1
|
Qin Y, Deng H, Liu L, Li M, Yang J, Zhang C, Zhou J, Xiao Y. Serum phoenixin levels in girls with central precocious puberty and premature thelarche. Endocrine 2025; 87:857-865. [PMID: 39427109 DOI: 10.1007/s12020-024-04074-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND AND AIM Phoenixin (PNX), a newly discovered neuropeptide associated with reproduction, has been speculated to be involved in precocious puberty. Therefore, we assessed serum PNX levels in girls with precocious puberty. METHODS Serum phoenixin-14 (PNX-14) and phoenixin-20 (PNX-20) levels were determined in girls with central precocious puberty (CPP) and premature thelarche (PT) and in healthy controls (n = 58 per group). Spearman's correlation was used to analyze the correlations between variables. Receiver operating characteristic curves were used to evaluate the performance of PNX for the diagnosis of CPP. Significant predictors of serum PNX levels were determined using least absolute shrinkage and selection operator regression and multiple linear regression analyses. RESULTS Serum PNX-14 and PNX-20 levels were significantly higher in girls with CPP than in the controls; however, no significant differences in serum PNX-14 and PNX-20 levels were observed between girls with PT and the controls. PNX-20 levels were positively correlated with basal luteinizing hormone (LH) levels, peak LH levels, the peak LH to follicle-stimulating hormone (FSH) ratio, and estradiol levels. No significant correlation was observed between PNX-14 levels and any of these parameters. Multivariate linear regression analysis revealed that PNX-20 levels exhibited the strongest correlation with peak LH/FSH values. The areas under the curve (AUCs) of PNX-14 and PNX-20 for predicting CPP were 0.628 (cut-off value, 100.12 pg/mL; sensitivity, 44.6%; specificity, 77.6%) and 0.775 (cut-off value, 360.03 pg/mL; sensitivity, 66.5%; specificity, 79.3%), respectively. When these two indicators were combined, the AUC was 0.785. CONCLUSIONS Serum PNX levels may be associated with precocious puberty in girls and can be used as an auxiliary CPP indicator. However, given the low sensitivity and specificity of PNX, it should not be used as a single diagnostic indicator of CPP.
Collapse
Affiliation(s)
- Yujie Qin
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hongyang Deng
- Department of Pediatrics, Xijing Hospital, Xi'an, China
| | - Lujie Liu
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Li
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiong Yang
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chenglin Zhang
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jing Zhou
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Yanfeng Xiao
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
2
|
Xiao Q, Gong S, Huang Z, Peng W, Han Z, Gan Y, Shen Y, You W, Ke C, Luo X. Ten Candidate Genes Were Identified to Be Associated with the Great Growth Differentiation in the Three-Way Cross Hybrid Abalone. Animals (Basel) 2025; 15:211. [PMID: 39858210 PMCID: PMC11758661 DOI: 10.3390/ani15020211] [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: 11/21/2024] [Revised: 12/29/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Abalone is an economically important mollusk, whose slow growth has impeded the recovery of its wild populations and development of aquaculture. The three-way cross hybrid abalone ((Haliotis discus hannai♀ × H. fulgens♂)♀ × H. gigantea♂, DF × SS) demonstrated notable diversity in growth traits across the population with genetic differentiation, offering a model for exploring the molecular mechanisms of abalone growth. In this study, a total of 89 SNPs and 97 candidate genes were identified to be associated with growth-related traits of abalone using whole-genome resequencing and a genome-wide association study (GWAS) analysis. Then, ten overlap genes were found among these candidate genes by combining the results of GWAS and comparative transcriptomic analyses between the large individuals (L group) and small individuals (S group) of DF × SS. These overlap genes include up-regulated genes (fabG) and down-regulated genes (HMCN1, TLR3, ITIH3) between the L and the S groups, which are thought to function in growth in other organisms. The biological functions of these candidate genes in abalone still have to be confirmed, but they have improved our understanding of the molecular mechanisms behind abalone growth traits and provided molecular markers for abalone breeding programs.
Collapse
Affiliation(s)
- Qizhen Xiao
- Fujian Provincial Key Lab of Coastal Basin Environment, Fujian Polytechnic Normal University, Fuqing 350300, China;
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China; (S.G.); (Z.H.); (W.P.); (Z.H.); (Y.G.); (Y.S.); (W.Y.); (C.K.)
| | - Shihai Gong
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China; (S.G.); (Z.H.); (W.P.); (Z.H.); (Y.G.); (Y.S.); (W.Y.); (C.K.)
| | - Zekun Huang
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China; (S.G.); (Z.H.); (W.P.); (Z.H.); (Y.G.); (Y.S.); (W.Y.); (C.K.)
| | - Wenzhu Peng
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China; (S.G.); (Z.H.); (W.P.); (Z.H.); (Y.G.); (Y.S.); (W.Y.); (C.K.)
| | - Zhaofang Han
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China; (S.G.); (Z.H.); (W.P.); (Z.H.); (Y.G.); (Y.S.); (W.Y.); (C.K.)
| | - Yang Gan
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China; (S.G.); (Z.H.); (W.P.); (Z.H.); (Y.G.); (Y.S.); (W.Y.); (C.K.)
- Abalone Research Center, Fujian Minruibao Marine Biotechnology Co., Ltd., Xiamen 361102, China
| | - Yawei Shen
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China; (S.G.); (Z.H.); (W.P.); (Z.H.); (Y.G.); (Y.S.); (W.Y.); (C.K.)
| | - Weiwei You
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China; (S.G.); (Z.H.); (W.P.); (Z.H.); (Y.G.); (Y.S.); (W.Y.); (C.K.)
| | - Caihuan Ke
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China; (S.G.); (Z.H.); (W.P.); (Z.H.); (Y.G.); (Y.S.); (W.Y.); (C.K.)
| | - Xuan Luo
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China; (S.G.); (Z.H.); (W.P.); (Z.H.); (Y.G.); (Y.S.); (W.Y.); (C.K.)
- Abalone Research Center, Fujian Minruibao Marine Biotechnology Co., Ltd., Xiamen 361102, China
| |
Collapse
|
3
|
Karadağ MA, Gram A, Schäfer-Somi S, Aslan S, Kaya D. Expression of GnRH, Kisspeptin, and Their Specific Receptors in the Ovary and Uterus in Deslorelin-Treated Late-Prepubertal Bitches. Vet Sci 2024; 11:591. [PMID: 39728931 DOI: 10.3390/vetsci11120591] [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: 10/17/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
In this study, the expression and localization of gonadotropin-releasing hormone (GnRH1) and kisspeptin (KISS1) and their specific receptors in canine ovarian and uterine tissues were investigated after the application of deslorelin acetate (Suprelorin®, 4.7 mg, Virbac, France) in the late prepubertal period. We hypothesized that prolonged treatment of prepubertal dogs with deslorelin would alter the expression of GnRH and kisspeptin genes in the uterus and ovaries. Ovarian and uterine samples of 25 dogs with an average age of 7.8 ± 0.2 months and from mixed breeds were used. Following implant insertion, dogs entered estrus (EST; n = 6); dogs without estrus (N-EST; n = 10) comprised the experimental groups. Nine dogs with placebo implants served as a control (CONT). Ovarian and uterine tissues were investigated for expression of GnRH1, GnRHR, KISS1, and KISS1R/GPR54 mRNA and protein by using IHC and RT-qPCR. In the uterus, expression of GnRH1 significantly decreased in response to deslorelin treatment in the N-EST, compared with the control group. Compared with CONT, KISS1R expression in ovarian samples was significantly lower in the EST group. Uterine protein expression of GnRH1 appeared weaker in N-EST than in CONT. While GnRH1-system members and KISS1 protein were localized in the follicles at various stages and stroma, no or only weak signals were detected for KISS1R in the ovarian samples. Deslorelin-mediated induction of puberty by changing the expression of some of the GnRH and KISS1-system members seems to have an effect on ovarian and uterine functionality. Deslorelin implants can, therefore, not be considered a valuable alternative to induce fertile estrus in late-prepubertal bitches. However, further studies with a larger number of animals are needed to clarify the effect of deslorelin-mediated induction of puberty.
Collapse
Affiliation(s)
- Muhammet Ali Karadağ
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Kafkas University, Kars 36100, Türkiye
| | - Aykut Gram
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, Kayseri 38039, Türkiye
| | | | - Selim Aslan
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Near East University, Nicosia 99138, Cyprus
| | - Duygu Kaya
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Dokuz Eylül University, İzmir 35890, Türkiye
| |
Collapse
|
4
|
Navin AK, Aruldhas MM, Mani KK, Navaneethabalakrishnan S, Venkatachalam S, Banu SK. Unraveling Hypothalamus-Pituitary dysregulation: Hypergonadotropism in F 1 progeny due to prenatal exposure to hexavalent chromium. J Biochem Mol Toxicol 2024; 38:e23699. [PMID: 38532648 DOI: 10.1002/jbt.23699] [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: 10/16/2023] [Revised: 02/18/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024]
Abstract
The endocrine disruptor hexavalent chromium [Cr(VI)] is a proven reproductive toxicant. We recently demonstrated that prenatal Cr(VI) exposure causes testicular resistance to gonadotropins, resulting in hypergonadotropic hypoandrogenism in F1 rats. However, the mechanism driving hypergonadotropism in F1 rats exposed to Cr(VI) prenatally remains an enigma. Therefore, we hypothesized that 'Prenatal Cr(VI) exposure may disrupt steroid hormones-mediated negative feedback regulation of the hypothalamic GnRH, and its receptor in the pituitary of F1 rats, leading to hypergonadotropism.' We administered potassium dichromate (50, 100, or 200 mg/L) to pregnant rats through drinking water between days 9 and 14, and their male F1 offspring were euthanized at 60 days of age. Prenatal Cr(VI) exposure in F1 rats resulted in the accumulation of Cr in the hypothalamus and pituitary. Western blot detected decreased hypothalamic GnRH, Kisspeptin1, and its receptor GPR54, along with diminished ERα, AR, aromatase, and 5α reductase, and GnRH regulatory transcription factors Pit-1 and GATA-4 proteins. Immunohistochemical studies revealed increased immunopositivity of GnRH receptor, AR, 5α reductase, ERα, ERβ, and aromatase proteins in the pituitary, whereas decreased Kisspeptin1, GPR54, and inhibin β. Our findings imply that Cr(VI) exposure during the prenatal period disrupts the hypothalamic Kisspeptin-GPR54-Pit-1/GATA4-GnRH network, boosting the pituitary GnRH receptor. We conclude that prenatal exposure to Cr(VI) alters GnRH expression in the hypothalamus and its receptor in the pituitary of F1 progeny through interfering with the negative feedback effect of androgens and estrogens.
Collapse
Affiliation(s)
- Ajit Kumar Navin
- Department of Endocrinology, Dr. A.L.M. Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Taramani-Velachery Link Road, Chennai, Tamil Nadu, India
| | - Mariajoseph Michael Aruldhas
- Department of Endocrinology, Dr. A.L.M. Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Taramani-Velachery Link Road, Chennai, Tamil Nadu, India
| | - Kathiresh Kumar Mani
- Department of Endocrinology, Dr. A.L.M. Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Taramani-Velachery Link Road, Chennai, Tamil Nadu, India
| | - Shobana Navaneethabalakrishnan
- Department of Endocrinology, Dr. A.L.M. Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Taramani-Velachery Link Road, Chennai, Tamil Nadu, India
| | - Sankar Venkatachalam
- Department of Anatomy, Dr. A.L.M. Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Taramani-Velachery Link Road, Chennai, Tamil Nadu, India
| | - Sakhila K Banu
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, TAMU-4458, Texas A&M University, College Station, TX, USA
| |
Collapse
|
5
|
Sliwowska JH, Woods NE, Alzahrani AR, Paspali E, Tate RJ, Ferro VA. Kisspeptin a potential therapeutic target in treatment of both metabolic and reproductive dysfunction. J Diabetes 2024; 16:e13541. [PMID: 38599822 PMCID: PMC11006622 DOI: 10.1111/1753-0407.13541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/21/2023] [Accepted: 02/03/2024] [Indexed: 04/12/2024] Open
Abstract
Kisspeptins (KPs) are proteins that were first recognized to have antimetastatic action. Later, the critical role of this peptide in the regulation of reproduction was proved. In recent years, evidence has been accumulated supporting a role for KPs in regulating metabolic processes in a sexual dimorphic manner. It has been proposed that KPs regulate metabolism both indirectly via gonadal hormones and/or directly via the kisspeptin receptor in the brain, brown adipose tissue, and pancreas. The aim of the review is to provide both experimental and clinical evidence indicating that KPs are peptides linking metabolism and reproduction. We propose that KPs could be used as a potential target to treat both metabolic and reproductive abnormalities. Thus, we focus on the consequences of disruptions in KPs and their receptors in metabolic conditions such as diabetes, undernutrition, obesity, and reproductive disorders (hypogonadotropic hypogonadism and polycystic ovary syndrome). Data from both animal models and human subjects indicate that alterations in KPs in the case of metabolic imbalance lead also to disruptions in reproductive functions. Changes both in the hypothalamic and peripheral KP systems in animal models of the aforementioned disorders are discussed. Finally, an overview of current clinical studies involving KP in fertility and metabolism show fewer studies on metabolism (15%) and only one to date on both. Presented data indicate a dynamic and emerging field of KP studies as possible therapeutic targets in treatments of both reproductive and metabolic dysfunctions.
Collapse
Affiliation(s)
- Joanna Helena Sliwowska
- Department of Veterinary Medicine and Animal Sciences, Laboratory of Neurobiology, Poznan University of Life Sciences, Poznan, Poland
| | - Nicola Elizabeth Woods
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Abdullah Rzgallah Alzahrani
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Elpiniki Paspali
- Department of Chemical Engineering, University of Strathclyde, Glasgow, UK
| | - Rothwelle Joseph Tate
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Valerie Anne Ferro
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
6
|
Anderson GM, Hill JW, Kaiser UB, Navarro VM, Ong KK, Perry JRB, Prevot V, Tena-Sempere M, Elias CF. Metabolic control of puberty: 60 years in the footsteps of Kennedy and Mitra's seminal work. Nat Rev Endocrinol 2024; 20:111-123. [PMID: 38049643 PMCID: PMC10843588 DOI: 10.1038/s41574-023-00919-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/19/2023] [Indexed: 12/06/2023]
Abstract
An individual's nutritional status has a powerful effect on sexual maturation. Puberty onset is delayed in response to chronic energy insufficiency and is advanced under energy abundance. The consequences of altered pubertal timing for human health are profound. Late puberty increases the chances of cardiometabolic, musculoskeletal and neurocognitive disorders, whereas early puberty is associated with increased risks of adult obesity, type 2 diabetes mellitus, cardiovascular diseases and various cancers, such as breast, endometrial and prostate cancer. Kennedy and Mitra's trailblazing studies, published in 1963 and using experimental models, were the first to demonstrate that nutrition is a key factor in puberty onset. Building on this work, the field has advanced substantially in the past decade, which is largely due to the impressive development of molecular tools for experimentation and population genetics. In this Review, we discuss the latest advances in basic and translational sciences underlying the nutritional and metabolic control of pubertal development, with a focus on perspectives and future directions.
Collapse
Affiliation(s)
- Greg M Anderson
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH, USA
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Victor M Navarro
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken K Ong
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - John R B Perry
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Vincent Prevot
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain.
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain.
| | - Carol F Elias
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
- Department of Obstetrics & Gynecology, University of Michigan, Ann Arbor, MI, USA.
- Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
Gutási A, Hammer SE, El-Matbouli M, Saleh M. Review: Recent Applications of Gene Editing in Fish Species and Aquatic Medicine. Animals (Basel) 2023; 13:1250. [PMID: 37048506 PMCID: PMC10093118 DOI: 10.3390/ani13071250] [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: 02/23/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/08/2023] Open
Abstract
Gene editing and gene silencing techniques have the potential to revolutionize our knowledge of biology and diseases of fish and other aquatic animals. By using such techniques, it is feasible to change the phenotype and modify cells, tissues and organs of animals in order to cure abnormalities and dysfunctions in the organisms. Gene editing is currently experimental in wide fields of aquaculture, including growth, controlled reproduction, sterility and disease resistance. Zink finger nucleases, TALENs and CRISPR/Cas9 targeted cleavage of the DNA induce favorable changes to site-specific locations. Moreover, gene silencing can be used to inhibit the translation of RNA, namely, to regulate gene expression. This methodology is widely used by researchers to investigate genes involved in different disorders. It is a promising tool in biotechnology and in medicine for investigating gene function and diseases. The production of food fish has increased markedly, making fish and seafood globally more popular. Consequently, the incidence of associated problems and disease outbreaks has also increased. A greater investment in new technologies is therefore needed to overcome such problems in this industry. To put it concisely, the modification of genomic DNA and gene silencing can comprehensively influence aquatic animal medicine in the future. On the ethical side, these precise genetic modifications make it more complicated to recognize genetically modified organisms in nature and can cause several side effects through created mutations. The aim of this review is to summarize the current state of applications of gene modifications and genome editing in fish medicine.
Collapse
Affiliation(s)
- Anikó Gutási
- Department of Farm Animals and Veterinary Public Health, Division of Fish Health, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Sabine E. Hammer
- Department of Pathobiology, Institute of Immunology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Mansour El-Matbouli
- Department of Farm Animals and Veterinary Public Health, Division of Fish Health, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Mona Saleh
- Department of Farm Animals and Veterinary Public Health, Division of Fish Health, University of Veterinary Medicine, 1210 Vienna, Austria
| |
Collapse
|
8
|
Medeiros LDS, Rodrigues PDS, Santos DNL, Silva-Sampaio AC, Kirsten TB, Suffredini IB, Coque ADC, da Silva RA, Bernardi MM. Prenatal restraint stress downregulates the hypothalamic kisspeptidergic system transcripts genes, reduces the estrogen plasma levels, delayed the onset of puberty, and reduced the sexual behavior intensity in female rats. Physiol Behav 2023; 260:114055. [PMID: 36563733 DOI: 10.1016/j.physbeh.2022.114055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 10/28/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
AIMS This study investigated the possible relationships between the expression of the Kiss1 and Gpr54 gene expressions and the pituitary-gonadal hormones with the female onset of puberty and sexual behavior. The Kiss1 and Gpr54 gene expressions were examined because they are critical to controlling the hypothalamic activation of GnRH neurons and, in turn, the pituitary-gonadal hormones related to the early onset of puberty and sexual behavior. Further, it was evaluated that the pituitary and gonadal hormones involved in the vaginal opening and the expression of sexual behavior. METHODS Pregnant rats exposed to PRS from gestation days 17 to 20 were evaluated for maternal and open-field behaviors. The maternal behavior was analyzed because it may alter brain sexual organization affecting the pups development. It was observed in female pups the physical and development and, in adult age, the open-field behavior, the anxiety-like behavior, the estrous cycle, the sexual behavior, the serum FSH, LH, estrogen, progesterone, and testosterone levels, and the gene expression of kisspeptin protein (Kiss1) and Gpr54 in the hypothalamus. RESULTS the maternal and open-field behaviors were unaffected. In the F1 generation, PRS reduced weight at weaning, delayed the day of the vaginal opening and reduced the intensity of lordosis, the estrogen levels, and the Kiss1 and Gpr54 gene expression. These effects were attributed to hypothalamic kisspeptidergic system downregulation of transcripts genes and the reduced estrogen levels affected by the PRS.
Collapse
Affiliation(s)
- Loren da Silva Medeiros
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Paula da Silva Rodrigues
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Daniel Nascimento Lago Santos
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Ana Claudia Silva-Sampaio
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Thiago Berti Kirsten
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Ivana Barbosa Suffredini
- Núcleo de Pesquisas em Biodiversidade, Laboratório de Extração, Universidade Paulista - UNIP, Av. Paulista, 900, São Paulo, SP 01310-100, Brazil
| | - Alex de Camargo Coque
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Rodrigo Augusto da Silva
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil; School of Dentistry, Graduate Program in Health Sciences, University of Taubaté, Rua dos Operários, 9, Taubaté, SP 12020-340, Brazil
| | - Maria Martha Bernardi
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil.
| |
Collapse
|
9
|
Sobrino V, Avendaño MS, Perdices-López C, Jimenez-Puyer M, Tena-Sempere M. Kisspeptins and the neuroendocrine control of reproduction: Recent progress and new frontiers in kisspeptin research. Front Neuroendocrinol 2022; 65:100977. [PMID: 34999056 DOI: 10.1016/j.yfrne.2021.100977] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 12/31/2022]
Abstract
In late 2003, a major breakthrough in our understanding of the mechanisms that govern reproduction occurred with the identification of the reproductive roles of kisspeptins, encoded by the Kiss1 gene, and their receptor, Gpr54 (aka, Kiss1R). The discovery of this unsuspected reproductive facet attracted an extraordinary interest and boosted an intense research activity, in human and model species, that, in a relatively short period, established a series of basic concepts on the physiological roles of kisspeptins. Such fundamental knowledge, gathered in these early years of kisspeptin research, set the scene for the more recent in-depth dissection of the intimacies of the neuronal networks involving Kiss1 neurons, their precise mechanisms of regulation and the molecular underpinnings of the function of kisspeptins as pivotal regulators of all key aspects of reproductive function, from puberty onset to pulsatile gonadotropin secretion and the metabolic control of fertility. While no clear temporal boundaries between these two periods can be defined, in this review we will summarize the most prominent advances in kisspeptin research occurred in the last ten years, as a means to provide an up-dated view of the state of the art and potential paths of future progress in this dynamic, and ever growing domain of Neuroendocrinology.
Collapse
Affiliation(s)
- Veronica Sobrino
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Hospital Universitario Reina Sofia, 14004 Cordoba, Spain
| | - Maria Soledad Avendaño
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Hospital Universitario Reina Sofia, 14004 Cordoba, Spain
| | - Cecilia Perdices-López
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Hospital Universitario Reina Sofia, 14004 Cordoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004 Cordoba, Spain
| | - Manuel Jimenez-Puyer
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Hospital Universitario Reina Sofia, 14004 Cordoba, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Hospital Universitario Reina Sofia, 14004 Cordoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004 Cordoba, Spain; Institute of Biomedicine, University of Turku, FIN-20520 Turku, Finland.
| |
Collapse
|
10
|
Umaña Sedó S, Figueiredo C, Gonzalez T, Duarte G, Ugarte Marin M, Crawford C, Pohler K, Chebel R, Bilby T, Bisinotto R. Evaluation of luteolysis, follicle size, and time to ovulation in Holstein heifers treated with two different analogs and doses of prostaglandin-F2α. J Dairy Sci 2022; 105:5506-5518. [DOI: 10.3168/jds.2021-21487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/21/2022] [Indexed: 11/19/2022]
|
11
|
Hudson AD, Kauffman AS. Metabolic actions of kisspeptin signaling: Effects on body weight, energy expenditure, and feeding. Pharmacol Ther 2022; 231:107974. [PMID: 34530008 PMCID: PMC8884343 DOI: 10.1016/j.pharmthera.2021.107974] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 12/18/2022]
Abstract
Kisspeptin (encoded by the Kiss1 gene) and its receptor, KISS1R (encoded by the Kiss1r gene), have well-established roles in stimulating reproduction via central actions on reproductive neural circuits, but recent evidence suggests that kisspeptin signaling also influences metabolism and energy balance. Indeed, both Kiss1 and Kiss1r are expressed in many metabolically-relevant peripheral tissues, including both white and brown adipose tissue, the liver, and the pancreas, suggesting possible actions on these tissues or involvement in their physiology. In addition, there may be central actions of kisspeptin signaling, or factors co-released from kisspeptin neurons, that modulate metabolic, feeding, or thermoregulatory processes. Accumulating data from animal models suggests that kisspeptin signaling regulates a wide variety of metabolic parameters, including body weight and energy expenditure, adiposity and adipose tissue function, food intake, glucose metabolism, respiratory rates, locomotor activity, and thermoregulation. Herein, the current evidence for the involvement of kisspeptin signaling in each of these physiological parameters is reviewed, gaps in knowledge identified, and future avenues of important research highlighted. Collectively, the discussed findings highlight emerging non-reproductive actions of kisspeptin signaling in metabolism and energy balance, in addition to previously documented roles in reproductive control, but also emphasize the need for more research to resolve current controversies and uncover underlying molecular and physiological mechanisms.
Collapse
Affiliation(s)
- Alexandra D Hudson
- Dept. of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Alexander S Kauffman
- Dept. of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, United States of America.
| |
Collapse
|
12
|
Hatef A, Rajeswari JJ, Unniappan S. Kisspeptin stimulates oocyte maturation, and food deprivation modulates the abundance of kisspeptin system in zebrafish gonads. AQUACULTURE AND FISHERIES 2022. [DOI: 10.1016/j.aaf.2022.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
13
|
Knoedler JR, Inoue S, Bayless DW, Yang T, Tantry A, Davis CH, Leung NY, Parthasarathy S, Wang G, Alvarado M, Rizvi AH, Fenno LE, Ramakrishnan C, Deisseroth K, Shah NM. A functional cellular framework for sex and estrous cycle-dependent gene expression and behavior. Cell 2022; 185:654-671.e22. [PMID: 35065713 PMCID: PMC8956134 DOI: 10.1016/j.cell.2021.12.031] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/22/2021] [Accepted: 12/20/2021] [Indexed: 01/05/2023]
Abstract
Sex hormones exert a profound influence on gendered behaviors. How individual sex hormone-responsive neuronal populations regulate diverse sex-typical behaviors is unclear. We performed orthogonal, genetically targeted sequencing of four estrogen receptor 1-expressing (Esr1+) populations and identified 1,415 genes expressed differentially between sexes or estrous states. Unique subsets of these genes were distributed across all 137 transcriptomically defined Esr1+ cell types, including estrous stage-specific ones, that comprise the four populations. We used differentially expressed genes labeling single Esr1+ cell types as entry points to functionally characterize two such cell types, BNSTprTac1/Esr1 and VMHvlCckar/Esr1. We observed that these two cell types, but not the other Esr1+ cell types in these populations, are essential for sex recognition in males and mating in females, respectively. Furthermore, VMHvlCckar/Esr1 cell type projections are distinct from those of other VMHvlEsr1 cell types. Together, projection and functional specialization of dimorphic cell types enables sex hormone-responsive populations to regulate diverse social behaviors.
Collapse
Affiliation(s)
- Joseph R Knoedler
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Sayaka Inoue
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Daniel W Bayless
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Taehong Yang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Adarsh Tantry
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Chung-Ha Davis
- Neurosciences Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Nicole Y Leung
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | | | - Grace Wang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Maricruz Alvarado
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Abbas H Rizvi
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Lief E Fenno
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | | | - Karl Deisseroth
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Nirao M Shah
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Department of Neurobiology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
14
|
Ikegami K, Watanabe Y, Nakamura S, Goto T, Inoue N, Uenoyama Y, Tsukamura H. Cellular and molecular mechanisms regulating the KNDy neuronal activities to generate and modulate GnRH pulse in mammals. Front Neuroendocrinol 2022; 64:100968. [PMID: 34808231 DOI: 10.1016/j.yfrne.2021.100968] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/18/2021] [Accepted: 11/15/2021] [Indexed: 12/30/2022]
Abstract
Accumulating findings during the past decades have demonstrated that the hypothalamic arcuate kisspeptin neurons are supposed to be responsible for pulsatile release of gonadotropin-releasing hormone (GnRH) to regulate gametogenesis and steroidogenesis in mammals. The arcuate kisspeptin neurons express neurokinin B (NKB) and dynorphin A (Dyn), thus, the neurons are also referred to as KNDy neurons. In the present article, we mainly focus on the cellular and molecular mechanisms underlying GnRH pulse generation, that is focused on the action of NKB and Dyn and an interaction between KNDy neurons and astrocytes to control GnRH pulse generation. Then, we also discuss the factors that modulate the activity of KNDy neurons and consequent pulsatile GnRH/LH release in mammals.
Collapse
Affiliation(s)
- Kana Ikegami
- Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Youki Watanabe
- Graduate School of Applied Life Science, Nippon Veterinary and Life Science University, Tokyo 180-8602, Japan
| | - Sho Nakamura
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime 794-8555, Japan
| | - Teppei Goto
- RIKEN Center for Biosystems Dynamics Research, Hyogo 650-0047, Japan
| | - Naoko Inoue
- Graduate School of Bioagricultural Science, Nagoya University, Nagoya 464-8601, Japan
| | - Yoshihisa Uenoyama
- Graduate School of Bioagricultural Science, Nagoya University, Nagoya 464-8601, Japan
| | - Hiroko Tsukamura
- Graduate School of Bioagricultural Science, Nagoya University, Nagoya 464-8601, Japan.
| |
Collapse
|
15
|
Ghaderpour S, Ghiasi R, Heydari H, Keyhanmanesh R. The relation between obesity, kisspeptin, leptin, and male fertility. Horm Mol Biol Clin Investig 2021; 43:235-247. [PMID: 34931507 DOI: 10.1515/hmbci-2021-0058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/22/2021] [Indexed: 11/15/2022]
Abstract
Over the past decades, obesity and infertility in men increased in parallel, and the association between both phenomena have been examined by several researchers. despite the fact that there is no agreement, obesity appears to affect the reproductive potential of men through various mechanisms, such as changes in the hypothalamic-pituitary-testicular (HPT) axis, spermatogenesis, sperm quality and/or alteration of sexual health. Leptin is a hormone produced by the adipose tissue, and its production elevates with increasing body fat. Many studies have supported the relationship between raised leptin production and reproductive function regulation. In fact, Leptin acts on the HPT axis in men at all levels. However, most obese men are insensitive to increased production of endogenous leptin and functional leptin resistance development. Recently, it has been recommended that Kisspeptin neurons mediate the leptin's effects on the reproductive system. Kisspeptin binding to its receptor on gonadotropin-releasing hormone (GnRH) neurons, activates the mammal's reproductive axis and stimulates GnRH release. Increasing infertility associated with obesity is probably mediated by the Kisspeptin-GnRH pathway. In this review, the link between obesity, kisspeptin, leptin, and male fertility will be discussed.
Collapse
Affiliation(s)
- Saber Ghaderpour
- Department of Physiology, Tabriz Faculty of Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rafighe Ghiasi
- Department of Physiology, Tabriz Faculty of Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Heydari
- Department of Physiology, Tabriz Faculty of Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Department of Physiology, Tabriz Faculty of Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Li Z, Liu J, Inuzuka H, Wei W. Functional analysis of the emerging roles for the KISS1/KISS1R signaling pathway in cancer metastasis. J Genet Genomics 2021; 49:181-184. [PMID: 34767970 DOI: 10.1016/j.jgg.2021.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 02/07/2023]
Abstract
Cancer metastasis, a process that primary tumor cells disseminate to secondary organs, is the most lethal and least effectively treated characteristic of human cancers. Kisspeptins are proteins encoded by the KISS1 gene that was originally described as a melanoma metastasis suppressor gene. Then, Kisspeptins were discovered as the natural ligands of the G-protein-coupled receptor 54 (GPR54) that is also called KISS1R. The KISS1/KISS1R signaling is essential to control GnRH secretion during puberty and to establish mammalian reproductive function through the hypothalamic-pituitary-gonadal (HPG) axis. Although KISS1 primarily plays a suppressive role in the metastasis progression in several cancer types, emerging evidence indicates that the physiological effect of KISS1/KISS1R in cancer metastasis is tissue context-dependent and still controversial. Here, we will discuss the epigenetic mechanism regulation of KISS1 gene expression, the context-dependent role of KISS1/KISS1R, pro-/anti-metastasis signaling pathways of KISS1/KISS1R, and the perspective anti-cancer therapeutics via targeting KISS1/KISS1R.
Collapse
Affiliation(s)
- Zhenxi Li
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Jing Liu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
17
|
Kırgezen T, Yücetaş U, Server EA, Övünç O, Yiğit Ö. Possible effects of low testosterone levels on olfactory function in males. Braz J Otorhinolaryngol 2021; 87:702-710. [PMID: 32345533 PMCID: PMC9422496 DOI: 10.1016/j.bjorl.2020.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/05/2020] [Accepted: 03/08/2020] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION Functions attributed to androgens have increased, ranging from the role in hypothalamic-pituitary-gonadal axis and reproductive behaviors to modulation of cognition, mood and some other functions. Sex differences and changes in circulating sex hormones affect human sensory function. In the literature, authors reported this kind of influence for olfaction predominantly in females. OBJECTIVE To investigate the effects of low testosterone levels on olfactory functions in males, in this prospective clinical study. METHODS Male patients diagnosed with prostate cancer were included. Thirty-nine patients with prostate cancer whose testosterone levels were lower than 50ng/dL due to castration, were the study group. Thirty-one patients with prostate cancer who were not castrated with testosterone levels higher than 50ng/dL were selected as the control group. Acoustic rhinometry and peak nasal inspiratory flow tests were performed for all participants; and for evaluation of olfactory function, both groups completed the Connecticut chemosensory clinical research center olfactory test. RESULTS The mean ages of the patients and controls were 69.6±7.2 (57-89) and 66.3±5.8 (50-78) years, respectively (p=0.039). There was a significant difference between groups in terms of testosterone levels (p<0.0001). The multivariate logistic regression revealed testosterone level as the only predictive factor determining the difference between the groups. In terms of olfactory parameters, all scores were lower in the emasculated group (butanol threshold test p=0.019, identification p=0.059, and Connecticut center score p=0.029) There was a significant correlation between testosterone levels and olfactory parameters (p=0.023; p=0.025 for identification and Connecticut center scores, respectively). CONCLUSION Low testosterone levels in males have negative effects on olfactory functions. Further molecular research is required to understand the connection between testosterone and olfaction.
Collapse
Affiliation(s)
- Tolga Kırgezen
- University of Health Sciences, Istanbul Training and Research Hospital, Department of Otorhinolaryngology/Head and Neck Surgery, Istanbul, Turkey.
| | - Uğur Yücetaş
- University of Health Sciences, Istanbul Training and Research Hospital, Department of Urology, Istanbul, Turkey
| | - Ela Araz Server
- University of Health Sciences, Istanbul Training and Research Hospital, Department of Otorhinolaryngology/Head and Neck Surgery, Istanbul, Turkey
| | - Okan Övünç
- University of Health Sciences, Istanbul Training and Research Hospital, Department of Otorhinolaryngology/Head and Neck Surgery, Istanbul, Turkey
| | - Özgür Yiğit
- University of Health Sciences, Istanbul Training and Research Hospital, Department of Otorhinolaryngology/Head and Neck Surgery, Istanbul, Turkey
| |
Collapse
|
18
|
Gametogenic and steroidogenic action of kisspeptin-10 in the Asian catfish, Clarias batrachus: Putative underlying mechanistic cascade. Comp Biochem Physiol B Biochem Mol Biol 2021; 256:110642. [PMID: 34197962 DOI: 10.1016/j.cbpb.2021.110642] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/21/2021] [Accepted: 06/24/2021] [Indexed: 12/18/2022]
Abstract
Unlike mammals, two kisspeptins genes encoding, kiss1 and kiss2 are detected in fishes with highly varied and contradictory difference in their reproductive activities. The present study was undertaken to examine the direct action of kisspeptin-10 and its role in gonadal activities in the gonadally quiescent Asian catfish using native mammalian kisspeptin decapeptide (KP-10) involving in vivo and in vitro approaches. The in vivo KP-10 treatment caused precocious onset of gametogenesis and its rapid progression, as was evident from the appearance of advanced stages of ovarian follicles in ovary, and advanced germ cells (spermatocytes/ spermatids) in the testis of the treated Clarias batrachus in comparison to the control gonads. It also elevated the steroid levels in gonads of the catfish in vivo and in vitro conditions. Simultaneously, it increased the expressions of key steroidogenic enzymes like 3β-HSD, 17β-HSD, and StAR protein, responsible for transfer of cholesterol from outer to inner membrane of the mitochondria of steroidogenic cells. Concurrently, it augmented the activities of 3β-HSD and 17β-HSD in the ovarian explants. The expressions of MAPK component (pERK1/2 and ERK1/2) were also up-regulated by KP-10 in gonadal explants. Thus, the data suggest that kisspeptin-10 stimulates gametogenesis by enhancing gonadal steroid production. The study also describes the putative mechanistic cascade of steroidogenic actions of kisspeptin-10 in the catfish so much so in teleost fish. The study also suggests that, kisspeptin may act locally to regulate gonadal activities in an autocrine/paracine manner, independent of known extra-gonadal factors in the catfish.
Collapse
|
19
|
Hua X, Jiang H, Guo N, Du Y, Yuan X, Deng T, Teng X, Yao Y, Li Y. Effects of Prepubertal Exposure to Aroclor-1221 on Reproductive Development and Transcriptional Gene Expression in Female Rats. Reprod Sci 2021; 28:393-405. [PMID: 32816245 DOI: 10.1007/s43032-020-00290-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 08/04/2020] [Indexed: 12/23/2022]
Abstract
Polychlorinated biphenyls (PCBs), as persistent organic pollutants, are environmental endocrine-disrupting chemicals (EDCs). We aim to investigate the effects of prepubertal exposure to PCBs on the reproductive development and expression and regulation of related genes in rats. Female rats were treated with Aroclor-1221 (A-1221) (4 mg/kg/day, 0.4 mg/kg/day) or castor oil daily from postnatal day (PND) 28 for 2 weeks by gavage. Morphological, histological, hormonal, and biochemical parameters were studied. Lower weight and relative weight of hypothalamus, earlier puberty onset, a longer length of the estrous cycle, lower serum estradiol and progesterone levels, accelerated ovarian folliculogenesis, and higher apoptotic index in the ovary were found. The in vitro fertilization study showed a lower fertilization rate and cleavage rate. The genetic study revealed higher expression of Kiss-1 mRNA and lower expression of GnRH mRNA in the hypothalamus and higher expression of AMH mRNA and lower expression of C-myc mRNA in the ovary. These confirmed the reproductive damage of A-1221 in rats.
Collapse
Affiliation(s)
- Xiang Hua
- Reproductive Medical Center, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Huahua Jiang
- Reproductive Medical Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Na Guo
- Reproductive Medical Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yaoyao Du
- Reproductive Medical Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaoqiong Yuan
- Reproductive Medical Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Taoran Deng
- Reproductive Medical Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xuemei Teng
- Reproductive Medical Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yangcheng Yao
- Reproductive Medical Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yufeng Li
- Reproductive Medical Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
20
|
Dees WL, Hiney JK, Srivastava VK. IGF-1 Influences Gonadotropin-Releasing Hormone Regulation of Puberty. Neuroendocrinology 2021; 111:1151-1163. [PMID: 33406521 PMCID: PMC8257778 DOI: 10.1159/000514217] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/08/2020] [Indexed: 11/19/2022]
Abstract
The pubertal process is initiated as a result of complex neuroendocrine interactions within the preoptic and hypothalamic regions of the brain. These interactions ultimately result in a timely increase in the secretion of gonadotropin-releasing hormone (GnRH). Researchers for years have believed that this increase is due to a diminished inhibitory tone which has applied a prepubertal brake on GnRH secretion, as well as to the gradual development of excitatory inputs driving the increased release of the peptide. Over the years, insulin-like growth factor-1 (IGF-1) has emerged as a prime candidate for playing an important role in the onset of puberty. This review will first present initial research demonstrating that IGF-1 increases in circulation as puberty approaches, is able to induce the release of prepubertal GnRH, and can advance the timing of puberty. More recent findings depict an early action of IGF-1 to activate a pathway that releases the inhibitory brake on prepubertal GnRH secretion provided by dynorphin, as well as demonstrating that IGF-1 can also act later in the process to regulate the synthesis and release of kisspeptin, a potent stimulator of GnRH at puberty.
Collapse
Affiliation(s)
- William L Dees
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas, USA,
| | - Jill K Hiney
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas, USA
| | - Vinod K Srivastava
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
21
|
Xia Q, Di R, He XY, Wei CH, Chu MX. Expression analysis of DIO2, EYA3, KISS1 and GPR54 genes in year-round estrous and seasonally estrous rams. Arch Anim Breed 2020; 63:451-460. [PMID: 33473370 PMCID: PMC7810231 DOI: 10.5194/aab-63-451-2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/14/2020] [Indexed: 12/30/2022] Open
Abstract
The expression characteristics of the hypothalamic-pituitary-gonadal (HPG) axis-related candidate genes, DIO2, EYA3, KISS1 and GPR54, were analyzed in year-round estrous rams (small-tail Han sheep, STH) and seasonally estrous rams (Sunite sheep, SNT) using qPCR. The results were as follows: DIO2 was mainly expressed in pituitary, and KISS1 was specifically expressed in hypothalamus in the two groups. However, EYA3 and GPR54 were widely expressed in the cerebrum, cerebellum, hypothalamus, pituitary, testis, epididymis, vas deferens and adrenal gland tissues in both breeds, with significant differences in the cerebellum, hypothalamus, pituitary, testis and vas deferens tissues. We speculated that DIO2 and KISS1 may have positive roles in different regions in ram year-round estrus. Moreover, the expression patterns of EYA3 and GPR54 suggested that they may regulate the estrous mode of ram via testis and vas deferens. This is the first study to systematically analyze the expression patterns of HPG axis-related genes in rams.
Collapse
Affiliation(s)
- Qing Xia
- Key Laboratory of Animal Genetics and Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Ran Di
- Key Laboratory of Animal Genetics and Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Xiao-Yun He
- Key Laboratory of Animal Genetics and Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Cai-Hong Wei
- Key Laboratory of Animal Genetics and Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Ming-Xing Chu
- Key Laboratory of Animal Genetics and Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| |
Collapse
|
22
|
D’Occhio MJ, Campanile G, Baruselli PS. Peripheral action of kisspeptin at reproductive tissues-role in ovarian function and embryo implantation and relevance to assisted reproductive technology in livestock: a review. Biol Reprod 2020; 103:1157-1170. [PMID: 32776148 PMCID: PMC7711897 DOI: 10.1093/biolre/ioaa135] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/23/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022] Open
Abstract
Kisspeptin (KISS1) is encoded by the KISS1 gene and was initially found to be a repressor of metastasis. Natural mutations in the KISS1 receptor gene (KISS1R) were subsequently shown to be associated with idiopathic hypothalamic hypogonadism and impaired puberty. This led to interest in the role of KISS1 in reproduction. It was established that KISS1 had a fundamental role in the control of gonadotropin releasing hormone (GnRH) secretion. KISS1 neurons have receptors for leptin and estrogen receptor α (ERα), which places KISS1 at the gateway of metabolic (leptin) and gonadal (ERα) regulation of GnRH secretion. More recently, KISS1 has been shown to act at peripheral reproductive tissues. KISS1 and KISS1R genes are expressed in follicles (granulosa, theca, oocyte), trophoblast, and uterus. KISS1 and KISS1R proteins are found in the same tissues. KISS1 appears to have autocrine and paracrine actions in follicle and oocyte maturation, trophoblast development, and implantation and placentation. In some studies, KISS1 was beneficial to in vitro oocyte maturation and blastocyst development. The next phase of KISS1 research will explore potential benefits on embryo survival and pregnancy. This will likely involve longer-term KISS1 treatments during proestrus, early embryo development, trophoblast attachment, and implantation and pregnancy. A deeper understanding of the direct action of KISS1 at reproductive tissues could help to achieve the next step change in embryo survival and improvement in the efficiency of assisted reproductive technology.
Collapse
Affiliation(s)
- Michael J D’Occhio
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Giuseppe Campanile
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Pietro S Baruselli
- Department of Animal Reproduction, Faculty of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
23
|
Thayil AJ, Wang X, Bhandari P, vom Saal FS, Tillitt DE, Bhandari RK. Bisphenol A and 17α-ethinylestradiol-induced transgenerational gene expression differences in the brain-pituitary-testis axis of medaka, Oryzias latipes†. Biol Reprod 2020; 103:1324-1335. [PMID: 32940650 PMCID: PMC7711903 DOI: 10.1093/biolre/ioaa169] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 07/15/2020] [Accepted: 09/15/2020] [Indexed: 11/13/2022] Open
Abstract
Endocrine disrupting chemicals (EDCs), such as bisphenol A (BPA) and 17α-ethinylestradiol (EE2), can have far reaching health effects, including transgenerational abnormalities in offspring that never directly contacted either chemical. We previously reported reduced fertilization rates and embryo survival at F2 and F3 generations caused by 7-day embryonic exposure (F0) to 100 μg/L BPA or 0.05 μg/L EE2 in medaka. Crossbreeding of fish in F2 generation indicated subfertility in males. To further understand the mechanisms underlying BPA or EE2-induced adult onset and transgenerational reproductive defects in males, the present study examined the expression of genes regulating the brain-pituitary-testis (BPT) axis in the same F0 and F2 generation male medaka. Embryonic exposure to BPA or EE2 led to hyperactivation of brain and pituitary genes, which are actively involved in reproduction in adulthood of the F0 generation male fish, and some of these F0 effects continued to the F2 generation (transgenerational effects). Particularly, the F2 generation inherited the hyperactivated state of expression for kisspeptin (kiss1 and kiss2) and their receptors (kiss1r and kiss2r), and gnrh and gnrh receptors. At F2 generation, expression of DNA methyltransferase 1 (dnmt1) decreased in brain of the BPA treatment lineage, while EE2 treatment lineage showed increased dnmt3bb expression. Global hypomethylation pattern was observed in the testis of both F0 and F2 generation fish. Taken together, these results demonstrated that BPA or EE2-induced transgenerational reproductive impairment in the F2 generation was associated with alterations of reproductive gene expression in brain and testis and global DNA methylation in testis.
Collapse
Affiliation(s)
- Albert J Thayil
- Department of Biology, University of North Carolina Greensboro, Greensboro, NC, USA
| | - Xuegeng Wang
- Department of Biology, University of North Carolina Greensboro, Greensboro, NC, USA
| | - Pooja Bhandari
- Department of Biology, University of North Carolina Greensboro, Greensboro, NC, USA
| | | | - Donald E Tillitt
- United States Geological Survey, Columbia Environmental Research Center, Columbia, MO, USA
| | - Ramji K Bhandari
- Department of Biology, University of North Carolina Greensboro, Greensboro, NC, USA
| |
Collapse
|
24
|
Neuroendocrine disruption is associated to infertility in chronically stressed female rats. Reprod Biol 2020; 20:474-483. [PMID: 32807716 DOI: 10.1016/j.repbio.2020.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
Infertility is a growing worldwide public health problem, and stress is a main factor exerting detrimental effects on female reproduction. However, knowledge regarding the neuroendocrine changes caused by chronic stress in females is limited. Therefore, this study assessed the effects of stress on hormones that control female reproduction during the proestrus and diestrus stages of the estrous cycle, as well as its effects on fertility. Adult females were assigned to either a control or a stress group. Stress consisted of exposure, for 15 min, to cold-water immersion daily for 30 days. Estrous cyclicity, female sexual behavior, as well as hypothalamic kisspeptin, gonadotropin releasing hormone (GnRH) content, serum luteinizing hormone (LH), estradiol (E2), progesterone (P4), corticosterone (CORT) and fertility were assessed after chronic stress. The results show that chronically stressed females exhibited disrupted estrous cyclicity, decreased receptivity, low pregnancy rates and lower numbers of fetuses. The content of Kisspeptin and GnRH in the Anteroventral Periventricular/medial Preoptic Area decreased during proestrus, while Kisspeptin increased in the Arcuate nucleus in proestrus and diestrus. Serum LH decreased only during proestrus, whereas E2 and P4 concentrations decreased during proestrus and diestrus, with a concomitant increase in CORT levels in both stages. As a whole, these results indicate that chronic stress decreases Kisspeptin content in AVPV nucleus and GnRH in POA in females, and might induce disruption of the LH surge, consequently disrupting estrous cyclicity and fertility, leading to lower rates of pregnancy and number of fetuses.
Collapse
|
25
|
Wang T, Cao Z, Shen Z, Yang J, Chen X, Yang Z, Xu K, Xiang X, Yu Q, Song Y, Wang W, Tian Y, Sun L, Zhang L, Guo S, Zhou N. Existence and functions of a kisspeptin neuropeptide signaling system in a non-chordate deuterostome species. eLife 2020; 9:e53370. [PMID: 32513385 PMCID: PMC7282810 DOI: 10.7554/elife.53370] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
The kisspeptin system is a central modulator of the hypothalamic-pituitary-gonadal axis in vertebrates. Its existence outside the vertebrate lineage remains largely unknown. Here, we report the identification and characterization of the kisspeptin system in the sea cucumber Apostichopus japonicus. The gene encoding the kisspeptin precursor generates two mature neuropeptides, AjKiss1a and AjKiss1b. The receptors for these neuropeptides, AjKissR1 and AjKissR2, are strongly activated by synthetic A. japonicus and vertebrate kisspeptins, triggering a rapid intracellular mobilization of Ca2+, followed by receptor internalization. AjKissR1 and AjKissR2 share similar intracellular signaling pathways via Gαq/PLC/PKC/MAPK cascade, when activated by C-terminal decapeptide. The A. japonicus kisspeptin system functions in multiple tissues that are closely related to seasonal reproduction and metabolism. Overall, our findings uncover for the first time the existence and function of the kisspeptin system in a non-chordate species and provide new evidence to support the ancient origin of intracellular signaling and physiological functions that are mediated by this molecular system.
Collapse
Affiliation(s)
- Tianming Wang
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean UniversityZhoushanChina
- Programs in Human Genetics and Biological Sciences, Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
| | - Zheng Cao
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang UniversityHangzhouChina
| | - Zhangfei Shen
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang UniversityHangzhouChina
| | - Jingwen Yang
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean UniversityZhoushanChina
- Programs in Human Genetics and Biological Sciences, Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
| | - Xu Chen
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean UniversityZhoushanChina
| | - Zhen Yang
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean UniversityZhoushanChina
| | - Ke Xu
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean UniversityZhoushanChina
| | - Xiaowei Xiang
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean UniversityZhoushanChina
| | - Qiuhan Yu
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean UniversityZhoushanChina
| | - Yimin Song
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean UniversityZhoushanChina
| | - Weiwei Wang
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang UniversityHangzhouChina
| | - Yanan Tian
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang UniversityHangzhouChina
| | - Lina Sun
- Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of SciencesQingdaoChina
| | - Libin Zhang
- Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of SciencesQingdaoChina
- Center for Ocean Mega-Science, Chinese Academy of SciencesQingdaoChina
| | - Su Guo
- Programs in Human Genetics and Biological Sciences, Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
| | - Naiming Zhou
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang UniversityHangzhouChina
| |
Collapse
|
26
|
Umayal B, Jayakody SN, Chandrasekharan NV, Wijesundera WS, Wijeyaratne CN. Polycystic ovary syndrome (PCOS) and kisspeptin - A Sri Lankan study. J Postgrad Med 2020; 65:18-23. [PMID: 30004037 PMCID: PMC6380135 DOI: 10.4103/jpgm.jpgm_683_17] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Context Polycystic ovary syndrome (PCOS) is the commonest endocrine disorder affecting young women. Kisspeptins are a family of closely related peptides encoded by Kiss1 gene that controls the hypothalamic-pituitary-gonadal axis by binding to its receptor (GPR54) expressed in gonadotropin-releasing hormone (GnRH) neurons and releases GnRH. Since GnRH secretion is deregulated in PCOS, we hypothesized that dysregulated gonadotropin secretion in PCOS is reflected by kisspeptin levels. Aim We aimed to measure serum kisspeptin levels of subjects with well-characterized PCOS versus controls and explore any correlation between kisspeptin and PCOS-related reproductive and metabolic disturbances. Materials and Methods : Consecutive women with PCOS manifesting from adolescence (n = 55) and adult controls (n = 110) were recruited. Pre-treatment baseline clinical, anthropometry, and biochemical parameters were measured in all. Serum kisspeptin and testosterone levels were determined by enzyme-linked immunosorbent assay method. Results : Serum kisspeptin and testosterone concentrations were significantly higher in women with PCOS (kisspeptin 4.873 nmol/L; testosterone 4.713 nmol/L) than controls (kisspeptin 4.127 nmol/L; testosterone 3.415 nmol/L; P < 0.05). Serum kisspeptin levels were positively associated with PCOS (odds ratio: 1.853; 95% confidence interval: 1.246-2.755; P = 0.002) in our studied population. Conclusion Serum kisspeptin levels are higher in Sri Lankan women with PCOS manifesting from adolescence compared with controls regardless of body mass index. We propose serum kisspeptin concentration as a useful marker to recognize PCOS that manifests from adolescence.
Collapse
Affiliation(s)
- B Umayal
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Sri Jayewardenepura, Sri Lanka
| | - S N Jayakody
- Department of Community Medicine, Faculty of Medical Sciences, University of Sri Jayewardenepura, Sri Lanka
| | - N V Chandrasekharan
- Department of Chemistry, Faculty of Science, University of Colombo, Colombo, Sri Lanka
| | - W Ss Wijesundera
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - C N Wijeyaratne
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Sri Jayewardenepura, Sri Lanka
| |
Collapse
|
27
|
Azizi V, Oryan S, Khazali H, Hosseini A. Upregulation of Kiss-1 and Gpr54 Genes Expression in Pituitary of Male Rats Following the Central Administration of Neuropeptide Y. INTERNATIONAL JOURNAL OF BASIC SCIENCE IN MEDICINE 2019. [DOI: 10.34172/ijbsm.2019.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introduction: The neuropeptide Y (NPY) in the neural circuits of the hypothalamus has a stimulating effect on reproductive activities in mammals. Kisspeptin (KiSS1) is a quintessential neurotransmitter in the reproductive axis which directly stimulates gonadotropin-releasing hormone neurons in the hypothalamus. The distribution of KiSS1 expressing cells in the pituitary was described previously. Despite earlier reports showing the KiSS1 receptor, G-protein coupled receptor 54 (GPR54) expression in the pituitary, the potential physiological roles of kisspeptin at this gland have remained obscure. Accordingly, this study investigated the role of NPY on the relative expression of Kiss1 and Gpr54 genes in the pituitary gland in male Wistar rats. Methods: In general, 20 male Wistar rats weighing 200-250 g in 4 groups (5 in each group) received saline, NPY (2.3 nM), BIBP3226 (NPY receptor antagonist, 7.8 nM), and NPY+ BIBP3226. Then, they received the simultaneous injection of these molecules through the third ventricle of the brain. Finally, the relative mean expressions of Kiss1 and Gpr54 genes in the anterior pituitary were quantitatively analyzed by the real-time polymerase chain reaction. Results: The central injection of NPY increased the relative mean expressions of Kiss1 and Gpr54 genes in the pituitary gland compared to the control group although the injection of BIBP3226 eradicated these effects. However, the gene expression of Gpr54 in the rats receiving NPY coupled with BIBP3226 in hypophysis in comparison to the group receiving only NPY demonstrated a significant reduction (P<0.05). Conclusion: Overall, the central injection of NPY stimulated the gene expression of Kiss1 and Gpr54 in the pituitary gland.
Collapse
Affiliation(s)
- Vahid Azizi
- Department of Animal Sciences and Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Shahrbanoo Oryan
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Homayuon Khazali
- Department of Animal Sciences and Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Abdolkarim Hosseini
- Department of Animal Sciences and Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
28
|
Dees WL, Hiney JK, Srivastava VK. Regulation of prepubertal dynorphin secretion in the medial basal hypothalamus of the female rat. J Neuroendocrinol 2019; 31:e12810. [PMID: 31715027 PMCID: PMC6916394 DOI: 10.1111/jne.12810] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/21/2019] [Accepted: 11/08/2019] [Indexed: 11/27/2022]
Abstract
The onset of puberty is the result of an increase in secretion of hypothalamic gonadotrophin-releasing hormone (GnRH). This action is a result of not only the development of stimulatory inputs to its release, but also the gradual decrease in inhibitory inputs that restrain release of the peptide prior to pubertal onset. Dynorphin (DYN) is one of the inhibitory inputs produced in the medial basal hypothalamus (MBH); however, little is known about what substance(s) control its prepubertal synthesis and release. Because neurokinin B (NKB) increases in the hypothalamus as puberty approaches, we considered it a candidate for such a role. An initial study investigated the acute effects of an NKB agonist, senktide, on the secretion of DYN from MBH tissues incubated in vitro. In other experiments, central injections of senktide were administered to animals for 4 days then MBHs were collected for assessment of DYN synthesis or for the in vitro secretion of both DYN and GnRH. Because insulin-like growth factor (IGF)-1 has been shown to play an important role at puberty, additional animals received central injections of this peptide for 4 days to assess NKB and DYN synthesis or the in vitro secretion of NKB. The results obtained show that senktide administration up-regulates the NKB receptor protein, at the same time as suppressing the DYN and its receptor. Senktide consistently suppressed DYN and elevated GnRH secretion in the same tissue incubates from both the acute and chronic studies. IGF-1 administration caused an increase in NKB protein, at the same time as decreasing DYN protein. Furthermore, the central administration of IGF-1 caused an increase in NKB release, an action blocked by the IGF-1 receptor blocker, JB-1. These results indicate that the IGF-1/NKB pathway contributes to suppressing the DYN inhibitory tone on prepubertal GnRH secretion and thus facilitates the puberty-related increase in the release of GnRH to accelerate the onset of puberty.
Collapse
Affiliation(s)
- William L. Dees
- Department of Veterinary Integrative BiosciencesCollege of Veterinary MedicineTexas A&M UniversityCollege StationTXUSA
| | - Jill K. Hiney
- Department of Veterinary Integrative BiosciencesCollege of Veterinary MedicineTexas A&M UniversityCollege StationTXUSA
| | - Vinod K. Srivastava
- Department of Veterinary Integrative BiosciencesCollege of Veterinary MedicineTexas A&M UniversityCollege StationTXUSA
| |
Collapse
|
29
|
Zhang Z, Wen H, Li Y, Li Q, Li W, Zhou Y, Wang L, Liu Y, Lyu L, Qi X. TAC3 Gene Products Regulate Brain and Digestive System Gene Expression in the Spotted Sea Bass ( Lateolabrax maculatus). Front Endocrinol (Lausanne) 2019; 10:556. [PMID: 31474940 PMCID: PMC6702303 DOI: 10.3389/fendo.2019.00556] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Neurokinin B (NKB) is a member of the tachykinin (tac) family that plays important roles in mammalian growth by modulating prolactin (PRL) synthesis and secretion and causing contraction of the stomach and intestine. However, its potential role in regulating growth of teleosts is less clear. We aimed to explore the role that NKB plays in regulating fish growth using the spotted sea bass (Lateolabrax maculatus) as a model. In the present study, two tac3 and two tacr3 genes were identified in the spotted sea bass. Sequence analysis showed that two tac3 transcripts, tac3a and tac3b, encode four NKBs: NKBa-13, NKBa-10, NKBb-13, and NKBb-10. Expression analysis in different tissues showed that both genes are highly expressed in the brain, stomach and intestine of the spotted sea bass. In situ hybridization indicated that the tac3a and tac3b mRNAs are both localized in several brain regions, such as the telencephalon and hypothalamus, and that tacr3a and tacr3b are localized in the intestinal villus and gastric gland. To investigate the potential role of NKBs in regulating growth, in vitro experiments were performed to detect the effect of NKBs on growth-related gene expression in the brain and brain-gut peptide (BGP)-related genes in the stomach and intestine. NKBb-13 was the most critical ligand in regulating the expression of growth-related genes in the brain and brain-gut peptide (BGP)-related genes in the stomach. The expression of cholecystokinin (cck) was enhanced by NKBa-13, NKBa-10, and NKBb-10 but not NKBb-13 in the intestine. In general, our results showed that NKBs participate in regulating the growth of spotted sea bass.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Xin Qi
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| |
Collapse
|
30
|
Amelkina O, Tanyapanyachon P, Thongphakdee A, Chatdarong K. Identification of feline Kiss1 and distribution of immunoreactive kisspeptin in the hypothalamus of the domestic cat. J Reprod Dev 2019; 65:335-343. [PMID: 31142694 PMCID: PMC6708855 DOI: 10.1262/jrd.2018-101] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In recent years, the Kiss1 gene has been reported in a number of vertebrate species, and a substantial dataset has been acquired to demonstrate the critical role of
kisspeptins in the reproductive system; yet limited information is available for carnivores. In the present study, we identified and characterized feline Kiss1 by isolating
and cloning its full-length cDNA in the domestic cat hypothalamus and caracal testis, using the method of rapid amplification of cDNA ends. Additionally, we isolated and cloned the 3′ end of
Kiss1 cDNA, containing kisspeptin-10 (Kp10), from the ovaries of a clouded leopard and Siberian tiger. Nucleotide sequencing revealed that domestic cat
Kiss1 cDNA is of 711 base pairs and caracal Kiss1 cDNA is of 792 base pairs, both having an open reading frame of 450 base pairs, encoding a precursor
protein Kiss1 of 149 amino acids. The core sequence of the feline kisspeptin Kp10 was found to be identical in all species analyzed here and is highly conserved in other
vertebrate species. Using an anti-Kp10 antibody, we found the immunoreactive kisspeptin to be localized in the periventricular and infundibular nuclei of the cat hypothalamus. The results
show that kisspeptin is highly conserved among different feline families, and its immunoreactive distribution in the hypothalamus may indicate its physiological function in the domestic
cat.
Collapse
Affiliation(s)
- Olga Amelkina
- Research Unit of Obstetrics and Reproduction in Animals, Department of Obstetrics, Gynaecology and Reproduction, Faculty of Veterinary Sciences, Chulalongkorn University, Bangkok 10330, Thailand.,Center for Species Survival, Smithsonian Conservation Biology Institute, National Zoological Park, Washington, DC 20013, USA
| | - Prattana Tanyapanyachon
- Research Unit of Obstetrics and Reproduction in Animals, Department of Obstetrics, Gynaecology and Reproduction, Faculty of Veterinary Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Ampika Thongphakdee
- Wildlife Reproductive Innovation Center, Bureau of Conservation and Research, Zoological Park Organization under the Royal Patronage of H.M. the King, Bangkok, Thailand
| | - Kaywalee Chatdarong
- Research Unit of Obstetrics and Reproduction in Animals, Department of Obstetrics, Gynaecology and Reproduction, Faculty of Veterinary Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
31
|
Wassie T, Liu G, Jiang X, Tesema B, Han Y, Zhao J, Girmay S, Ahmad HI. Immunization against Kisspeptin-54 perturb hypothalamic–pituitary–testicular signaling pathway in ram lambs. Theriogenology 2019; 125:193-202. [DOI: 10.1016/j.theriogenology.2018.10.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 02/01/2023]
|
32
|
Ozaki S, Higo S, Iwata K, Saeki H, Ozawa H. Region-specific changes in brain kisspeptin receptor expression during estrogen depletion and the estrous cycle. Histochem Cell Biol 2019; 152:25-34. [PMID: 30671658 DOI: 10.1007/s00418-018-01767-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2018] [Indexed: 11/28/2022]
Abstract
Kisspeptin acts as a potent neuropeptide regulator of reproduction through modulation of the hypothalamic-pituitary-gonadal axis. Previous studies revealed sex differences in brain expression patterns as well as regulation of expression by estrogen. Alternatively, sex differences and estrogen regulation of the kisspeptin receptor (encoded by Kiss1r) have not been examined at cellular resolution. In the current study, we examined whether Kiss1r mRNA expression also exhibits estrogen sensitivity and sex-dependent differences using in situ hybridization. We compared Kiss1r mRNA expression between ovariectomized (OVX) rats and estradiol (E2)-replenished OVX rats to examine estrogen sensitivity, and compared expression between gonadally intact male rats and female rats in diestrus or proestrus to examine sex differences. In OVX rats, E2 replenishment significantly reduced Kiss1r expression specifically in the hypothalamic arcuate nucleus (ARC). A difference in Kiss1r expression was also observed between diestrus and proestrus rats in the hypothalamic paraventricular nucleus (PVN), but not in the ARC. Thus, estrogen appears to have region- and context-specific effects on Kiss1r expression. However, immunostaining revealed minimal colocalization of estrogen receptor alpha (ERα) in Kiss1r-expressing neuronal populations of ARC and PVN, indicating indirect or ERα-independent regulation of Kiss1r expression. Surprisingly, unlike the kisspeptin ligand, no sexual dimorphisms were observed in either the brain distribution of Kiss1r expression or in the number of Kiss1r-expressing neurons within enriched brain nuclei. The current study reveals marked differences in regulation between kisspeptin and kisspeptin receptor, and provides an essential foundation for further study of kisspeptin signaling and function in reproduction.
Collapse
Affiliation(s)
- Saeko Ozaki
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, Sendagi 1-1-5, Bunkyo-ku, Tokyo, 113-8602, Japan.,Department of Dermatology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Shimpei Higo
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, Sendagi 1-1-5, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Kinuyo Iwata
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, Sendagi 1-1-5, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Hidehisa Saeki
- Department of Dermatology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Hitoshi Ozawa
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, Sendagi 1-1-5, Bunkyo-ku, Tokyo, 113-8602, Japan.
| |
Collapse
|
33
|
Smith JJ, Timoshevskaya N, Timoshevskiy VA, Keinath MC, Hardy D, Voss SR. A chromosome-scale assembly of the axolotl genome. Genome Res 2019; 29:317-324. [PMID: 30679309 PMCID: PMC6360810 DOI: 10.1101/gr.241901.118] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 11/26/2018] [Indexed: 01/14/2023]
Abstract
The axolotl (Ambystoma mexicanum) provides critical models for studying regeneration, evolution, and development. However, its large genome (∼32 Gb) presents a formidable barrier to genetic analyses. Recent efforts have yielded genome assemblies consisting of thousands of unordered scaffolds that resolve gene structures, but do not yet permit large-scale analyses of genome structure and function. We adapted an established mapping approach to leverage dense SNP typing information and for the first time assemble the axolotl genome into 14 chromosomes. Moreover, we used fluorescence in situ hybridization to verify the structure of these 14 scaffolds and assign each to its corresponding physical chromosome. This new assembly covers 27.3 Gb and encompasses 94% of annotated gene models on chromosomal scaffolds. We show the assembly's utility by resolving genome-wide orthologies between the axolotl and other vertebrates, identifying the footprints of historical introgression events that occurred during the development of axolotl genetic stocks, and precisely mapping several phenotypes including a large deletion underlying the cardiac mutant. This chromosome-scale assembly will greatly facilitate studies of the axolotl in biological research.
Collapse
Affiliation(s)
- Jeramiah J Smith
- Department of Biology, University of Kentucky, Lexington, Kentucky 40506, USA
| | | | | | - Melissa C Keinath
- Department of Biology, University of Kentucky, Lexington, Kentucky 40506, USA.,Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland 21218, USA
| | - Drew Hardy
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky 40506, USA.,Ambystoma Genetic Stock Center, University of Kentucky, Lexington, Kentucky 40506, USA
| | - S Randal Voss
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky 40506, USA.,Ambystoma Genetic Stock Center, University of Kentucky, Lexington, Kentucky 40506, USA.,Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40506, USA
| |
Collapse
|
34
|
Nomoto K, Ikumi M, Otsuka M, Asaba A, Kato M, Koshida N, Mogi K, Kikusui T. Female mice exhibit both sexual and social partner preferences for vocalizing males. Integr Zool 2019; 13:735-744. [PMID: 30019858 DOI: 10.1111/1749-4877.12357] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Acoustic signals are widely used as courtship signals in the animal kingdom. It has long been known that male mice emit ultrasonic vocalizations (USVs) in the presence of female mice or in response to female secretions. This observation led to the hypothesis that male USVs play a role in courtship behavior. Although previous studies showed that female mice have a social partner preference for vocalizing males, it is not known if they exhibit a sexual partner preference when given a choice. To address this issue, we examined the copulatory behaviors of female mice with either devocalized males (with or without the playback of the USVs) or sham-operated males in 2 different behavioral paradigms: the no-choice paradigm in the home cage of a male mouse (without choice of mating partners) or the mate-choice paradigm in a 3-chambered apparatus (with choice of mating partners). In the no-choice paradigm, female mice exhibited comparable sexual receptivity with sham-operated and devocalized males. In addition, we found that female mice showed more approach behavior towards devocalized males when male USVs were played back. In the mate-choice paradigm, female mice visited more frequently and stayed longer with sham-operated than devocalized males. Furthermore, we showed that female mice received more intromissions from sham-operated males than devocalized males. In summary, our results suggested that, although female mice can copulate equally with both devocalized and vocalizing males when given no choice of mating partner, female mice exhibit both sexual and social partner preferences for vocalizing males in the mate-choice paradigm.
Collapse
Affiliation(s)
- Kensaku Nomoto
- Companion Animal Research Laboratory, School of Veterinary Medicine, Azabu University, Kanagawa, Japan
| | - Mayu Ikumi
- Companion Animal Research Laboratory, School of Veterinary Medicine, Azabu University, Kanagawa, Japan
| | - Monami Otsuka
- Companion Animal Research Laboratory, School of Veterinary Medicine, Azabu University, Kanagawa, Japan
| | - Akari Asaba
- Companion Animal Research Laboratory, School of Veterinary Medicine, Azabu University, Kanagawa, Japan
| | | | - Nobuyoshi Koshida
- Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kazutaka Mogi
- Companion Animal Research Laboratory, School of Veterinary Medicine, Azabu University, Kanagawa, Japan
| | - Takefumi Kikusui
- Companion Animal Research Laboratory, School of Veterinary Medicine, Azabu University, Kanagawa, Japan
| |
Collapse
|
35
|
Zhang H, Zhang B, Qin G, Li S, Lin Q. The Roles of the Kisspeptin System in the Reproductive Physiology of the Lined Seahorse ( Hippocampus erectus), an Ovoviviparous Fish With Male Pregnancy. Front Neurosci 2018; 12:940. [PMID: 30618569 PMCID: PMC6298243 DOI: 10.3389/fnins.2018.00940] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/29/2018] [Indexed: 11/13/2022] Open
Abstract
The kisspeptin/GPR54 system plays a crucial role in the regulation of the reproductive axis in vertebrates. Male pregnancy and ovoviviparity are special reproductive phenomena among vertebrates. To better understand the neuroendocrine mechanisms of male pregnancy, cDNAs encoding kiss2 and GPR54 were cloned and functionally characterized from the lined seahorse, Hippocampus erectus, an ovoviviparous teleost with male pregnancy. The core mature peptide of seahorse Kiss2 is high conserved among seahorses, but unique among vertebrate Kiss orthologs. In the phylogenic analysis, the seahorse Kiss clustered with the teleost Kiss2 clade. The kiss2 transcripts were shown to be widely expressed in various tissues, notably in the brain and gonad of the seahorse, while GPR54-2 mRNA was expressed exclusively in the brain. In addition, kiss2 mRNA found in male seahorse brain tissue increased significantly at the early pubertal stage, and decreased significantly during pregnancy. Intraperitoneal administration of seahorse Kiss2-10 to sexual mature male seahorses demonstrated to stimulate lutropin β (LHβ) and follitropin β (FSHβ) release and increased serum testosterone levels. In summary, we first identified the kisspeptin/GPR54 system in an ovoviviparous fish with male pregnancy, which might be involved in the regulation of the reproductive functions of pubertal onset, gonadal development, and male pregnancy via regulating the synthesis of both gonadotropic hormone (GTH) and testosterone.
Collapse
Affiliation(s)
- Huixian Zhang
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Institution of South China Sea Ecology and Environmental Engineering, Chinese Academy of Sciences, Guangzhou, China
| | - Bo Zhang
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Institution of South China Sea Ecology and Environmental Engineering, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Geng Qin
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Institution of South China Sea Ecology and Environmental Engineering, Chinese Academy of Sciences, Guangzhou, China
| | - Shuisheng Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Qiang Lin
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Institution of South China Sea Ecology and Environmental Engineering, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
36
|
Andrews CJ, Thomas DG, Yapura J, Potter MA. Reproductive biology of the 38 extant felid species: a review. Mamm Rev 2018. [DOI: 10.1111/mam.12145] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Christopher J. Andrews
- Animal Science Group; School of Agriculture and Environment; Massey University; Private Bag 11-222 Palmerston North 4442 New Zealand
| | - David G. Thomas
- Animal Science Group; School of Agriculture and Environment; Massey University; Private Bag 11-222 Palmerston North 4442 New Zealand
| | - Jimena Yapura
- School of Veterinary Science; Massey University; Private Bag 11-222 Palmerston North 4442 New Zealand
| | - Murray A. Potter
- Wildlife and Ecology Group; School of Agriculture and Environment; Massey University; Private Bag 11-222 Palmerston North 4442 New Zealand
| |
Collapse
|
37
|
Olaniyan OT, Kunle-Alabi OT, Raji Y. Protective effects of methanol extract of Plukenetia conophora seeds and 4H-Pyran-4-One 2,3-Dihydro-3,5-Dihydroxy-6-Methyl on the reproductive function of male Wistar rats treated with cadmium chloride. JBRA Assist Reprod 2018; 22:289-300. [PMID: 30106540 PMCID: PMC6210618 DOI: 10.5935/1518-0557.20180048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 06/01/2018] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES Male infertility caused by exposure to heavy metals is a current global issue. Exposure to cadmium chloride (CdCl2) negatively affects the male reproductive system. Many infertile people, especially in developing countries, resort to folkloric treatment. Plukenetia conophora is used in Nigerian folk medicine to promote fertility. This study investigated the effects of Plukenetia conophora (PC) and 4H-Pyran-4-One 2,3-Dihydro-3,5-Dihydroxy-6-Methyl (DDMP) on Wistar rats with cadmium chloride-induced testicular damage. METHODS Forty-two male Wistar rats (150-190g) were divided into seven groups (n=6) and treated daily for 54 days as follows: Controls (normal saline); CdCl2 (2mg/kg single IP dose); CdCl2 + 200 mg/kg vitamin E; CdCl2 + 100 or 200 mg/kg PC; and CdCl2 + 25 or 50 mg/kg DDMP. The rats were sacrificed 55 days after the start of the study; Samples were collected for analysis. Biochemical parameters malondialdehyde, nitric oxide, antioxidant enzymes, and proton pumps were measured by spectrophotometry. Reproductive hormones were measured using ELISA. Data were analysed using ANOVA and differences in mean values were considered significant at p<0.05. RESULTS Significant increases in sperm count, motility, and viability were observed in the groups given CdCl2+Vitamin E, CdCl2+PC or CdCl2+DDMP as compared with the CdCl2 group. Malondialdehyde and nitric oxide levels in the groups treated with CdCl2+PC or CdCl2+DDMP decreased significantly when compared with the group given CdCl2. Significant increases were observed in antioxidant enzymes, proton pump, and testosterone in the groups treated with CdCl2+PC or CdCl2+DDMP, respectively. CONCLUSION Plukenetia conophora alleviated male reproductive toxicity induced by cadmium chloride in Wistar rats. 4H-Pyran-4-One 2,3-Dihydro-3,5-Dihydroxy-6-Methyl present in Plukenetia conophora may be responsible for the ameliorative effects.
Collapse
Affiliation(s)
- Olugbemi Tope Olaniyan
- Department of Physiology, Bingham University Karu, Nasarawa State,
Nigeria
- Department of Physiology, University of Ibadan, Ibadan, Oyo State,
Nigeria
| | | | - Yinusa Raji
- Department of Physiology, University of Ibadan, Ibadan, Oyo State,
Nigeria
| |
Collapse
|
38
|
Estradiol Increases Glutamate and GABA Neurotransmission into GnRH Neurons via Retrograde NO-Signaling in Proestrous Mice during the Positive Estradiol Feedback Period. eNeuro 2018; 5:eN-NWR-0057-18. [PMID: 30079374 PMCID: PMC6073979 DOI: 10.1523/eneuro.0057-18.2018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/26/2018] [Accepted: 07/09/2018] [Indexed: 12/20/2022] Open
Abstract
Surge release of gonadotropin-releasing hormone (GnRH) is essential in the activation of pituitary gonadal unit at proestrus afternoon preceded by the rise of serum 17β-estradiol (E2) level during positive feedback period. Here, we describe a mechanism of positive estradiol feedback regulation acting directly on GnRH-green fluorescent protein (GFP) neurons of mice. Whole-cell clamp and loose patch recordings revealed that a high physiological dose of estradiol (200 pM), significantly increased firing rate at proestrus afternoon. The mPSC frequency at proestrus afternoon also increased, whereas it decreased at metestrus afternoon and had no effect at proestrus morning. Inhibition of the estrogen receptor β (ERβ), intracellular blockade of the Src kinase and phosphatidylinositol 3 kinase (PI3K) and scavenge of nitric oxide (NO) inside GnRH neurons prevented the facilitatory estradiol effect indicating involvement of the ERβ/Src/PI3K/Akt/nNOS pathway in this fast, direct stimulatory effect. Immunohistochemistry localized soluble guanylate cyclase, the main NO receptor, in both glutamatergic and GABAergic terminals innervating GnRH neurons. Accordingly, estradiol facilitated neurotransmissions to GnRH neurons via both GABAA-R and glutamate/AMPA/kainate-R. These results indicate that estradiol acts directly on GnRH neurons via the ERβ/Akt/nNOS pathway at proestrus afternoon generating NO that retrogradely accelerates GABA and glutamate release from the presynaptic terminals contacting GnRH neurons. The newly explored mechanism might contribute to the regulation of the GnRH surge, a fundamental prerequisite of the ovulation.
Collapse
|
39
|
Srivastava VK, Hiney JK, Dees WL. Alcohol Delays the Onset of Puberty in the Female Rat by Altering Key Hypothalamic Events. Alcohol Clin Exp Res 2018; 42:1166-1176. [PMID: 29689132 DOI: 10.1111/acer.13762] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/13/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND Because alcohol (ALC) delays signs of pubertal development, we assessed the time course of events associated with the synthesis of critical hypothalamic peptides that regulate secretion of luteinizing hormone-releasing hormone (LHRH), the peptide that drives the pubertal process. METHODS Immature female rats were administered either laboratory chow or BioServe isocaloric control or ALC-liquid diets from 27 through 33 days of age. On days 28, 29, 31, and 33, animals were killed by decapitation and tissue blocks containing the medial basal hypothalamus (MBH) and the rostral hypothalamic area (RHA) were isolated and stored frozen until assessed by Western blot analysis. RESULTS Synthesis of dynorphin (DYN), a prepubertal inhibitor of LHRH secretion, was increased (p < 0.05) in the MBH of ALC-treated animals by day 29. DYN was further elevated (p < 0.01) on day 33 and was associated with an increase (p < 0.01) in DYN receptor expression. ALC did not affect synthesis of neurokinin B (NKB), a prepubertal stimulator of LHRH; however, it did suppress (p < 0.05) NKB receptor expression in the MBH by day 31. The most potent stimulator of prepubertal LHRH secretion, kisspeptin (Kp), was also decreased (p < 0.05) in the MBH as early as day 29, with continued suppression (p < 0.01) through day 33. Similar timely suppressions of mammalian target of rapamycin (mTOR), an immediate upstream regulator of Kp, were also noted. These decreases in mTOR and Kp were consistent with ALC stimulating (p < 0.05) the p-AMP-activated protein kinase/Raptor inhibitory pathway to mTOR on day 29, then later suppressing (p < 0.001) an Akt-mediated induction pathway to mTOR by day 31. In the RHA, ALC affected the pathways regulating Kp in a manner similar to that described in the MBH; however, these effects were not noted until day 33. CONCLUSIONS ALC acts within the MBH as early as 29 days to induce inhibitor and repressor inputs to LHRH, while depressing stimulatory inputs to the peptide. Collectively, these events lead to delayed signs of pubertal development.
Collapse
Affiliation(s)
- Vinod K Srivastava
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas
| | - Jill K Hiney
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas
| | - William L Dees
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas
| |
Collapse
|
40
|
Xing R, Liu F, Yang Y, Cui X, Wang T, Xie L, Zhao Y, Fang L, Yi T, Zheng B, Liu M, Chen H. GPR54 deficiency reduces the Treg population and aggravates experimental autoimmune encephalomyelitis in mice. SCIENCE CHINA-LIFE SCIENCES 2018; 61:675-687. [PMID: 29931449 DOI: 10.1007/s11427-017-9269-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/17/2017] [Indexed: 12/20/2022]
Abstract
GPR54 is highly expressed in the central nervous system and plays a crucial role in pubertal development. However, GRP54 is also expressed in the immune system, implying possible immunoregulatory functions. Here we investigated the role of GPR54 in T cell and immune tolerance. GPR54 deficiency led to an enlarged thymus, an increased number of thymocytes, and altered thymic micro-architecture starting around puberty, indicating GPR54 function in T-cell development through its regulatory effect on the gonadal system. However, flow cytometry revealed a significant reduction in the peripheral regulatory T cell population and a moderate decrease in CD4 single-positive thymocytes in prepubertal Gpr54-/- mice. These phenotypes were confirmed in chimeric mice with GPR54 deficient bone marrow-derived cells. In addition, we found elevated T cell activation in peripheral and thymic T cells in Gpr54-/- mice. When intact mice were immunized with myelin oligodendrocyte glycoprotein, a more severe experimental autoimmune encephalomyelitis (EAE) developed in the Gpr54-/- mice. Interestingly, aggravated EAE disease was also manifested in castrated and bone marrow chimeric Gpr54-/- mice compared to the respective wild-type control, suggesting a defect in self-tolerance resulting from GPR54 deletion through a mechanism that bypassed sex hormones. These findings demonstrate a novel role for GPR54 in regulating self-tolerant immunity in a sex hormone independent manner.
Collapse
MESH Headings
- Animals
- Disease Susceptibility
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Gene Expression
- Immune Tolerance/immunology
- Lymphocyte Activation/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myelin-Oligodendrocyte Glycoprotein/administration & dosage
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Receptors, Kisspeptin-1/deficiency
- Receptors, Kisspeptin-1/genetics
- Receptors, Kisspeptin-1/physiology
- Spleen/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Regulatory/immunology
- Thymus Gland/immunology
Collapse
Affiliation(s)
- Roumei Xing
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Fang Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yiqing Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xueqin Cui
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Tongtong Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Ling Xie
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yongliang Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Lei Fang
- Third Venture Biotechnology Co., Ltd., Nanjing, 210042, China
| | - Tingfang Yi
- Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas, 77030, USA
| | - Biao Zheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas, 77030, USA.
| | - Huaqing Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
41
|
Shakya M, Shrestha PK, Briski KP. Hindbrain 5'-Adenosine Monophosphate-activated Protein Kinase Mediates Short-term Food Deprivation Inhibition of the Gonadotropin-releasing Hormone-Luteinizing Hormone Axis: Role of Nitric Oxide. Neuroscience 2018; 383:46-59. [PMID: 29746990 DOI: 10.1016/j.neuroscience.2018.04.040] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 04/24/2018] [Accepted: 04/27/2018] [Indexed: 10/17/2022]
Abstract
Hindbrain-derived stimuli restrain the gonadotropin-releasing hormone (GnRH)-pituitary luteinizing hormone (LH) reproductive neuroendocrine axis during energy insufficiency. Interruption of food intake, planned or unplanned, is emblematic of modern life. This study investigated the premise that the hindbrain energy sensor 5'-adenosine monophosphate-activated protein kinase (AMPK) inhibits reproductive neuroendocrine function in short term, e.g. 18-h food-deprived (FD) estradiol (E)-implanted ovariectomized female rats. Intra-caudal fourth ventricular administration of the AMPK inhibitor Compound C (Cc) reversed FD-induced inhibition of rostral preoptic (rPO) GnRH protein expression and LH release in animals given E to replicate proestrus (high-E dose-, but not metestrus (low-E dose)-stage plasma steroid levels. FD caused Cc-reversible augmentation or diminution of preoptic norepinephrine (NE) activity in high- versus low-E rats, respectively, and AMPK-independent reductions in hypothalamic NE accumulation in the latter. Nitric oxide (NO) and kisspeptin are key stimulatory signals for the preovulatory LH surge. Here, FD inhibited rPO neuronal nitric oxide synthase protein expression in high-, but not low-E-dosed animals. Lateral ventricular delivery of the NO donor 3-morpholinosydnonimine (SIN-1) reversed inhibitory GnRH and LH responses to FD in high-E rats, and normalized rPO Vglut2, anteroventral periventricular KiSS1, and dorsomedial hypothalamic RFRP-3 mRNA and/or protein profiles. Data show that FD curtails reproductive neuroendocrine outflow by hindbrain AMPK-dependent mechanisms in the presence of peak estrous cycle E levels. Results indicate that neural networks linking this sensor to GnRH neurons likely involve NO signaling, which may function upstream of one or more neurotransmitters identified here by SIN-1-reversible inhibitory responses to FD.
Collapse
Affiliation(s)
- Manita Shakya
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, College of Health and Pharmaceutical Sciences, The University of Louisiana at Monroe, Monroe, LA 71201, United States
| | - Prem K Shrestha
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, College of Health and Pharmaceutical Sciences, The University of Louisiana at Monroe, Monroe, LA 71201, United States
| | - Karen P Briski
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, College of Health and Pharmaceutical Sciences, The University of Louisiana at Monroe, Monroe, LA 71201, United States.
| |
Collapse
|
42
|
Chen H, Xiao L, Liu Y, Li S, Li G, Zhang Y, Lin H. Neurokinin B signaling in hermaphroditic species, a study of the orange-spotted grouper (Epinephelus coioides). Gen Comp Endocrinol 2018; 260:125-135. [PMID: 29355534 DOI: 10.1016/j.ygcen.2018.01.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/16/2017] [Accepted: 01/11/2018] [Indexed: 12/11/2022]
Abstract
Neurokinin B (NKB) plays important roles in the mammalian reproductive axis by modulating the release of gonadotropin-releasing hormone (GnRH) and gonadotropins. In the present study, the tac3 cDNA was cloned from a hermaphroditic species, the orange-spotted grouper. Sequence analysis showed that the grouper Tac3 precursor encoded two tachykinin peptides, NKB and NKB-related peptide (NKBRP). Expression analysis in different tissues revealed that tac3 mRNA was highly expressed in the brain of the orange-spotted grouper. In situ hybridization further revealed that it was localized in some hypothalamic nuclei associated with reproductive regulation. During ovarian development, an increase of tac3 expression in the hypothalamus was observed at vitellogenesis stage. Intraperitoneal administration of NKB could increase the gnrh1 and lhβ mRNA levels, and enhance the serum estrogen levels, but did not significantly influence lhβ expression in cultured pituitary cells, indicating that NKB does not directly exert its actions on the pituitary gland. However, it was found that NKBRP had no effect on the expression of two gnrhs and two gths in vivo and in vitro. Effects of sex steroids on tac3 expression were further investigated. During the 17-methyltestosterone-induced sex change in the orange-spotted grouper, hypothalamic tac3 expression showed no significant change. Interestingly, ovariectomy greatly stimulated tac3 expression, while the 17β-estradiol treatment reversed this effect. In general, our data highly indicated that NKB signaling could activate the reproductive axis in the orange-spotted grouper. Our study is the first description of the NKB signaling in the hermaphroditic species.
Collapse
Affiliation(s)
- Huapu Chen
- Zhanjiang City Key Laboratory of Marine Ecology and Environment, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
| | - Ling Xiao
- State Key Laboratory of Biocontrol, and the Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China
| | - Yali Liu
- State Key Laboratory of Biocontrol, and the Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China
| | - Shuisheng Li
- State Key Laboratory of Biocontrol, and the Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China.
| | - Guangli Li
- Zhanjiang City Key Laboratory of Marine Ecology and Environment, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China.
| | - Yong Zhang
- State Key Laboratory of Biocontrol, and the Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China
| | - Haoran Lin
- State Key Laboratory of Biocontrol, and the Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
43
|
Zhang J, Jin PP, Gong M, Yi QT, Zhu RJ. Role of leptin and the leptin receptor in the pathogenesis of varicocele-induced testicular dysfunction. Mol Med Rep 2018; 17:7065-7072. [PMID: 29568885 PMCID: PMC5928662 DOI: 10.3892/mmr.2018.8753] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/01/2018] [Indexed: 11/17/2022] Open
Abstract
The present study investigated the expression of leptin and its receptor in the left testis and hypothalamus of rats with varicocele and clarified their roles in the pathogenesis of varicocele-induced testicular dysfunction. A total of 40 male rats were divided randomly into four groups. Groups 1 (G1) and 3 (G3) underwent a sham operation. Groups 2 (G2) and 4 (G4) underwent operations to form a varicocele created by partial ligation of the left renal vein. G1 and G2 rats were euthanized 4 weeks after the operation while G3 and G4 rats were euthanized at 8 weeks. The expression of leptin and its receptor was analyzed by immunohistochemistry. The mRNA levels of leptin, its receptor, kisspeptin (KiSS-1), G-protein coupled receptor 54 (GPR54), gonadotropin releasing hormone (GnRH), luteinizing hormone (LH), and follicle-stimulating hormone (FSH) were measured by reverse transcription-quantitative polymerase chain reaction. Testicular spermatogenesis function and gonadal hormone levels were measured. Compared with G1 and G3, the expression of leptin and its receptor in rat testis was significantly higher in G2 and G4, respectively. Leptin expression was inversely associated with the number of sperm in the left epididymis, thickness of the seminiferous epithelium and the diameter of seminiferous tubules. The expression of leptin receptors in the hypothalamus of G2 and G4 was significantly increased compared with that in G1 and G3, respectively. The mRNA levels of KiSS-1, GPR54, GnRH, LH and FSH in G2 and G4 were significantly increased compared with that in G1 and G3, respectively. Serum testosterone levels in G2 and G4 rats were significantly lower than those in G1 and G3 rats, respectively. There was no significant difference between the serum levels of FSH, LH and leptin. These results suggest that leptin and its receptor may serve significant roles in the pathogenesis of varicocele-induced testicular dysfunction.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Urology, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, P.R. China
| | - Peng-Peng Jin
- Center for Medical Research, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, P.R. China
| | - Min Gong
- Department of Urology, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, P.R. China
| | - Qing-Tong Yi
- Department of Urology, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, P.R. China
| | - Ru-Jian Zhu
- Department of Urology, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, P.R. China
| |
Collapse
|
44
|
Altered aspects of anxiety-related behavior in kisspeptin receptor-deleted male mice. Sci Rep 2018; 8:2794. [PMID: 29434234 PMCID: PMC5809376 DOI: 10.1038/s41598-018-21042-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/29/2018] [Indexed: 12/24/2022] Open
Abstract
The roles of kisspeptin signaling outside the hypothalamus in the brain are unknown. We examined here the impact of Kiss1r-deletion on hippocampus-related behaviors of anxiety and spatial learning in adult male mice using two mouse models. In the first, global Kiss1r-null and control mice were gonadectomized (GDX KISS1R-KO). In the second, KISS1R signalling was rescued selectively in gonadotropin-releasing hormone neurons to generate Kiss1r-null mice with normal testosterone levels (intact KISS1R-KO). Intact KISS1R-KO rescue mice were found to spend twice as much time in the open arms of the elevated plus maze (EPM) compared to controls (P < 0.01). GDX KISS1R-KO mice showed a similar but less pronounced trend. No differences were detected between intact KISS1R-KO mice and controls in the open field test (OFT), although a marked reduction in time spent in the centre quadrant was observed for all GDX mice (P < 0.001). No effects of KISS1R deletion or gonadectomy were detected in the Morris water maze. These observations demonstrate that KISS1R signalling impacts upon anxiogenic neural circuits operative in the EPM, while gonadal steroids appear important for anxiety behaviour observed in the OFT. The potential anxiogenic role of kisspeptin may need to be considered in the development of kisspeptin analogs for the clinic.
Collapse
|
45
|
Li G, Tang H, Chen Y, Yin Y, Ogawa S, Liu M, Guo Y, Qi X, Liu Y, Parhar IS, Liu X, Lin H. Estrogen directly stimulates LHb expression at the pituitary level during puberty in female zebrafish. Mol Cell Endocrinol 2018; 461:1-11. [PMID: 28801227 DOI: 10.1016/j.mce.2017.08.003] [Citation(s) in RCA: 11] [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: 02/17/2017] [Revised: 08/05/2017] [Accepted: 08/06/2017] [Indexed: 11/16/2022]
Abstract
The LHb expression is up-regulated during puberty in female zebrafish. However, the molecular mechanism underlying how LHb expression is regulated during puberty remains largely unknown. In this study, we found that the mRNA expression levels of lhb, fshb and cyp19a1b were up-regulated along with the puberty onset in zebrafish. Among the three nuclear estrogen receptors (nERs), the esr2b is the only type whose expression is significantly up-regulated during puberty onset in the pituitary. However, in situ hybridization results revealed that lhb mRNA was colocalized with esr1 and esr2a but not esr2b. Exposure to estradiol (E2) significantly stimulates LHb expression in both wild-type and kiss1-/-;kiss2-/-;gnrh3-/- triple knockout pubertal zebrafish. Moreover, exposure of cultured pituitary cells to E2 increased the LHb expression, indicating that the estrogenic effect on LHb expression could be acted at the pituitary level. Finally, we cloned and analyzed the promoter of lhb by luciferase assay. Our results indicated that the E2 responsive regions of lhb promoter for ERα and ERβ2 are identical, suggesting that ERα and ERβ2 could bind to the same half ERE region of the promoter of lhb, exhibiting a classical ERE-dependent pathway. In summary, we demonstrate that E2 could directly act on the pituitary level to stimulate LHb transcription during puberty in zebrafish.
Collapse
Affiliation(s)
- Gaofei Li
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Haipei Tang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yu Chen
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yike Yin
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Satoshi Ogawa
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Meifeng Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yin Guo
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Xin Qi
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yun Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Ishwar S Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Xiaochun Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, China.
| | - Haoran Lin
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, China.
| |
Collapse
|
46
|
Riddy DM, Delerive P, Summers RJ, Sexton PM, Langmead CJ. G Protein-Coupled Receptors Targeting Insulin Resistance, Obesity, and Type 2 Diabetes Mellitus. Pharmacol Rev 2018; 70:39-67. [PMID: 29233848 DOI: 10.1124/pr.117.014373] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 09/13/2017] [Indexed: 03/21/2025] Open
Abstract
G protein-coupled receptors (GPCRs) continue to be important discovery targets for the treatment of type 2 diabetes mellitus (T2DM). Many GPCRs are directly involved in the development of insulin resistance and β-cell dysfunction, and in the etiology of inflammation that can lead to obesity-induced T2DM. This review summarizes the current literature describing a number of well-validated GPCR targets, but also outlines several new and promising targets for drug discovery. We highlight the importance of understanding the role of these receptors in the disease pathology, and their basic pharmacology, which will pave the way to the development of novel pharmacological probes that will enable these targets to fulfill their promise for the treatment of these metabolic disorders.
Collapse
Affiliation(s)
- Darren M Riddy
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Philippe Delerive
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Patrick M Sexton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Christopher J Langmead
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| |
Collapse
|
47
|
Hiney JK, Srivastava VK, Vaden Anderson DN, Hartzoge NL, Dees WL. Regulation of Kisspeptin Synthesis and Release in the Preoptic/Anterior Hypothalamic Region of Prepubertal Female Rats: Actions of IGF-1 and Alcohol. Alcohol Clin Exp Res 2017; 42:61-68. [PMID: 29072778 DOI: 10.1111/acer.13539] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 10/18/2017] [Indexed: 12/01/2022]
Abstract
BACKGROUND Alcohol (ALC) causes suppressed secretion of prepubertal luteinizing hormone-releasing hormone (LHRH). Insulin-like growth factor-1 (IGF-1) and kisspeptin (Kp) are major regulators of LHRH and are critical for puberty. IGF-1 may be an upstream mediator of Kp in the preoptic area and rostral hypothalamic area (POA/RHA) of the rat brain, a region containing both Kp and LHRH neurons. We investigated the ability of IGF-1 to stimulate prepubertal Kp synthesis and release in POA/RHA, and the potential inhibitory effects of ALC. METHODS Immature female rats were administered either ALC (3 g/kg) or water via gastric gavage at 0730 hours. At 0900 hours, both groups were subdivided where half received either saline or IGF-1 into the brain third ventricle. A second dose of ALC (2 g/kg) or water was administered at 1130 hours. Rats were killed 6 hours after injection and POA/RHA region collected. RESULTS IGF-1 stimulated Kp, an action blocked by ALC. Upstream to Kp, IGF-1 receptor (IGF-1R) activation, as demonstrated by the increase in insulin receptor substrate 1, resulted in activation of Akt, tuberous sclerosis 2, ras homologue enriched in brain, and mammalian target of rapamycin (mTOR). ALC blocked the central action of IGF-1 to induce their respective phosphorylation. IGF-1 specificity and ALC specificity for the Akt-activated mTOR pathway were demonstrated by the absence of effects on PRAS40. Furthermore, IGF-1 stimulated Kp release from POA/RHA incubated in vitro. CONCLUSIONS IGF-1 stimulates prepubertal Kp synthesis and release following activation of a mTOR signaling pathway, and ALC blocks this pathway at the level of IGF-1R.
Collapse
Affiliation(s)
- Jill K Hiney
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas
| | - Vinod K Srivastava
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas
| | - Danielle N Vaden Anderson
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas
| | - Nicole L Hartzoge
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas
| | - William L Dees
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas
| |
Collapse
|
48
|
Srilanchakon K, Thadsri T, Jantarat C, Thengyai S, Nosoognoen W, Supornsilchai V. Higher phthalate concentrations are associated with precocious puberty in normal weight Thai girls. J Pediatr Endocrinol Metab 2017; 30:1293-1298. [PMID: 29176028 DOI: 10.1515/jpem-2017-0281] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/04/2017] [Indexed: 01/14/2023]
Abstract
BACKGROUND The cause of precocious puberty may be associated with genetics and other conditions such as central nervous system (CNS) insults, or the exposure to endocrine disrupting chemicals (EDCs). Phthalates is known to be one of the EDCs and have estrogenic and antiandrogenic activities, and may be associated with advanced puberty. The objective of the study was to determine the association between urinary phthalate metabolites and advanced puberty. METHODS A cross-sectional study was conducted in patients with precocious puberty (breast onset <8 years, n=42) and early puberty (breast onset 8-9 years, n=17), compared to age-matched controls (n=77). Anthropometric measurements, estradiol, basal and gonadotropin releasing hormone (GnRH)-stimulated follicle stimulating hormone (FSH) and luteinizing hormone (LH) levels, uterine sizes, ovarian diameters and bone ages (BA) were obtained. Urine samples were collected and mono-methyl phthalate (MMP) and mono-ethyl phthalate (MEP) were analyzed by high performance liquid chromatography (HPLC) and adjusted with urine creatinine. RESULTS The median adjusted-MEP concentration in girls with precocious puberty, was greater than in normal girls (6105.09 vs. 4633.98 μg/g Cr: p<0.05), and had the same trend among early puberty and normal puberty (5141.41 vs. 4633.98 μg/g Cr: p=0.4), but was not statistically significant. CONCLUSIONS Precocious puberty girls had an association with increased MEP concentration. This is the first report of the association between urinary phthalate levels and precocious puberty in Thai girls.
Collapse
|
49
|
Adekunbi DA, Li XF, Li S, Adegoke OA, Iranloye BO, Morakinyo AO, Lightman SL, Taylor PD, Poston L, O’Byrne KT. Role of amygdala kisspeptin in pubertal timing in female rats. PLoS One 2017; 12:e0183596. [PMID: 28846730 PMCID: PMC5573137 DOI: 10.1371/journal.pone.0183596] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/07/2017] [Indexed: 01/27/2023] Open
Abstract
To investigate the mechanism by which maternal obesity disrupts reproductive function in offspring, we examined Kiss1 expression in the hypothalamic arcuate (ARC) and anteroventral periventricular (AVPV) nuclei, and posterodorsal medial amygdala (MePD) of pre-pubertal and young adult offspring. Sprague-Dawley rats were fed either a standard or energy-dense diet for six weeks prior to mating and throughout pregnancy and lactation. Male and female offspring were weaned onto normal diet on postnatal day (pnd) 21. Brains were collected on pnd 30 or 100 for qRT-PCR to determine Kiss1 mRNA levels. Maternal obesity increased Kiss1 mRNA expression in the MePD of pre-pubertal male and female offspring, whereas Kiss1 expression was not affected in the ARC or AVPV at this age. Maternal obesity reduced Kiss1 expression in all three brain regions of 3 month old female offspring, but only in MePD of males. The role of MePD kisspeptin on puberty, estrous cyclicity and preovulatory LH surges was assessed directly in a separate group of post-weanling and young adult female rats exposed to a normal diet throughout their life course. Bilateral intra-MePD cannulae connected to osmotic mini-pumps for delivery of kisspeptin receptor antagonist (Peptide 234 for 14 days) were chronically implanted on pnd 21 or 100. Antagonism of MePD kisspeptin delayed puberty onset, disrupted estrous cyclicity and reduced the incidence of LH surges. These data show that the MePD plays a key role in pubertal timing and ovulation and that maternal obesity may act via amygdala kisspeptin signaling to influence reproductive function in the offspring.
Collapse
Affiliation(s)
- Daniel A. Adekunbi
- Division of Women’s Health, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
- Department of Physiology, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Xiao Feng Li
- Division of Women’s Health, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Shengyun Li
- Division of Women’s Health, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Olufeyi A. Adegoke
- Department of Physiology, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Bolanle O. Iranloye
- Department of Physiology, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Ayodele O. Morakinyo
- Department of Physiology, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Stafford L. Lightman
- Henry Wellcome Laboratory for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, United Kingdom
| | - Paul D. Taylor
- Division of Women’s Health, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Lucilla Poston
- Division of Women’s Health, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Kevin T. O’Byrne
- Division of Women’s Health, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| |
Collapse
|
50
|
Diethylstilbestrol administration inhibits theca cell androgen and granulosa cell estrogen production in immature rat ovary. Sci Rep 2017; 7:8374. [PMID: 28827713 PMCID: PMC5567288 DOI: 10.1038/s41598-017-08780-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/17/2017] [Indexed: 01/28/2023] Open
Abstract
Diethylstilbestrol (DES), a strong estrogenic compound, is well-known to affect the reproductive system. In this study, we investigated the effects of DES administration on gonadotropin levels and ovarian steroidogenesis in prepubertal rats. DES treatment acutely reduced serum LH levels, followed by a reduction in the expression of various steroidogenesis-related genes in theca cells. Serum FSH levels were almost unaffected by DES-treatment, even though Cyp19a1 expression was markedly reduced. Serum progesterone, testosterone and estradiol levels were also declined at this time. LH levels recovered from 12 h after DES-treatment and gradually increased until 96 h with a reduction of ERα expression observed in the pituitary. Steroidogenesis-related genes were also up-regulated during this time, except for Cyp17a1 and Cyp19a1. Consistent with observed gene expression pattern, serum testosterone and estradiol concentrations were maintained at lower levels, even though progesterone levels recovered. DES-treatment induced the inducible nitric oxide synthase (iNOS) in granulosa cells, and a nitric oxide generator markedly repressed Cyp19a1 expression in cultured granulosa cells. These results indicate that DES inhibits thecal androgen production via suppression of pituitary LH secretion and ovarian Cyp17a1 expression. In addition, DES represses Cyp19a1 expression by inducing iNOS gene expression for continuous inhibition of estrogen production in granulosa cells.
Collapse
|