIE group. SE and DIE groups had similar kisspeptin values, significantly higher than controls. CA125 and kisspeptin levels were not correlated in study groups. Serum kisspeptin levels were significantly different between endometriosis patients and healthy controls. However, kisspeptin levels were unable to differentiate endometriosis severity. Our results suggest that kisspeptins might play a role in the pathogenesis of endometriosis, which needs further assessment in more comprehensive studies.
Collapse
Affiliation(s)
- Mesut Önal
- Department of Gynecology and Obstetrics, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
- * Correspondence: Mesut Önal, Department of Gynecology and Obstetrics, Ondokuz Mayis University, Samsun 55200, Turkey (e-mail: )
| | - Pervin Karli
- Department of Gynecology and Obstetrics, Medical Park Hospital, Samsun, Turkey
| | - Ayşe Zehra Özdemir
- Department of Gynecology and Obstetrics, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Adem Kocaman
- Department of Histology and Embryology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Yunus Katirci
- Department of Gynecology and Obstetrics, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Gülnur Çoban
- Department of Gynecology and Obstetrics, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Gülen Kübra Nakişli
- Department of Gynecology and Obstetrics, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Yeşim Civil
- Department of Histology and Embryology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Bahattin Avci
- Department of Biochemistry, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| |
Collapse
|
14
|
Granata L, Gildawie KR, Ismail N, Brenhouse HC, Kopec AM. Immune signaling as a node of interaction between systems that sex-specifically develop during puberty and adolescence. Dev Cogn Neurosci 2022; 57:101143. [PMID: 35933922 PMCID: PMC9357835 DOI: 10.1016/j.dcn.2022.101143] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/08/2022] [Accepted: 08/01/2022] [Indexed: 01/18/2023] Open
Abstract
Adolescence is pivotal for neural and behavioral development across species. During this period, maturation occurs in several biological systems, the most well-recognized being activation of the hypothalamic-pituitary-gonadal axis marking pubertal onset. Increasing comparative studies of sex differences have enriched our understanding of systems integration during neurodevelopment. In recent years, immune signaling has emerged as a key node of interaction between a variety of biological signaling processes. Herein, we review the age- and sex-specific changes that occur in neural, hypothalamic-pituitary, and microbiome systems during adolescence. We then describe how immune signaling interacts with these systems, and review recent preclinical evidence indicating that immune signaling may play a central role in integrating changes in their typical and atypical development during adolescence. Finally, we discuss the translational relevance of these preclinical studies to human health and wellness.
Collapse
Affiliation(s)
- Lauren Granata
- Northeastern University, 125 Nightingale Hall, Boston, MA 02115, USA.
| | - Kelsea R Gildawie
- Tufts University Cummings School of Veterinary Medicine, 200 Westboro Rd. North Grafton, MA 01536, USA.
| | - Nafissa Ismail
- University of Ottawa, 136 Jean-Jacques Lussier, Vanier Hall 2076A, Ottawa, ON K1N 6N5 Canada.
| | | | - Ashley M Kopec
- Albany Medical College, 43 New Scotland Ave., Albany, NY 12208, USA.
| |
Collapse
|
15
|
Tsoutsouki J, Patel B, Comninos AN, Dhillo WS, Abbara A. Kisspeptin in the Prediction of Pregnancy Complications. Front Endocrinol (Lausanne) 2022; 13:942664. [PMID: 35928889 PMCID: PMC9344876 DOI: 10.3389/fendo.2022.942664] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Kisspeptin and its receptor are central to reproductive health acting as key regulators of the reproductive endocrine axis in humans. Kisspeptin is most widely recognised as a regulator of gonadotrophin releasing hormone (GnRH) neuronal function. However, recent evidence has demonstrated that kisspeptin and its receptor also play a fundamental role during pregnancy in the regulation of placentation. Kisspeptin is abundantly expressed in syncytiotrophoblasts, and its receptor in both cyto- and syncytio-trophoblasts. Circulating levels of kisspeptin rise dramatically during healthy pregnancy, which have been proposed as having potential as a biomarker of placental function. Indeed, alterations in kisspeptin levels are associated with an increased risk of adverse maternal and foetal complications. This review summarises data evaluating kisspeptin's role as a putative biomarker of pregnancy complications including miscarriage, ectopic pregnancy (EP), preterm birth (PTB), foetal growth restriction (FGR), hypertensive disorders of pregnancy (HDP), pre-eclampsia (PE), gestational diabetes mellitus (GDM), and gestational trophoblastic disease (GTD).
Collapse
Affiliation(s)
| | | | | | | | - Ali Abbara
- *Correspondence: Waljit S. Dhillo, ; Ali Abbara,
| |
Collapse
|
16
|
Kisspeptin Modulation of Reproductive Function. ENDOCRINES 2022. [DOI: 10.3390/endocrines3030029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Kisspeptin is a peptide expressed mainly in the infundibular nucleus of the hypothalamus. Kisspeptin plays a crucial role in the regulation of reproductive functions. It is regarded as the most important factor responsible for the control of the hypothalamic–pituitary–gonadal axis, the onset of puberty, and the regulation of menstruation and fertility. Kisspeptin activity influences numerous processes such as steroidogenesis, follicular maturation, ovulation, and ovarian senescence. The identification of kisspeptin receptor mutations that cause hypogonadotropic hypogonadism has initiated studies on the role of kisspeptin in puberty. Pathologies affecting the neurons secreting kisspeptin play a major role in the development of PCOS, functional hypothalamic amenorrhea, and perimenopausal vasomotor symptoms. Kisspeptin analogs (both agonists and antagonists), therefore, may be beneficial as therapy in those afflicted with such pathologies. The aim of this review is to summarize the influence of kisspeptin in the physiology and pathology of the reproductive system in humans, as well as its potential use in therapy.
Collapse
|
17
|
Szydełko-Gorzkowicz M, Poniedziałek-Czajkowska E, Mierzyński R, Sotowski M, Leszczyńska-Gorzelak B. The Role of Kisspeptin in the Pathogenesis of Pregnancy Complications: A Narrative Review. Int J Mol Sci 2022; 23:ijms23126611. [PMID: 35743054 PMCID: PMC9223875 DOI: 10.3390/ijms23126611] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/10/2022] [Accepted: 06/12/2022] [Indexed: 12/15/2022] Open
Abstract
Kisspeptins are the family of neuropeptide products of the KISS-1 gene that exert the biological action by binding with the G-protein coupled receptor 54 (GPR54), also known as the KISS-1 receptor. The kisspeptin level dramatically increases during pregnancy, and the placenta is supposed to be its primary source. The role of kisspeptin has already been widely studied in hypogonadotropic hypogonadism, fertility, puberty disorders, and insulin resistance-related conditions, including type 2 diabetes mellitus, polycystic ovary syndrome, and obesity. Gestational diabetes mellitus (GDM), preeclampsia (PE), preterm birth, fetal growth restriction (FGR), or spontaneous abortion affected 2 to 20% of pregnancies worldwide. Their occurrence is associated with numerous short and long-term consequences for mothers and newborns; hence, novel, non-invasive predictors of their development are intensively investigated. The study aims to present a comprehensive review emphasizing the role of kisspeptin in the most common pregnancy-related disorders and neonatal outcomes. The decreased level of kisspeptin is observed in women with GDM, FGR, and a high risk of spontaneous abortion. Nevertheless, there are still many inconsistencies in kisspeptin concentration in pregnancies with preterm birth or PE. Further research is needed to determine the usefulness of kisspeptin as an early marker of gestational and neonatal complications.
Collapse
|
18
|
Hu KL, Chen Z, Li X, Cai E, Yang H, Chen Y, Wang C, Ju L, Deng W, Mu L. Advances in clinical applications of kisspeptin-GnRH pathway in female reproduction. Reprod Biol Endocrinol 2022; 20:81. [PMID: 35606759 PMCID: PMC9125910 DOI: 10.1186/s12958-022-00953-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/30/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Kisspeptin is the leading upstream regulator of pulsatile and surge Gonadotrophin-Releasing Hormone secretion (GnRH) in the hypothalamus, which acts as the key governor of the hypothalamic-pituitary-ovary axis. MAIN TEXT Exogenous kisspeptin or its receptor agonist can stimulate GnRH release and subsequent physiological gonadotropin secretion in humans. Based on the role of kisspeptin in the hypothalamus, a broad application of kisspeptin and its receptor agonist has been recently uncovered in humans, including central control of ovulation, oocyte maturation (particularly in women at a high risk of ovarian hyperstimulation syndrome), test for GnRH neuronal function, and gatekeepers of puberty onset. In addition, the kisspeptin analogs, such as TAK-448, showed promising agonistic activity in healthy women as well as in women with hypothalamic amenorrhoea or polycystic ovary syndrome. CONCLUSION More clinical trials should focus on the therapeutic effect of kisspeptin, its receptor agonist and antagonist in women with reproductive disorders, such as hypothalamic amenorrhoea, polycystic ovary syndrome, and endometriosis.
Collapse
Affiliation(s)
- Kai-Lun Hu
- Center for Reproductive Medicine, Peking University Third Hospital, No.49 Huayuan North Road, Haidian District, Beijing, People's Republic of China, 100191
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Zimiao Chen
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Xiaoxue Li
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Enci Cai
- Department of Nutrition and Food Science, College of Food, Agricultural and Natural Resource Sciences, University of Minnesota, Twin Cities, Minneapolis, MN, 55455, USA
| | - Haiyan Yang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Yi Chen
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Congying Wang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Liping Ju
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Wenhai Deng
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China, 325006.
| | - Liangshan Mu
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200.
| |
Collapse
|
19
|
Xu S, Wang M, Li Y, Tang N, Zhang X, Chen H, Zhang S, Liu Y, Wang J, Chen D, Li Z. Cloning and expression of kiss genes and regulation of feeding in Siberian sturgeon (Acipenser baerii). FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:419-436. [PMID: 35184249 DOI: 10.1007/s10695-022-01055-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/31/2022] [Indexed: 06/14/2023]
Abstract
In 1996, kiss was reported to regulate feeding in mammals, but studies are limited in fish. Our study aimed to explore the possible role of kiss in the regulation of feeding in Siberian sturgeon (Acipenser baerii). kiss1 and kiss2 were cloned, and the expression patterns were analyzed in Siberian sturgeon. The complete coding regions of kiss1 and kiss2 genes were 393 and 471 bp. Both kiss1 and kiss2 showed the highest expression level in the hypothalamus. During the periprandial and fasting experiments, the expression of kiss1 and kiss2 highly significantly increased in the hypothalamus after feeding (P < 0.01). Compared with the feeding group, in hypothalamus, kiss1 expression in the fasting group highly significantly decreased (P < 0.01). In contrast, kiss2 expression had no significant difference on days 1 and 7 (P > 0.05) but highly significantly increased on day 14 (P < 0.01). Subsequently, the feeding function was verified by intraperitoneal (i.p.) injection of Kp1(10) and Kp1(10) into fish. The results showed that i.p. injection of 1 µg/g BW Kp1(10) or 0.01 µg/g BW Kp2(10) could significantly reduce 0-1 h food intake (P < 0.05) and affected the expression levels of apelin, ghrelin, leptin, nmu, etc. in the hypothalamus. These results suggested that kiss1 plays an anorexic role in both short- and long-term feeding regulation, while kiss2 plays a short-term anorexic and long-term orexigenic role. This study described kiss as a novel regulator of appetite in fish and laid the groundwork for further studies focused on physiological function. HIGHLIGHTS: • The kiss1 and kiss2 of Siberian sturgeon were cloned. • The expression levels of kiss1 and kiss2 mRNA were the highest in the hypothalamus. • Postprandial hypothalamic kiss1 and kiss2 expression levels increased in the periprandial experiment. • In the fasting test, the expression of hypothalamic kiss1 decreased after fasting, while the expression of kiss2 increased after fasting on the 14th day. • Siberian sturgeon food intake was reduced, and appetite factors expression levels in the hypothalamus were altered after intraperitoneal injection of Kp1(10) and Kp2(10).
Collapse
Affiliation(s)
- Shaoqi Xu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Mei Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Ya Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Ni Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xin Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Hu Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Shupeng Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yanling Liu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jun Wang
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, Sichuan, People's Republic of China
| | - Defang Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Zhiqiong Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China.
| |
Collapse
|
20
|
Aberrant Notch Signaling Pathway as a Potential Mechanism of Central Precocious Puberty. Int J Mol Sci 2022; 23:ijms23063332. [PMID: 35328752 PMCID: PMC8950842 DOI: 10.3390/ijms23063332] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 02/07/2023] Open
Abstract
The Notch signaling pathway is highly conserved during evolution. It has been well documented that Notch signaling regulates cell proliferation, migration, and death in the nervous, cardiac, and endocrine systems. The Notch pathway is relatively simple, but its activity is regulated by numerous complex mechanisms. Ligands bind to Notch receptors, inducing their activation and cleavage. Various post-translational processes regulate Notch signaling by affecting the synthesis, secretion, activation, and degradation of Notch pathway-related proteins. Through such post-translational regulatory processes, Notch signaling has versatile effects in many tissues, including the hypothalamus. Recently, several studies have reported that mutations in genes related to the Notch signaling pathway were found in patients with central precocious puberty (CPP). CPP is characterized by the early activation of the hypothalamus–pituitary–gonadal (HPG) axis. Although genetic factors play an important role in CPP development, few associated genetic variants have been identified. Aberrant Notch signaling may be associated with abnormal pubertal development. In this review, we discuss the current knowledge about the role of the Notch signaling pathway in puberty and consider the potential mechanisms underlying CPP.
Collapse
|
21
|
Khan HL, Bhatti S, Sehole Z, Younas H, Nathaniel S, Abbas S, Kaloglu C, Ziders R, Yildiz A, Isa AM. Putative Role of the Kisspeptin/Kiss1R System in Promoting Hypothalamic GnRH Release, Pubertal Maturation, and Regulation of Ovulation Considering the Central Reproductive Axis. FERTILITY & REPRODUCTION 2022. [DOI: 10.1142/s2661318222500062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Kisspeptin is a class of neuropeptides that are the product of the Kiss1 gene. These neuropeptides play an important role in maintaining gonadotropin-releasing hormone (GnRH) levels and their release through hypothalamic neurons. Subsequently, they also play an important role in maintaining gonadotropin levels, as GnRH levels stimulate the release of follicle-stimulating hormone (FSH) and luteinizing hormone (LH), which allow induction of gametogenesis of pubertal maturation. The importance of the Kiss1 gene in reproduction became evident when natural mutations in this gene were discovered, which were associated with hypothalamic hypogonadism (HH) and delayed puberty. Kisspeptin and its KISS1R receptors are expressed in the mammalian ovary. The putative role of the Kisspeptin system in the ovary directly controls oocyte maturation, follicular development, and ovulation in an autocrine and paracrine fashion. These essential facts of kisspeptin and its receptor are necessary to maintain the central reproductive axis.
Collapse
Affiliation(s)
- Haroon Latif Khan
- Lahore Institute of Fertility and Endocrinology, Hameed Latif Hospital, Lahore, Pakistan
| | - Shahzad Bhatti
- Lahore Institute of Fertility and Endocrinology, Hameed Latif Hospital, Lahore, Pakistan
| | - Zirva Sehole
- Department of Biochemistry, Kinnaird College for Women, Lahore, Pakistan
| | - Hooria Younas
- Department of Biochemistry, Kinnaird College for Women, Lahore, Pakistan
| | - Sammar Nathaniel
- Department of Biochemistry, Kinnaird College for Women, Lahore, Pakistan
| | - Sana Abbas
- Lahore Institute of Fertility and Endocrinology, Hameed Latif Hospital, Lahore, Pakistan
| | - Celal Kaloglu
- Department of Histology and Embryology, Cumhuriyet University Faculty of Medicine, Sivas, Turkey
| | - Rachel Ziders
- You Family Fertility Buffalo-Niagara Falls Area, University at Buffalo, Buffalo, NY, USA
| | - Aysegul Yildiz
- Department of Molecular Biology and Genetics, Faculty of Science, Mugla Sitki Kocman University, Kotekli, Mugla, Turkey
| | - Ahmed M. Isa
- Assisted Conception Unit, Obstetrics and Gynecology Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
22
|
Xie Q, Kang Y, Zhang C, Xie Y, Wang C, Liu J, Yu C, Zhao H, Huang D. The Role of Kisspeptin in the Control of the Hypothalamic-Pituitary-Gonadal Axis and Reproduction. Front Endocrinol (Lausanne) 2022; 13:925206. [PMID: 35837314 PMCID: PMC9273750 DOI: 10.3389/fendo.2022.925206] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/30/2022] [Indexed: 01/07/2023] Open
Abstract
The discovery of kisspeptin as a critical central regulatory factor of GnRH release has given people a novel understanding of the neuroendocrine regulation in human reproduction. Kisspeptin activates the signaling pathway by binding to its receptor kisspeptin receptor (KISS1R) to promote GnRH secretion, thereby regulating the hypothalamic-pituitary-gonadal axis (HPG) axis. Recent studies have shown that kisspeptin neurons located in arcuate nucleus (ARC) co-express neurokinin B (NKB) and dynorphin (Dyn). Such neurons are called KNDy neurons. KNDy neurons participate in the positive and negative feedback of estrogen to GnRH secretion. In addition, kisspeptin is a key factor in the initiation of puberty, and also regulates the processes of female follicle development, oocyte maturation, and ovulation through the HPG axis. In male reproduction, kisspeptin also plays an important role, getting involved in the regulation of Leydig cells, spermatogenesis, sperm functions and reproductive behaviors. Mutations in the KISS1 gene or disorders of the kisspeptin/KISS1R system may lead to clinical symptoms such as idiopathic hypogonadotropic hypogonadism (iHH), central precocious puberty (CPP) and female infertility. Understanding the influence of kisspeptin on the reproductive axis and related mechanisms will help the future application of kisspeptin in disease diagnosis and treatment. In this review, we critically appraise the role of kisspeptin in the HPG axis, including its signaling pathways, negative and positive feedback mechanisms, and its control on female and male reproduction.
Collapse
Affiliation(s)
- Qinying Xie
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yafei Kang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenlu Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ye Xie
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuxiong Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiang Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Caiqian Yu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hu Zhao
- Department of Human Anatomy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Donghui Huang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Khan S, Batool B, Zubair H, Bano R, Ahmad S, Shahab M. Expression and co-localization of RFRP-3 and kisspeptin during breeding and non-breeding season in the hypothalamus of male rhesus monkey ( Macaca mulatta). Reprod Med Biol 2022; 21:e12479. [PMID: 35847413 PMCID: PMC9270642 DOI: 10.1002/rmb2.12479] [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: 04/28/2022] [Revised: 06/12/2022] [Accepted: 06/21/2022] [Indexed: 12/01/2022] Open
Abstract
Propose The mechanism that underpins how RFRP-3 and kisspeptin interacts are not fully understood in higher primates. This study therefore set out to assess RFRP-3 and kisspeptin expression and their morphological interactions in the breeding, and in the non-breeding period in monkey hypothalamus. Methods Eight mature male macaques (Macaca mulatta) in the breeding season (February; n = 4) and non-breeding season (June; n = 4) were used. To reveal the expression and co-localization of RFRP-3 and kisspeptin, double-labeled immunohistochemistry was performed. Testicular volume, sperm count, and plasma testosterone level were also measured to validate the breeding and non-breeding paradigms. Results Testicular volume, plasma testosterone level, and sperm count showed a significant reduction during non-breeding season. The number of kisspeptin-positive cells was significantly increased during the breeding season (p < 0.05), whereas more RFRP-3-positive cell bodies were seen in the non-breeding season (p < 0.01). Close contacts of RFRP-3 fibers with kisspeptin cells showed no significant difference (p > 0.05) across seasons. However, co-localization of RFRP-3-ir cell bodies onto kisspeptin IR cell bodies showed a statistical increase (p < 0.01) in non-breeding season. Conclusion In higher primates, RFRP-3 decreases kisspeptin drives from the same cells to GnRH neurons in an autocrine manner causing suppression of the reproductive axis during the non-breeding period.
Collapse
Affiliation(s)
- Safdar Khan
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Bakhtwar Batool
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Hira Zubair
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Riffat Bano
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Shakil Ahmad
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Muhammad Shahab
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
- Shaheed Benazir Bhutto University, SheringalDir UpperPakistan
| |
Collapse
|
24
|
Abstract
Pubertal onset is a complex process, which is influenced by genetic and environmental factors, such as obesity and endocrine-disrupting chemicals. In addition, the timing of normal puberty varies between individuals and is a highly polygenic trait with both rare and common variants. Central precocious puberty (CPP) is defined as the early activation of the hypothalamic-pituitary-gonadal axis. Genetic factors are suggested to account for 50% to 80% of the variation in puberty initiation, as indicated by the greater concordance of pubertal timing observed in monozygotic twins than in dizygotic twins. Although genetic factors play a crucial role in CPP development, only few associated genes have been identified. To date, four monogenic genes have been identified: KISS1, KISS1R, MKRN3, and DLK1. Moreover, mutation prevalence in these genes varies considerably depending on the ethnicity of patients with CPP. This article reviews the current knowledge on the normal pubertal timing and physiology and discusses the CPP-causing genes.
Collapse
|
25
|
Chaudhary H, Patel J, Jain NK, Joshi R. The role of polymorphism in various potential genes on polycystic ovary syndrome susceptibility and pathogenesis. J Ovarian Res 2021; 14:125. [PMID: 34563259 PMCID: PMC8466925 DOI: 10.1186/s13048-021-00879-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/09/2021] [Indexed: 12/27/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrinopathies affecting the early reproductive age in women, whose pathophysiology perplexes many researchers till today. This syndrome is classically categorized by hyperandrogenism and/or hyperandrogenemia, menstrual and ovulatory dysfunction, bulky multi follicular ovaries on Ultrasonography (USG), and metabolic abnormalities such as hyperinsulinemia, dyslipidemia, obesity. The etiopathogenesis of PCOS is not fully elucidated, but it seems that the hypothalamus-pituitary-ovarian axis, ovarian, and/or adrenal androgen secretion may contribute to developing the syndrome. Infertility and poor reproductive health in women's lives are highly associated with elevated levels of androgens. Studies with ovarian theca cells taken from PCOS women have demonstrated increased androgen production due to augmented ovarian steroidogenesis attributed to mainly altered expression of critical enzymes (Cytochrome P450 enzymes: CYP17, CYP21, CYP19, CYP11A) in the steroid hormone biosynthesis pathway. Despite the heterogeneity of PCOS, candidate gene studies are the widely used technique to delineate the genetic variants and analyze for the correlation of androgen biosynthesis pathway and those affecting the secretion or action of insulin with PCOS etiology. Linkage and association studies have predicted the relationship between genetic variants and PCOS risk among families or populations. Several genes have been proposed as playing a role in the etiopathogenesis of PCOS, and the presence of mutations and/or polymorphisms has been discovered, which suggests that PCOS has a vital heritable component. The following review summarizes the influence of polymorphisms in crucial genes of the steroidogenesis pathway leading to intraovarian hyperandrogenism which can result in PCOS.
Collapse
Affiliation(s)
- Hiral Chaudhary
- Department of Biochemistry and Forensic Science, University School of Sciences, Gujarat University, Ahmedabad, Gujarat 380009 India
| | - Jalpa Patel
- Department of Biochemistry and Forensic Science, University School of Sciences, Gujarat University, Ahmedabad, Gujarat 380009 India
| | - Nayan K. Jain
- Department of Life Science, University School of Sciences, Gujarat University, Ahmedabad, Gujarat 380009 India
| | - Rushikesh Joshi
- Department of Biochemistry and Forensic Science, University School of Sciences, Gujarat University, Ahmedabad, Gujarat 380009 India
| |
Collapse
|
26
|
Sengupta P, Dutta S, Karkada IR, Akhigbe RE, Chinni SV. Irisin, Energy Homeostasis and Male Reproduction. Front Physiol 2021; 12:746049. [PMID: 34621189 PMCID: PMC8490744 DOI: 10.3389/fphys.2021.746049] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/31/2021] [Indexed: 11/17/2022] Open
Abstract
Irisin is a novel skeletal muscle- and adipose tissue-secreted peptide. It is conventionally regarded as an adipomyokine and is a cleaved fragment of Fibronectin type III domain-containing protein 5 (FNDC5). It is involved in the browning of white adipose tissue, glucose tolerance, and reversing of metabolic disruptions. Fertility is closely linked to energy metabolism and the endocrine function of the adipose tissue. Moreover, there is established association between obesity and male infertility. Irisin bears strong therapeutic promise in obesity and its associated disorders, as well as shown to improve male reproductive functions. Thus, irisin is a molecule of great interest in exploring the amelioration of metabolic syndrome or obesity-induced male infertility. In this review we aim to enumerate the most significant aspects of irisin actions and discuss its involvement in energy homeostasis and male reproduction. Though current and future research on irisin is very promiscuous, a number of clarifications are still needed to reveal its full potential as a significant medicinal target in several human diseases including male infertility.
Collapse
Affiliation(s)
- Pallav Sengupta
- Physiology Unit, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Kuala Lumpur, Malaysia
| | - Sulagna Dutta
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Kuala Lumpur, Malaysia
| | - Ivan Rolland Karkada
- Physiology Unit, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Kuala Lumpur, Malaysia
| | - Roland Eghoghosoa Akhigbe
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
- Reproductive Biology and Toxicology Research Laboratories, Oasis of Grace Hospital, Osogbo, Nigeria
| | - Suresh V. Chinni
- Department of Biotechnology, Faculty of Applied Sciences, AIMST University, Bedong, Malaysia
| |
Collapse
|
27
|
Padda J, Khalid K, Moosa A, Syam M, Kakani V, Imdad U, Ismail D, Cooper AC, Jean-Charles G. Role of Kisspeptin on Hypothalamic-Pituitary-Gonadal Pathology and Its Effect on Reproduction. Cureus 2021; 13:e17600. [PMID: 34646652 PMCID: PMC8482951 DOI: 10.7759/cureus.17600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2021] [Indexed: 01/25/2023] Open
Abstract
Kisspeptin is a neuropeptide that plays a significant role in human reproduction by its action on the hypothalamic-pituitary-gonadal (HPG) axis and functions through a G-protein-coupled receptor called G-protein-coupled receptor 54/kisspeptin 1 receptor (GPR54/KISS1R). It is encoded by the kisspeptin 1 (KISS1) gene that is mainly expressed in the hypothalamus. Kisspeptins are also recognized as vital aspects of maturation and proper function of the reproductive system in both males and females. It also plays its role in the onset of puberty, sexual patterns, desires, ovum development in women, sperm quality in men, feedback mechanisms, pregnancy, and lactation. Studies proved the pathological role of kisspeptin dysregulation in disorders like polycystic ovarian syndrome (PCOS) and infertility. Mutations in the KISS1 gene also contribute to precocious puberty or hypogonadotropic hypogonadism, depending upon the nature of mutations. Levels of kisspeptin also aid in the identification of a few pregnancy-related complications like preeclampsia, intrauterine growth restriction, and act as a marker of miscarriage. Due to the wide range of effects that kisspeptin has on the reproductive axis, investigations are being carried out to develop it as a diagnostic marker, treat diseases like hypogonadism and PCOS, and solve infertility issues.
Collapse
Affiliation(s)
| | | | - Amir Moosa
- Internal Medicine, JC Medical Center, Orlando, USA
| | | | | | - Urooj Imdad
- Internal Medicine, JC Medical Center, Orlando, USA
| | - Dina Ismail
- Internal Medicine, JC Medical Center, Orlando, USA
| | | | - Gutteridge Jean-Charles
- Internal Medicine, JC Medical Center, Orlando, USA
- Internal Medicine, Advent Health & Orlando Health Hospital, Orlando, USA
| |
Collapse
|
28
|
Clinical Potential of Kisspeptin in Reproductive Health. Trends Mol Med 2021; 27:807-823. [PMID: 34210598 DOI: 10.1016/j.molmed.2021.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 12/31/2022]
Abstract
Kisspeptins are a family of hypothalamic neuropeptides that are essential for the regulation of reproductive physiology. Their importance in reproductive health became apparent in 2003, when loss-of-function variants in the gene encoding the kisspeptin receptor were reported to result in isolated congenital hypogonadotropic hypogonadism (CHH). It has since been ascertained that hypothalamic kisspeptin neurons regulate gonadotropin-releasing hormone (GnRH) secretion to thus stimulate the remainder of the reproductive endocrine axis. In this review, we discuss genetic variants that affect kisspeptin receptor signaling, summarize data on KISS1R agonists, and posit possible clinical uses of native and synthetic kisspeptin receptor agonists for the investigation and treatment of reproductive disorders.
Collapse
|
29
|
Gomes VCL, Sones JL. From inhibition of trophoblast cell invasion to proapoptosis: what are the potential roles of kisspeptins in preeclampsia? Am J Physiol Regul Integr Comp Physiol 2021; 321:R41-R48. [PMID: 34009045 DOI: 10.1152/ajpregu.00258.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preeclampsia (PE) is a life-threatening human gestational syndrome with incompletely understood etiopathogenesis. The disorder has a spectrum of clinical features, likely due to a complex interaction between maternal predisposing factors and abnormalities at the maternal-fetal interface. Poor trophoblast cell invasion, inadequate uterine vascular remodeling, and placental hypoperfusion are considered as key placental events leading to PE. Kisspeptins, a family of small peptides derived from the KISS1 gene, have been implicated in the development of this syndrome. Most studies of kisspeptin expression in PE have reported an upregulation of kisspeptins and/or their cognate receptor in preeclamptic placentas. Conversely, maternal peripheral blood concentration of kisspeptins is reportedly lower in PE than in uncomplicated pregnancies. This apparent paradox remains to be further elucidated. Although kisspeptins were initially known for inhibiting cellular migration and invasion, other biological activities attributed to these peptides include neuroendocrine regulation of reproduction, metabolism regulation, inhibition of angiogenesis, and induction of apoptosis. This review summarizes the current knowledge on expression and biological activity of kisspeptins at the maternal-fetal interface in the context of PE.
Collapse
Affiliation(s)
- Viviane C L Gomes
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Jenny L Sones
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| |
Collapse
|
30
|
Pérez-López FR, Ornat L, López-Baena MT, Santabárbara J, Savirón-Cornudella R, Pérez-Roncero GR. Circulating kisspeptin and anti-müllerian hormone levels, and insulin resistance in women with polycystic ovary syndrome: A systematic review, meta-analysis, and meta-regression. Eur J Obstet Gynecol Reprod Biol 2021; 260:85-98. [PMID: 33744505 DOI: 10.1016/j.ejogrb.2021.03.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE This systematic review and meta-analysis aimed to summarize the available evidence regarding circulating kisspeptin and anti-müllerian hormone (AMH) and the homeostasis model assessment of insulin resistance (HOMA-IR) index in adolescents and women with and without polycystic ovary syndrome (PCOS). METHOD We performed a comprehensive literature search in Medline, Embase, Cochrane, Scopus, and Web of Science for studies evaluating circulating kisspeptin levels in women with and without PCOS published until September 24th, 2020. Co-primary outcomes were the HOMA-IR index and AMH. The quality of included studies was assessed using the Newcastle-Ottawa Scale. Random-effects models were used to estimate outcomes, and effects reported as mean difference (MD) or standardized MD (SMD) and their 95 % confidence interval (CI). The systematic review and meta-analysis was registered in the International Prospective Register of Systematic Reviews (PROSPERO) as number CRD42020205030. RESULTS We evaluated 18 studies including, 1282 PCOS cases and 977 controls. Participants with PCOS were younger (MD = -2.38 years, 95 %CI -4.32 to -0.44), with higher BMI (MD = 1.16, 95 % CI 0.54-1.78), waist-to-hip ratio (MD = 0.04, 95 %CI 0.02 to 0.05), circulating kisspeptin (SMD = 1.15, 95 %CI 0.68-1.62), luteinizing hormone (SMD = 1.29, 95 %CI 0.76-1.83), AMH (SMD = 0.97, 95 %CI 0.60-1,34), total testosterone (SMD = 2.48, 95 %CI 1.73-3.23), free testosterone (SMD = 1.37, 95 %CI 0.56-2.17), and dehydroepiandrosterone sulphate (SMD = 0.72, 95 %CI 0.32-1.13) levels, and Ferriman-Gallwey score (SMD = 5.08, 95 %CI 2.76-7.39), and lower sex hormone-binding globulin level (SMD = -1.34, 95 %CI -2.15 to -0.52). Besides, participants with PCOS had higher HOMA-IR index (SMD = 0.76, 95 %CI 0.35-1.17), and circulating insulin (SMD = 0.75, 95 %CI 0.30-1.19), leptin (SMD = 2.82, 95 %CI 1.35-4.29), and triglycerides (SMD = 2.15, 95 %CI 1.08-3.23) levels than participants without the syndrome. The meta-regression did not identify significant factors influencing circulating kisspeptin. CONCLUSION Patients with PCOS showed higher kisspeptin, LH, insulin, AMH, and androgen levels and HOMA-IR index, and lower sex hormone-binding globulin levels than those without the syndrome.
Collapse
Affiliation(s)
- Faustino R Pérez-López
- Department of Obstetrics and Gynecology, University of Zaragoza Faculty of Medicine, Zaragoza, 50009, Spain; Aragón Health Research Institute, Zaragoza, 50009, Spain.
| | - Lía Ornat
- Department of Obstetrics and Gynecology, University of Zaragoza Faculty of Medicine, Zaragoza, 50009, Spain
| | | | - Javier Santabárbara
- Aragón Health Research Institute, Zaragoza, 50009, Spain; Department of Microbiology, Preventive Medicine and Public Health, University of Zaragoza Faculty of Medicine, Zaragoza, 50009, Spain
| | | | | |
Collapse
|
31
|
Gorbunova OL, Shirshev SV. Role of Kisspeptin in Regulation of Reproductive and Immune Reactions. BIOCHEMISTRY (MOSCOW) 2021; 85:839-853. [PMID: 33045946 DOI: 10.1134/s0006297920080015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The work is focused on physiological role of the hormone kisspeptin produced by neurons of the hypothalamus anterior zone, which is a key regulator of reproduction processes. Role of the hormone in transmission of information on metabolic activity and induction of the secretion of gonadotropin-releasing hormone (GnRH) by the hypothalamus that determines gestation processes involving fertilization, placentation, fetal development, and child birth is considered. The literature data on molecular mechanisms and effects of kisspeptin on reproductive system including puberty initiation are summarized and analyzed. In addition, attention is paid to hormone-mediated changes in the cardiovascular system in pregnant women. For the first time, the review examines the effect of kisspeptin on functional activity of immune system cells presenting molecular mechanisms of the hormone signal transduction on the level of lymphoid cells that lead to the immune tolerance induction. In conclusion, a conceptual model is presented that determines the role of kisspeptin as an integrator of reproductive and immune functions during pregnancy.
Collapse
Affiliation(s)
- O L Gorbunova
- Perm Federal Research Center, Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences, Perm, 614081, Russia.
| | - S V Shirshev
- Perm Federal Research Center, Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences, Perm, 614081, Russia
| |
Collapse
|
32
|
Siahpoosh Z, Farsimadan M, Pazhohan M, Vaziri H, Mahmoudi Gomari M. KISS1R polymorphism rs587777844 (Tyr313His) is linked to female infertility. Br J Biomed Sci 2021; 78:98-100. [PMID: 33275081 DOI: 10.1080/09674845.2020.1856496] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Z Siahpoosh
- Department of Biology, Faculty of Sciences, University of Guilan , Rasht, Iran
| | - M Farsimadan
- Department of Biology, Faculty of Sciences, University of Guilan , Rasht, Iran
| | - M Pazhohan
- Department of Medical Microbiology Faculty of Medicine, Hacettepe University , Ankara, Turkey
| | - H Vaziri
- Department of Biology, Faculty of Sciences, University of Guilan , Rasht, Iran
| | - M Mahmoudi Gomari
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences , Tehran, Iran
| |
Collapse
|
33
|
Petrucci L, Maranesi M, Verini Supplizi A, Dall’Aglio C, Mandara MT, Quassinti L, Bramucci M, Miano A, Gobbetti A, Catone G, Boiti C, Zerani M. Kisspeptin/GnRH1 system in Leydig cells of horse (Equus caballus): Presence and function. Theriogenology 2020; 152:1-7. [DOI: 10.1016/j.theriogenology.2020.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 12/25/2022]
|
34
|
Zhu N, Zhao M, Song Y, Ding L, Ni Y. The KiSS-1/GPR54 system: Essential roles in physiological homeostasis and cancer biology. Genes Dis 2020; 9:28-40. [PMID: 35005105 PMCID: PMC8720660 DOI: 10.1016/j.gendis.2020.07.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/29/2022] Open
Abstract
KiSS-1, first identified as an anti-metastasis gene in melanoma, encodes C-terminally amidated peptide products, including kisspeptin-145, kisspeptin-54, kisspeptin-14, kisspeptin-13 and kisspeptin-10. These products are endogenous ligands coupled to G protein-coupled receptor 54 (GPR54)/hOT7T175/AXOR12. To date, the regulatory activities of the KiSS-1/GPR54 system, such as puberty initiation, antitumor metastasis, fertility in adulthood, hypothalamic-pituitary-gonadal axis (HPG axis) feedback, and trophoblast invasion, have been investigated intensively. Accumulating evidence has demonstrated that KiSS-1 played a key role in reproduction and served as a promising biomarker relative to the diagnosis, identification of therapeutic targets and prognosis in various carcinomas, while few studies have systematically summarized its subjective factors and concluded the functions of KiSS-1/GPR54 signaling in physiology homeostasis and cancer biology. In this review, we retrospectively summarized the regulators of the KiSS-1/GPR54 system in different animal models and reviewed its functions according to physiological homeostasis regulations and above all, cancer biology, which provided us with a profound understanding of applying the KiSS-1/GPR54 system into medical applications.
Collapse
Affiliation(s)
- Nisha Zhu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu 210008, PR China
| | - Mengxiang Zhao
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu 210008, PR China
| | - Yuxian Song
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu 210008, PR China
| | - Liang Ding
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu 210008, PR China
| | - Yanhong Ni
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu 210008, PR China
| |
Collapse
|
35
|
Watanabe T, Sato K. Roles of the kisspeptin/GPR54 system in pathomechanisms of atherosclerosis. Nutr Metab Cardiovasc Dis 2020; 30:889-895. [PMID: 32409274 DOI: 10.1016/j.numecd.2020.02.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/11/2019] [Accepted: 02/28/2020] [Indexed: 12/11/2022]
Abstract
AIMS Kisspeptin-10 (KP-10), a potent vasoconstrictor and inhibitor of angiogenesis, and its receptor, GPR54, have currently received much attention with respect to atherosclerosis, since both KP-10 and GPR54 are expressed at high levels in atheromatous plaques and restenotic lesions after wire-injury. The present review introduces the emerging roles of the KP-10/GPR54 system in atherosclerosis. DATA SYNTHESIS KP-10 suppresses migration and proliferation of human umbilical vein endothelial cells (HUVECs), and induces senescence in HUVECs. KP-10 increases adhesion of human monocytes to HUVECs. KP-10 also stimulates expression of interleukin-6, tumor necrosis factor-α, monocyte chemotactic protein-1, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and E-selectin genes in HUVECs. KP-10 enhances oxidized low-density lipoprotein-induced foam cell formation associated with upregulation of CD36 and acyl-coenzyme A: cholesterol acyltransferase-1 in human monocyte-derived macrophages. In human aortic smooth muscle cells, KP-10 suppresses angiotensin II-induced migration and proliferation, however, it enhances apoptosis and activities of matrix metalloproteinase (MMP)-2 and MMP-9 by upregulation of extracellular signal-regulated kinase 1/2, p38, Bax, and caspase-3. Four-week-infusion of KP-10 into Apoe-/- mice accelerates development of aortic atherosclerotic lesions with increased monocyte/macrophage infiltration and vascular inflammation, also, it decreases intraplaque vascular smooth muscle cell content. Proatherosclerotic effects of endogenous and exogenous KP-10 were completely attenuated upon infusion of P234, a GPR54 antagonist, in Apoe-/- mice. CONCLUSION These findings suggest that KP-10 may contribute to acceleration of progression and to the instability of atheromatous plaques, leading to rupture of plaques. This GPR54 antagonist may be useful for the prevention and treatment of atherosclerosis. Thus, the KP-10/GPR54 system may serve as a novel therapeutic target for atherosclerotic diseases.
Collapse
Affiliation(s)
- Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan; Department of Internal Medicine, Ushioda General Hospital/Clinic, Yokohama, Japan.
| | - Kengo Sato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan; Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| |
Collapse
|
36
|
Association of polymorphisms in metastasis suppressor genes NME1 and KISS1 with breast cancer development and metastasis. J Egypt Natl Canc Inst 2020; 32:24. [PMID: 32462300 DOI: 10.1186/s43046-020-00037-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 05/10/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND NME1 and KISS1 genes are two tumor metastasis suppressor genes, mapped to chromosomes 17q21.3 and 1q32 respectively. Here, we analyzed the association of EcoR1 (rs34214448-G/T) polymorphism in NME1 gene and 9 del T (rs5780218-A/-) polymorphism in KISS1 gene with breast cancer development and metastasis. RESULTS The study included 75 women newly diagnosed with breast cancer recruited from Oncology Center at Mansoura University Hospitals and 37 age-matched healthy female volunteers as a control group. DNA was extracted from peripheral blood samples and genotyping of rs34214448 and rs5780218 SNPs was carried out by PCR-RFLP technique. NME1 EcoR1 (rs34214448) polymorphism has a statistically significant association with breast cancer risk (P < 0.001). Most of breast cancer group (55%) had heterozygous (G/T) genotype while most of control group (95%) had homozygous wild (G/G) genotype (P < 0.0005). Also, KISS1 rs5780218 polymorphism has a statistically significant association with breast cancer risk. The wild (A/A) genotype was associated with lower risk of breast cancer (A/- + -/- vs. A/A: OR = 3.1, 95% CI = 1.15-8.36, P = 0.025). EcoR1 (rs34214448) polymorphism revealed a significant association with tumor stage and distant metastasis as patients. Carriers of the wild (G/G) genotype were more likely to present with advanced disease stages and distant metastasis. CONCLUSIONS Both EcoR1 (rs34214448) polymorphism of NME1 gene and rs5780218 polymorphism of KISS1 gene revealed significant association with increased risk of breast cancer development. The (G/G) genotype of EcoR1 polymorphism was associated with higher risk of breast cancer metastasis.
Collapse
|
37
|
Hu KL, Chang HM, Zhao HC, Yu Y, Li R, Qiao J. Potential roles for the kisspeptin/kisspeptin receptor system in implantation and placentation. Hum Reprod Update 2020; 25:326-343. [PMID: 30649364 PMCID: PMC6450039 DOI: 10.1093/humupd/dmy046] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/19/2018] [Accepted: 12/09/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Initially identified as suppressors of metastasis in various types of cancer, kisspeptins are a family of neuropeptides that are key regulators of the mammalian reproductive axis. Accumulating evidence has shown that kisspeptin is able to control both the pulsatile and surge GnRH release, playing fundamental roles in female reproduction, which include the secretion of gonadotropins, puberty onset, brain sex differentiation, ovulation and the metabolic regulation of fertility. Furthermore, recent studies have demonstrated the involvement of the kisspeptin system in the processes of implantation and placentation. This review summarizes the current knowledge of the pathophysiological role and utility of these local placental regulatory factors as potential biomarkers during the early human gestation. OBJECTIVE AND RATIONALE A successful pregnancy, from the initiation of embryo implantation to parturition, is a complex process that requires the orchestration of a series of events. This review aims to concisely summarize what is known about the role of the kisspeptin system in implantation, placentation, early human pregnancy and pregnancy-related disorders, and to develop strategies for predicting, diagnosing and treating these abnormalities. SEARCH METHODS Using the PubMed and Google Scholar databases, we performed comprehensive literature searches in the English language describing the advancement of kisspeptins and the kisspeptin receptor (KISS1R) in implantation, placentation and early pregnancy in humans, since its initial identification in 1996 and ending in July 2018. OUTCOMES Recent studies have shown the coordinated spatial and temporal expression patterns of kisspeptins and KISS1R during human pregnancy. The experimental data gathered recently suggest putative roles of kisspeptin signaling in the regulation of trophoblast invasion, embryo implantation, placentation and early pregnancy. Dysregulation of the kisspeptin system may negatively affect the processes of implantation as well as placentation. Clinical studies indicate that the circulating levels of kisspeptins or the expression levels of kisspeptin/KISS1R in the placental tissues may be used as potential diagnostic markers for women with miscarriage and gestational trophoblastic neoplasia. WIDER IMPLICATIONS Comprehensive research on the pathophysiological role of the kisspeptin/KISS1R system in implantation and placentation will provide a dynamic and powerful approach to understanding the processes of early pregnancy, with potential applications in observational and analytic screening as well as the diagnosis, prognosis and treatment of implantation failure and early pregnancy-related disorders.
Collapse
Affiliation(s)
- Kai-Lun Hu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
| | - Hsun-Ming Chang
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
| | - Hong-Cui Zhao
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
| | - Yang Yu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China.,National Clinical Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Rong Li
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China.,National Clinical Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Jie Qiao
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China.,National Clinical Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
38
|
Szeliga A, Podfigurna A, Bala G, Meczekalski B. Kisspeptin and neurokinin B analogs use in gynecological endocrinology: where do we stand? J Endocrinol Invest 2020; 43:555-561. [PMID: 31838714 DOI: 10.1007/s40618-019-01160-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/09/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Recent studies have found that kisspeptin/neurokinin B/dynorphin neurons (KNDy neurons) in the infundibular nucleus play a crucial role in the reproductive axis. Analogs, both agonists and antagonists, of kisspeptin and neurokinin B (NKB) are particularly important in explaining the physiological role of KNDy in the reproductive axis in animals. The use of kisspeptin and NKB analogs has helped elucidate the regulators of the hypothalamic reproductive axis. PURPOSE This review describes therapeutic uses of Kiss-1 and NKB agonists, most obviously the use of kisspeptin agonists in the treatment for infertility and the induction of ovulation. Kisspeptin antagonists may have potential clinical applications in patients suffering from diseases associated with enhanced LH pulse frequency, such as polycystic ovary syndrome or menopause. The inhibition of pubertal development using Kiss antagonists may be used as a therapeutic option in precocious puberty. Kisspeptin antagonists have been found capable of inhibiting ovulation and have been proposed as novel contraceptives. Hypothalamic amenorrhea and delayed puberty are conditions in which normalization of LH secretion may potentially be achieved by treatment with both kisspeptin and NKB agonists. NKB antagonists are used to treat vasomotor symptoms in postmenopausal women, providing rapid relief of symptoms while supplanting the need for exogenous estrogen exposure. CONCLUSIONS There is a wide spectrum of therapeutic uses of Kiss-1 and NKB agonists, including the management of infertility, treatment for PCOS, functional hypothalamic amenorrhea or postmenopausal vasomotor symptoms, as well as contraceptive issues. Nevertheless, further research is needed before kisspeptin and NKB analogs are fully incorporated in clinical practice.
Collapse
Affiliation(s)
- A Szeliga
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 33 Polna Street, 60-535, Poznan, Poland
| | - A Podfigurna
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 33 Polna Street, 60-535, Poznan, Poland
| | - G Bala
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 33 Polna Street, 60-535, Poznan, Poland
| | - B Meczekalski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 33 Polna Street, 60-535, Poznan, Poland.
| |
Collapse
|
39
|
Martins Trevisan C, Naslavsky MS, Monfardini F, Wang J, Zatz M, Peluso C, Pellegrino R, Mafra F, Hakonarson H, Ferreira FM, Nakaya H, Christofolini DM, Montagna E, Crandall KA, Barbosa CP, Bianco B. Variants in the Kisspeptin-GnRH Pathway Modulate the Hormonal Profile and Reproductive Outcomes. DNA Cell Biol 2020; 39:1012-1022. [PMID: 32352843 DOI: 10.1089/dna.2019.5165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Kisspeptin has been identified as a key regulatory protein in the release of gonadotropin-releasing hormone (GnRH), which subsequently increases gonadotropin secretion during puberty to establish reproductive function and regulate the hypothalamic-pituitary-gonadal axis. The effects of variants in the KISS1, KISS1R, and GNRHR genes and their possible association with assisted reproduction outcomes remain to be elucidated. In this study, we used next-generation sequencing to investigate the associations of the genetic diversity at the candidate loci for KISS1, KISS1R, and GNRHR with the hormonal profiles and reproductive outcomes in 86 women who underwent in vitro fertilization treatments. Variants in the KISS1 and KISS1R genes were associated with luteinizing hormone (rs35431622:T>C), anti-Mullerian hormone (rs71745629delT), follicle-stimulating hormone (rs73507529:C>A), and estradiol (rs73507527:G>A, rs350130:A>G, and rs73507529:C>A) levels, as well as with reproductive outcomes such as the number of oocytes retrieved (s35431622:T>C), metaphasis II oocytes (rs35431622:T>C), and embryos (rs1132506:G>C). Additionally, variants in the GNRHR UTR3' (rs1038426:C>A, rs12508464:A>C, rs13150734:C>A, rs17635850:A>G, rs35683646:G>A, rs35610027:C>G, rs35845954:T>C, rs17635749:C>T, and rs7666201:C>T) were associated with low prolactin levels. A conjoint analysis of clinical, hormonal, and genetic variables using a generalized linear model identified two variants of the KISS1 gene (rs71745629delT and rs1132506:G>C) that were significantly associated with hormonal variations and reproductive outcomes. The findings suggest that variants in KISS1, KISS1R, and GNRHR genes can modulate hormone levels and reproductive outcomes.
Collapse
Affiliation(s)
- Camila Martins Trevisan
- Discipline of Sexual and Reproductive Health and Populational Genetics, Department of Collective Health, Centro Universitário Saúde ABC, FMABC, Santo André, São Paulo, Brazil
| | - Michel Satya Naslavsky
- Human Genome and Stem Cell Research Center, Biosciences Institute, Universidade de São Paulo, São Paulo, Brazil
| | - Frederico Monfardini
- Human Genome and Stem Cell Research Center, Biosciences Institute, Universidade de São Paulo, São Paulo, Brazil
| | - Jaqueline Wang
- Human Genome and Stem Cell Research Center, Biosciences Institute, Universidade de São Paulo, São Paulo, Brazil
| | - Mayana Zatz
- Human Genome and Stem Cell Research Center, Biosciences Institute, Universidade de São Paulo, São Paulo, Brazil
| | - Carla Peluso
- Discipline of Sexual and Reproductive Health and Populational Genetics, Department of Collective Health, Centro Universitário Saúde ABC, FMABC, Santo André, São Paulo, Brazil
| | - Renata Pellegrino
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Fernanda Mafra
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Frederico Moraes Ferreira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Helder Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Denise Maria Christofolini
- Discipline of Sexual and Reproductive Health and Populational Genetics, Department of Collective Health, Centro Universitário Saúde ABC, FMABC, Santo André, São Paulo, Brazil
| | - Erik Montagna
- Postgraduation Program in Health Sciences, Research and Innovation, Centro Universitário Saúde ABC, FMABC, Santo André, São Paulo, Brazil
| | - Keith A Crandall
- Computational Biology Institute, Milken Institute School of Public Health, George Washington University, Washington, District of Columbia, USA
| | - Caio Parente Barbosa
- Discipline of Sexual and Reproductive Health and Populational Genetics, Department of Collective Health, Centro Universitário Saúde ABC, FMABC, Santo André, São Paulo, Brazil
| | - Bianca Bianco
- Discipline of Sexual and Reproductive Health and Populational Genetics, Department of Collective Health, Centro Universitário Saúde ABC, FMABC, Santo André, São Paulo, Brazil
| |
Collapse
|
40
|
Harihar S, Ray S, Narayanan S, Santhoshkumar A, Ly T, Welch DR. Role of the tumor microenvironment in regulating the anti-metastatic effect of KISS1. Clin Exp Metastasis 2020; 37:209-223. [PMID: 32088827 PMCID: PMC7339126 DOI: 10.1007/s10585-020-10030-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 02/19/2020] [Indexed: 12/29/2022]
Abstract
KISS1, a metastasis suppressor gene, has been shown to block metastasis without affecting primary tumor formation. Loss of KISS1 leads to invasion and metastasis in multiple cancers, which is the leading cause of cancer morbidity and mortality. The discovery of KISS1 has provided a ray of hope for early clinical diagnosis and for designing effective treatments targeting metastatic cancer. However, this goal requires greater holistic understanding of its mechanism of action. In this review, we go back into history and highlight some key developments, from the discovery of KISS1 to its role in regulating multiple physiological processes including cancer. We discuss key emerging roles for KISS1, specifically interactions with tissue microenvironment to promote dormancy and regulation of tumor cell metabolism, acknowledged as some of the key players in tumor progression and metastasis. We finally discuss strategies whereby KISS1 might be exploited clinically to treat metastasis.
Collapse
Affiliation(s)
- Sitaram Harihar
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| | - Srijit Ray
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Samyukta Narayanan
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Anirudh Santhoshkumar
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Thuc Ly
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
- The University Kansas Cancer Center, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
| | - Danny R Welch
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
- The University Kansas Cancer Center, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
| |
Collapse
|
41
|
Immunoreactivity of Kisspeptin and Kisspeptin Receptor in Eutopic and Ectopic Endometrial Tissue of Women With and Without Endometriosis. Reprod Sci 2020; 27:1731-1741. [PMID: 32072605 DOI: 10.1007/s43032-020-00167-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 01/13/2020] [Indexed: 01/10/2023]
Abstract
Endometriosis is characterized by the presence of ectopic endometrial tissues. Mechanisms of tissue dissemination in endometriosis may be similar to those involved in tumor metastasis. We hypothesize that dysregulation of kisspeptin (KISS1), a metastasis suppressor in endometrial carcinoma, may contribute to the pathogenesis of endometriosis. In this study, we characterized the immunoreactivity of kisspeptin and its receptor, KISS1R, in eutopic and ectopic endometrial tissue of women with and without endometriosis, in proliferative and secretory menstrual cycle phases. Immunohistochemistry was performed using KISS1 and KISS1R antibodies on samples from women with (n = 35) and without (n = 14) endometriosis. Samples from women with endometriosis included eutopic endometrium (n = 20) samples, superficial endometriotic implants (SUP, n = 10) deep infiltrating endometriotic implants (DIE, n = 15), and ovarian endometriomas (OMA, n = 15). Immunoreactivity was quantified using histoscores. KISS1 and KISS1R immunoreactivity was significantly lower in eutopic endometrial stroma of women with versus without endometriosis, regardless of the menstrual cycle phase (P = 0.001 and P = 0.015 respectively). In endometriotic implants, KISS1 levels were significantly lower in both glandular and stromal components of DIE (P < 0.01) and OMA (P < 0.01) compared to SUP. KISS1R immunoreactivity was lower in the glandular component of OMA (P = 0.035) compared to SUP. KISS1 and KISS1R levels are lower in eutopic endometrial stroma from women with versus without endometriosis, consistent with a role for decreased KISS1 expression in the pathogenesis of endometriosis. As deeply invasive lesions showed lower KISS1 levels than superficial lesions, downregulation of KISS1 levels may contribute to implant invasiveness.
Collapse
|
42
|
Ke R, Ma X, Lee LTO. Understanding the functions of kisspeptin and kisspeptin receptor (Kiss1R) from clinical case studies. Peptides 2019; 120:170019. [PMID: 30339828 DOI: 10.1016/j.peptides.2018.09.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/21/2018] [Accepted: 09/21/2018] [Indexed: 10/28/2022]
Abstract
It is widely acknowledged that kisspeptin and its receptor Kiss1R play central regulatory roles in the hypothalamus-pituitary-gonad (HPG) axis and reproduction. Mutations of KISS1 and KISS1R lead to disorders associated with pubertal development, such as central precocious puberty (CPP) and idiopathic hypogonadotropic hypogonadism (IHH). This review focuses on KISS1 and KISS1R mutations found in CPP and IHH and its purposes are twofold: Firstly, based on the mutations found in KISS1 and KISS1R, this review provides insights into the precise mechanism of kisspeptin and the kisspeptin/Kiss1R pathway in the reproductive axis and in puberty. Secondly, G protein-coupled receptors (GPCRs) are known to share highly conserved structural motifs; therefore, knowledge of mutations found at different structural domains of Kiss1R in the diseased state, and how they affect Kiss1R function can be used to decipher GPCR domain function.
Collapse
Affiliation(s)
- Ran Ke
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Xin Ma
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Leo T O Lee
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
43
|
Stathaki M, Stamatiou ME, Magioris G, Simantiris S, Syrigos N, Dourakis S, Koutsilieris M, Armakolas A. The role of kisspeptin system in cancer biology. Crit Rev Oncol Hematol 2019; 142:130-140. [PMID: 31401420 DOI: 10.1016/j.critrevonc.2019.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 02/01/2019] [Accepted: 07/18/2019] [Indexed: 02/08/2023] Open
Abstract
Kisspeptins are a family of neuropeptides that are known to be critical in puberty initiation and ovulation. Apart from that kisspeptin derived peptides (KPs) are also known for their antimetastatic activities in several malignancies. Herein we report recent evidence of the role of kisspeptins in cancer biology and we examine the prospective of targeting the kisspeptin pathways leading to a better prognosis in patients with malignant diseases.
Collapse
Affiliation(s)
- Martha Stathaki
- Physiology Laboratory, Athens Medical School, National and Kapodestrian University of Athens, Greece
| | - Maria Evanthia Stamatiou
- Physiology Laboratory, Athens Medical School, National and Kapodestrian University of Athens, Greece
| | - George Magioris
- Physiology Laboratory, Athens Medical School, National and Kapodestrian University of Athens, Greece
| | - Spyridon Simantiris
- Physiology Laboratory, Athens Medical School, National and Kapodestrian University of Athens, Greece
| | - Nikolaos Syrigos
- Physiology Laboratory, Athens Medical School, National and Kapodestrian University of Athens, Greece
| | - Spyridon Dourakis
- 2nd Academic Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens School of Medicine Hippokration General Hospital Athens Greece, Greece
| | - Michael Koutsilieris
- Physiology Laboratory, Athens Medical School, National and Kapodestrian University of Athens, Greece
| | - Athanasios Armakolas
- Physiology Laboratory, Athens Medical School, National and Kapodestrian University of Athens, Greece.
| |
Collapse
|
44
|
Kisspeptin/kisspeptin receptor system in pseudopregnant rabbit corpora lutea: presence and function. Sci Rep 2019; 9:5044. [PMID: 30911071 PMCID: PMC6433948 DOI: 10.1038/s41598-019-41623-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 03/11/2019] [Indexed: 11/09/2022] Open
Abstract
Kisspeptin (KiSS) and its related receptors (KiSS1R) have a critical role in the reproduction of mammals. The KiSS/KiSS1R system is expressed in numerous reproductive organs including the ovary. Here, we studied the expression of the KiSS/KiSS1R system and its functional role in rabbit corpora lutea (CL) at days 4 (early-), 9 (mid-), and 13 (late-stage) of pseudopregnancy. In vitro progesterone, prostaglandin (PG) F2α (PGF2α) and E2 (PGE2) productions and prostaglandin-endoperoxide synthase 1 (PTGS1) and 2 (PTGS2) activities were evaluated. Immune reactivity (IR) for KiSS and KiSS1R were detected in luteal cells at nuclear and cytoplasmic level at all luteal stage for KiSS and only at early- and mid-stage for KiSS1R; IR decreased from early- to later stages of pseudopregnancy. The KiSS-10 augmented progesterone and PGE2 and diminished PGF2α secretions by early- and mid-CL; KiSS-10 reduced PTGS2 activity at early- and mid-stages, but did not affect PTGS1 at any luteal stages. The antagonist KiSS-234 counteracted all KiSS-10 effects. This study shows that the KiSS/KiSS1R system is expressed in CL of pseudopregnant rabbits and exerts a luteotropic action by down-regulating PTGS2, which decreases PGF2α and increases PGE2 and progesterone.
Collapse
|
45
|
Association of Kiss1 and GPR54 Gene Polymorphisms with Polycystic Ovary Syndrome among Sri Lankan Women. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6235680. [PMID: 30993114 PMCID: PMC6434290 DOI: 10.1155/2019/6235680] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/21/2019] [Accepted: 02/25/2019] [Indexed: 01/07/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the commonest endocrine disorder affecting women of reproductive age. Its aetiology, though yet unclear, is presumed to have an oligogenic basis interacting with environmental factors. Kisspeptins are peptide products of Kiss1 gene that control the hypothalamic pituitary (HPG) axis by acting via G protein-coupled receptor known as GPR54. There is paucity of data on the role of Kiss1 and GPR54 gene in PCOS. We aimed to identify the polymorphisms in Kiss1 and GPR54 genes and explore their association with serum kisspeptin levels among Sri Lankan women with well-characterized PCOS. Consecutive women with PCOS manifesting from adolescence (n=55) and adult controls (n=110) were recruited. Serum kisspeptin and testosterone levels were determined by ELISA method. Whole gene sequencing was performed to identify the polymorphisms in Kiss1 and GPR54 genes. Serum kisspeptin and testosterone concentrations were significantly higher in women with PCOS than controls: kisspeptin 4.873nmol/L versus 4.127nmol/L; testosterone 4.713nmol/L versus 3.415 nmol/L, p<0.05. Sequencing the GPR54 gene revealed 5 single nucleotide polymorphisms (SNPs), rs10407968, rs1250729403, rs350131, chr19:918686, and chr19:918735, with two novel SNPs (chr19:918686 and chr19:918735), while sequencing the Kiss1 gene revealed 2 SNPs, rs5780218 and rs4889. All identified SNPs showed no significant difference in frequency between patients and controls. GPR54 gene rs350131 polymorphism (G/T) was detected more frequently in our study population. The heterozygous allele (AG) of GPR54 gene novel polymorphism chr19:918686 showed a marginal association with serum kisspeptin levels (p=0.053). Genetic variations in GPR54 and Kiss1 genes are unlikely to be associated with PCOS among Sri Lankan women manifesting from adolescence. Meanwhile the heterozygous allele of chr19:918686 is probably associated with serum kisspeptin concentrations, which suggests a potential role in the aetiology of PCOS.
Collapse
|
46
|
Computational study of putative functional variants in human kisspeptin. J Genet Eng Biotechnol 2019; 15:419-422. [PMID: 30647681 PMCID: PMC6296575 DOI: 10.1016/j.jgeb.2017.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 04/23/2017] [Accepted: 07/24/2017] [Indexed: 11/25/2022]
Abstract
Non-synonymous single nucleotide polymorphisms (nsSNPs) are a type of genetic mutations that result in amino acid substitution of the encoded proteins that may potentially affect its function and phenotype. An In Silico assay has been carried out by using bioinformatics prediction tools to identify nsSNPs which are responsible for important disorders in human kisspeptin (KISS1) gene. In this study, for the first time, KISS1 amino acid changes were discovered by tBlastn for EST database. A list of nsSNPs in human KISS1 gene from dbSNP, dbEST and UniProt databases were prepared. Computational analysis was performed using SIFT (Sorting Intolerant From Tolerant) and PolyPhen (Polymorphism Phenotyping) programs. Of the total 92 nsSNPs, 20 were found to be damaged by both servers. Six nsSNPs (P97L, G122R, W114C, R92C, R120H and N115K) are predicted with the highest damaging scores (SIFT = 0, PolyPhen = 1). These intolerant changes may suggest their functional significance in critical regions which may affect the function and stability of KISS1 protein. Identifying these nsSNPs among the thousands of them make an opportunity to screen only those predicted deleterious by programs.
Collapse
|
47
|
Kisspeptin Protein in Seminal Plasma Is Positively Associated with Semen Quality: Results from the MARHCS Study in Chongqing, China. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5129263. [PMID: 30729125 PMCID: PMC6343164 DOI: 10.1155/2019/5129263] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 12/17/2018] [Indexed: 01/17/2023]
Abstract
Objectives To study the associations between kisspeptin levels in seminal plasma and blood plasma and semen quality. Materials and Methods We conducted a male reproductive health survey in June 2014. A total of 666 volunteers were recruited from colleges in Chongqing, China. All volunteers completed a questionnaire including information on domestic characteristics and some potential confounders. We tested the kisspeptin levels in both blood and seminal plasma. Total seminal kisspeptin was calculated as the concentration of kisspeptin in seminal plasma multiplied by semen volume. Semen samples were tested according to the 2010 World Health Organization's (WHO) guidelines. Spearman correlation and multivariate linear regression were used to explore the association between kisspeptin concentrations in seminal plasma and blood plasma and semen quality. Potential confounders that were adjusted for included age, abstinence time, body mass index (BMI), grade, and smoking. Results The median of kisspeptin levels in seminal plasma was 60,000 times higher than kisspeptin in blood plasma (28.0 × 106 pg/ml versus 448.9 pg/ml). Each interquartile range (IQR) of kisspeptin in seminal plasma was associated with a 4.6% (95% confidence interval [CI]: 1.6%–7.6%) increase in sperm concentration. Each IQR of total kisspeptin was associated with a 7.7% (95% CI: 4.4%–11.0%) increase in total sperm number and a 7.8% (95% CI: 4.0%–11.7%) increase in total motile sperm count. Kisspeptin levels were further classified into quartiles and Q1 was set as the reference level. Subjects in the high total kisspeptin group had 57.5% (95% CI: 33.2%–86.2%) higher total sperm number than the reference group. Conclusion The positive association between kisspeptin levels in seminal plasma and semen quality supported an important role for the KISS1/GPR54 system in male reproductive health. Kisspeptin may be a potential marker of male reproductive health and an alternative strategy for treating infertility.
Collapse
|
48
|
KiSS1 in regulation of metastasis and response to antitumor drugs. Drug Resist Updat 2019; 42:12-21. [DOI: 10.1016/j.drup.2019.02.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/03/2019] [Accepted: 02/06/2019] [Indexed: 12/15/2022]
|
49
|
Mishra GK, Patra MK, Singh LK, Upmanyu V, Chakravarti S, M. K, Singh SK, Das GK, Kumar H, Krisnaswami N. Kiss1 and its receptor: molecular characterization and immunolocalization in the hypothalamus and corpus luteum of the buffalo. Anim Biotechnol 2018; 30:342-351. [DOI: 10.1080/10495398.2018.1520715] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Girish Kumar Mishra
- Animal Reproduction Division, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Manas Kumar Patra
- Animal Reproduction Division, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Laishram Kipjen Singh
- Animal Reproduction Division, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Vikramaditya Upmanyu
- Biological Standardization Division, Indian Veterinary research Institute, Bareilly, Uttar Pradesh, India
| | - Soumendu Chakravarti
- Division of Biological Products, Indian Veterinary Research Institute, Izatnagar, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Karikalan M.
- Centre for Wildlife Conservation Management and Disease Surveillance, Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Sanjay Kumar Singh
- Animal Reproduction Division, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Goutam Kumar Das
- Animal Reproduction Division, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Harendra Kumar
- Animal Reproduction Division, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Narayanan Krisnaswami
- Animal Reproduction Division, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| |
Collapse
|
50
|
Kaprara A, Huhtaniemi IT. The hypothalamus-pituitary-gonad axis: Tales of mice and men. Metabolism 2018; 86:3-17. [PMID: 29223677 DOI: 10.1016/j.metabol.2017.11.018] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/28/2017] [Accepted: 11/29/2017] [Indexed: 02/06/2023]
Abstract
Reproduction is controlled by the hypothalamic-pituitary-gonadal (HPG) axis. Gonadotropin-releasing hormone (GnRH) neurons play a central role in this axis through production of GnRH, which binds to a membrane receptor on pituitary gonadotrophs and stimulates the biosynthesis and secretion of follicle-stimulating hormone (FSH) and luteinizing hormone (LH). Multiple factors affect GnRH neuron migration, GnRH gene expression, GnRH pulse generator, GnRH secretion, GnRH receptor expression, and gonadotropin synthesis and release. Among them anosmin is involved in the guidance of the GnRH neuron migration, and a loss-of-function mutation in its gene leads to a failure of their migration from the olfactory placode to the hypothalamus, with consequent anosmic hypogonadotropic hypogonadism (Kallmann syndrome). There are also cases of hypogonadotropic hypogonadim with normal sense of smell, due to mutations of other genes. Another protein, kisspeptin plays a crucial role in the regulation of GnRH pulse generator and the pubertal development. GnRH is the main hypothalamic regulator of the release of gonadotropins. Finally, FSH and LH are the essential hormonal regulators of testicular functions, acting through their receptors in Sertoli and Leydig cells, respectively. The main features of the male HPG axis will be described in this review.
Collapse
Affiliation(s)
- Athina Kaprara
- Unit of Reproductive Endocrinology, Medical School, Aristotle University of Thessaloniki, Greece.
| | | |
Collapse
|