1
|
Griesgraber MJ, Coolen LM, Onslow KM, Corey JR, Rice RE, Aerts EG, Bowdridge EC, Hardy SL, Lehman MN, Goodman RL, Hileman SM. Critical role of arcuate nucleus kisspeptin and Kiss1R in regulation of the ovine luteinizing hormone surge. J Neuroendocrinol 2025; 37:e70010. [PMID: 40033679 PMCID: PMC12045732 DOI: 10.1111/jne.70010] [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: 09/27/2024] [Revised: 01/28/2025] [Accepted: 02/13/2025] [Indexed: 03/05/2025]
Abstract
Hypothalamic kisspeptin (Kiss), neurokinin B (NKB), and dynorphin-containing (KNDy) neurons in the arcuate nucleus (ARC) have consistently been shown to be the central generator of gonadotropin-releasing hormone (GnRH) and corresponding luteinizing hormone (LH) pulses in mammals and possibly contribute to surge secretion as well. Additionally, recent evidence from experiments in sheep suggests that ARC Kiss1R-containing neurons play an important role in regulating the timing and amplitude of LH pulses. In this study, we examined the functional role of ARC KNDy and Kiss1R-containing neurons in ovine LH surge secretion via injection of saporin-ligand conjugates (SAP) to ablate these neural populations. NKB-SAP injections significantly reduced the percentage of ARC Kiss1 (~65% decrease) cells compared to control animals, and a surge-like increase of LH was prevented in ewes with the greatest degree of Kiss1 cell ablation. Kiss-SAP injections had no effect on Kiss1 cell percentage or ARC Kiss1R cell number compared to controls, the latter perhaps due to Kiss1R suppression in control animals from elevated estradiol concentrations during the LH surge. However, Kiss-SAP injections consistently and robustly decreased LH surge amplitude, with 80% of Kiss-SAP-treated ewes failing to generate a surge. While the exact identity of these ARC Kiss1R neurons has yet to be fully elucidated, they likely act downstream or in concert with KNDy neurons and possibly integrate other surge-centric signaling pathways to generate the ovine LH surge. These results support the conclusion that KNDy neurons contribute significantly to the ovine LH surge, while ARC Kiss1R neurons appear to be necessary for a functional surge to occur in sheep.
Collapse
Affiliation(s)
- Max J Griesgraber
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
| | - Lique M Coolen
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Kayla M Onslow
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Jacob R Corey
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Rachel E Rice
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Eliana G Aerts
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
| | - Elizabeth C Bowdridge
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
| | - Steven L Hardy
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
| | - Michael N Lehman
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Robert L Goodman
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
- Department of Neuroscience, West Virginia University, Morgantown, West Virginia, USA
| | - Stanley M Hileman
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
- Department of Neuroscience, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
2
|
Reichard TM, Miller CH, Yang J, Sheehan MJ. Seasonality of the estrus cycle in laboratory mice under constant conditions. Lab Anim 2025:236772251318772. [PMID: 40159114 DOI: 10.1177/00236772251318772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Seasonality governs every aspect of life in the natural environment. Controlled laboratory settings are intended to keep animals under a constant set of environmental cues with no seasonality. However, prior research suggests that seasonal variation may exist despite aseasonal lab environments. Here, we examined whether the length of each phase of the estrus cycle varied seasonally in addition to seasonal changes in the overall estrus cycle length in a laboratory mouse strain (C57BL/6J) under standard laboratory housing conditions. We found that female C57BL/6J mice exhibited reproductive seasonality mirroring the outside environment, in a controlled "simulated summer" environment. In the winter and spring, females have longer ovulating phases (proestrus and estrus), compared to the fall. Females similarly experience lengthier quiescent phases (metestrus and diestrus) in the summer, compared to fall and winter. Interestingly, females showed no significant variation in overall estrus cycle length across seasons. Notably, females spent more time in ovulating phases across seasons than previously reported. Laboratory mice are sensitive to external seasonal changes, even when housed in standard laboratory environments designed to control light, temperature, and humidity. Humidity is indicated by some analyses as a potential seasonal cue, however, we cannot rule out other unidentified external cues that may provide information about external seasonal changes. These findings represent just one example of how seasonality may impact mouse physiology in laboratory settings, emphasizing the need to account for such influences in biomedical research and improve environmental control in mouse holding facilities.
Collapse
Affiliation(s)
- Tess M Reichard
- Laboratory for Animal Social Evolution and Recognition, Department of Neurobiology and Behavior, Cornell University, Ithaca, USA
| | - Caitlin H Miller
- Laboratory for Animal Social Evolution and Recognition, Department of Neurobiology and Behavior, Cornell University, Ithaca, USA
- Department of Psychology, University of California-San Diego, La Jolla, CA 92093, USA
| | - Jay Yang
- Laboratory for Animal Social Evolution and Recognition, Department of Neurobiology and Behavior, Cornell University, Ithaca, USA
| | - Michael J Sheehan
- Laboratory for Animal Social Evolution and Recognition, Department of Neurobiology and Behavior, Cornell University, Ithaca, USA
| |
Collapse
|
3
|
Belenichev I, Popazova O, Bukhtiyarova N, Ryzhenko V, Pavlov S, Suprun E, Oksenych V, Kamyshnyi O. Targeting Mitochondrial Dysfunction in Cerebral Ischemia: Advances in Pharmacological Interventions. Antioxidants (Basel) 2025; 14:108. [PMID: 39857442 PMCID: PMC11760872 DOI: 10.3390/antiox14010108] [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: 12/19/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
The study of mitochondrial dysfunction has become increasingly pivotal in elucidating the pathophysiology of various cerebral pathologies, particularly neurodegenerative disorders. Mitochondria are essential for cellular energy metabolism, regulation of reactive oxygen species (ROS), calcium homeostasis, and the execution of apoptotic processes. Disruptions in mitochondrial function, driven by factors such as oxidative stress, excitotoxicity, and altered ion balance, lead to neuronal death and contribute to cognitive impairments in several brain diseases. Mitochondrial dysfunction can arise from genetic mutations, ischemic events, hypoxia, and other environmental factors. This article highlights the critical role of mitochondrial dysfunction in the progression of neurodegenerative diseases and discusses the need for targeted therapeutic strategies to attenuate cellular damage, restore mitochondrial function, and enhance neuroprotection.
Collapse
Affiliation(s)
- Igor Belenichev
- Department of Pharmacology and Medical Formulation with Course of Normal Physiology, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine;
| | - Olena Popazova
- Department of Histology, Cytology and Embryology, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Nina Bukhtiyarova
- Department of Clinical Laboratory Diagnostics, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Victor Ryzhenko
- Department of Medical and Pharmaceutical Informatics and Advanced Technologies, Zaporizhzhia State Medical University, 69000 Zaporizhzhia, Ukraine
| | - Sergii Pavlov
- Department of Clinical Laboratory Diagnostics, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Elina Suprun
- The State Institute of Neurology, Psychiatry and Narcology of the National Academy of Medical Sciences of Ukraine, 46 Academician Pavlov Street, 61076 Kharkov, Ukraine
| | | | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology and Immunology, I. Horbachevsky Ternopil State Medical University, 46001 Ternopil, Ukraine;
| |
Collapse
|
4
|
Joy KP, Chaube R. Kisspeptin control of hypothalamus-pituitary-ovarian functions. VITAMINS AND HORMONES 2024; 127:153-206. [PMID: 39864941 DOI: 10.1016/bs.vh.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The discovery of Kisspeptin (Kiss) has opened a new direction in research on neuroendocrine control of reproduction in vertebrates. Belonging to the RF amide family of peptides, Kiss and its cognate receptor Gpr54 (Kissr) have a long and complex evolutionary history. Multiple forms of Kiss and Kissr are identified in non-mammalian vertebrates, with the exception of birds, and monotreme mammals. However, only a single form of the ligand (KISS1/Kiss1) and receptor (KISS1R/Kiss1r) is retained in higher mammals. Kiss1 is distributed in the hypothalamus-pituitary-gonadal (HPG) axis and its primary function is to stimulate gonadotropin-releasing hormone (GnRH) secretion. Kiss1 neurons are distributed in the rostral periventricular area of the third ventricle (RP3V) and arcuate/infundibular nucleus (ARN/IFN). The ARN/IFN is considered the GnRH pulse generator controlled by steroid negative feedback, and the RP3V neurons is concerned with GnRH surge induced by steroid positive feedback in females. The Kiss1-Kiss1r signaling is important in all aspects of reproduction: puberty onset, maintenance of adult gonadal functions and reproductive aging, and hence assumes therapeutic potentials in the treatment of reproductive dysfunctions and induction of artificial reproduction. This chapter reviews involvement of Kiss1 in the control of the HPG axis functions in female mammals.
Collapse
Affiliation(s)
- K P Joy
- Retired Professor, Department of Zoology, Banaras Hindu University, Varanasi, Uttar pradesh, India.
| | - R Chaube
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar pradesh, India
| |
Collapse
|
5
|
Aerts EG, Griesgraber MJ, Shuping SL, Bowdridge EC, Hardy SL, Goodman RL, Nestor CC, Hileman SM. The effect of NK3-Saporin injection within the arcuate nucleus on puberty, the LH surge, and the response to Senktide in female sheep†. Biol Reprod 2024; 110:275-287. [PMID: 37930247 DOI: 10.1093/biolre/ioad147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/09/2023] [Accepted: 10/24/2023] [Indexed: 11/07/2023] Open
Abstract
The timing of puberty onset is reliant on increased gonadotropin-releasing hormone (GnRH). This elicits a corresponding increase in luteinizing hormone (LH) due to a lessening of sensitivity to the inhibitory actions of estradiol (E2). The mechanisms underlying the increase in GnRH release likely involve a subset of neurons within the arcuate (ARC) nucleus of the hypothalamus that contain kisspeptin, neurokinin B (NKB), and dynorphin (KNDy neurons). We aimed to determine if KNDy neurons in female sheep are critical for: timely puberty onset; the LH surge; and the response to an intravenous injection of the neurokinin-3 receptor (NK3R) agonist, senktide. Prepubertal ewes received injections aimed at the ARC containing blank-saporin (control, n = 5) or NK3-saporin (NK3-SAP, n = 6) to ablate neurons expressing NK3R. Blood samples taken 3/week for 65 days following surgery were assessed for progesterone to determine onset of puberty. Control ewes exhibited onset of puberty at 33.2 ± 3.9 days post sampling initiation, whereas 5/6 NK3-SAP treated ewes didn't display an increase in progesterone. After an artificial LH surge protocol, surge amplitude was lower in NK3-SAP ewes. Finally, ewes were treated with senktide to determine if an LH response was elicited. LH pulses were evident in both groups in the absence of injections, but the response to senktide vs saline was similar between groups. These results show that KNDy cells are necessary for timely puberty onset and for full expresson of the LH surge. The occurrence of LH pulses in NK3-SAP treated ewes may indicate a recovery from an apulsatile state.
Collapse
Affiliation(s)
- Eliana G Aerts
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
| | - Max J Griesgraber
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
| | - Sydney L Shuping
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | | | - Steven L Hardy
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
| | - Robert L Goodman
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
- Department of Neuroscience, West Virginia University, Morgantown, WV 26506, USA
| | - Casey C Nestor
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Stanley M Hileman
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
- Department of Neuroscience, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
6
|
Schroeder HT, De Lemos Muller CH, Heck TG, Krause M, Homem de Bittencourt PI. The dance of proteostasis and metabolism: Unveiling the caloristatic controlling switch. Cell Stress Chaperones 2024; 29:175-200. [PMID: 38331164 PMCID: PMC10939077 DOI: 10.1016/j.cstres.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 02/10/2024] Open
Abstract
The heat shock response (HSR) is an ancient and evolutionarily conserved mechanism designed to restore cellular homeostasis following proteotoxic challenges. However, it has become increasingly evident that disruptions in energy metabolism also trigger the HSR. This interplay between proteostasis and energy regulation is rooted in the fundamental need for ATP to fuel protein synthesis and repair, making the HSR an essential component of cellular energy management. Recent findings suggest that the origins of proteostasis-defending systems can be traced back over 3.6 billion years, aligning with the emergence of sugar kinases that optimized glycolysis around 3.594 billion years ago. This evolutionary connection is underscored by the spatial similarities between the nucleotide-binding domain of HSP70, the key player in protein chaperone machinery, and hexokinases. The HSR serves as a hub that integrates energy metabolism and resolution of inflammation, further highlighting its role in maintaining cellular homeostasis. Notably, 5'-adenosine monophosphate-activated protein kinase emerges as a central regulator, promoting the HSR during predominantly proteotoxic stress while suppressing it in response to predominantly metabolic stress. The complex relationship between 5'-adenosine monophosphate-activated protein kinase and the HSR is finely tuned, with paradoxical effects observed under different stress conditions. This delicate equilibrium, known as caloristasis, ensures that cellular homeostasis is maintained despite shifting environmental and intracellular conditions. Understanding the caloristatic controlling switch at the heart of this interplay is crucial. It offers insights into a wide range of conditions, including glycemic control, obesity, type 2 diabetes, cardiovascular and neurodegenerative diseases, reproductive abnormalities, and the optimization of exercise routines. These findings highlight the profound interconnectedness of proteostasis and energy metabolism in cellular function and adaptation.
Collapse
Affiliation(s)
- Helena Trevisan Schroeder
- Laboratory of Cellular Physiology (FisCel) Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Carlos Henrique De Lemos Muller
- Laboratory of Inflammation, Metabolism and Exercise Research (LAPIMEX), Department of Physiology, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Thiago Gomes Heck
- Post Graduate Program in Integral Health Care (PPGAIS-UNIJUÍ/UNICRUZ/URI), Regional University of Northwestern Rio Grande Do Sul State (UNIJUI) and Post Graduate Program in Mathematical and Computational Modeling (PPGMMC), UNIJUI, Ijuí, Rio Grande do Sul, Brazil
| | - Mauricio Krause
- Laboratory of Inflammation, Metabolism and Exercise Research (LAPIMEX), Department of Physiology, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Paulo Ivo Homem de Bittencourt
- Laboratory of Cellular Physiology (FisCel) Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
7
|
Peltoniemi O, Tanskanen T, Kareskoski M. One Health challenges for pig reproduction. Mol Reprod Dev 2023; 90:420-435. [PMID: 36638261 DOI: 10.1002/mrd.23666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/15/2023]
Abstract
The current state of the world challenges pig reproduction as an important part of One Health, which involves interrelationships between animal, human and environmental health. The One Health concept underlines a comparative aspect in reproductive physiology and disease occurrence, bridging knowledge from one species to another. Seasonal changes in the environment affect pig reproduction and climate change may further strengthen those effects. Endocrine-disrupting chemicals (EDCs), and specifically phthalates and heavy metals, interfere with endocrine function, and thereby sexual behavior, fertilization capacity and steroidogenesis. Reproductive infections and extended semen storage are important indications for antimicrobial use. Innovative solutions are needed to explore alternatives to antimicrobials. Efforts to ensure reproductive efficiency have prolonged farrowing as litter size has doubled over the past three decades, compromising immune transfer and welfare. Physiological, metabolic and programming related events around parturition are key areas for future One Health research in pig reproduction. In conclusion, climate change challenges reproductive management and breeding. More resilient pigs that can tolerate harsh environment but maintain high reproductive performance are needed. EDCs continue to grow as an environmental challenge for reproductive management and alternatives to antibiotics will be required.
Collapse
Affiliation(s)
- Olli Peltoniemi
- Department of Production Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Faculty of Veterinary Medicine, Helsinki One Health, University of Helsinki, Helsinki, Finland
| | - Topi Tanskanen
- Faculty of Veterinary Medicine, Helsinki One Health, University of Helsinki, Helsinki, Finland
| | - Maria Kareskoski
- Department of Production Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
8
|
Merchán M, Coveñas R, Plaza I, Abecia JA, Palacios C. Anatomy of hypothalamic and diencephalic nuclei involved in seasonal fertility regulation in ewes. Front Vet Sci 2023; 10:1101024. [PMID: 36876003 PMCID: PMC9978410 DOI: 10.3389/fvets.2023.1101024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
In this study, we describe in detail the anatomy of nuclei involved in seasonal fertility regulation (SFR) in ewes. For this purpose, the intergeniculate leaflet of the visual thalamus, the caudal hypothalamic arcuate nucleus, and suprachiasmatic, paraventricular and supraoptic nuclei of the rostral hypothalamus were morphometrically and qualitatively analyzed in Nissl-stained serial sections, in the three anatomical planes. In addition, data were collected on calcium-binding proteins and cell phenotypes after immunostaining alternate serial sections for calretinin, parvalbumin and calbindin. For a complete neuroanatomical study, glial architecture was assessed by immunostaining and analyzing alternate sections for glial fibrillary acidic protein (GFAP) and ionized calcium-binding adapter molecule 1 (IBA1). The results showed a strong microglial and astroglia reaction around the hypothalamic nuclei of interest and around the whole 3rd ventricle of the ewe brain. Moreover, we correlated cytoarchitectonic coordinates of panoramic serial sections with their macroscopic localization and extension in midline sagittal-sectioned whole brain to provide guidelines for microdissecting nuclei involved in SFR.
Collapse
Affiliation(s)
- Miguel Merchán
- Animal Production Area, Department of Construction and Agronomy, Faculty of Agricultural and Environmental Sciences, University of Salamanca, Salamanca, Spain.,Laboratory of Neuroanatomy of the Peptidergic Systems, Institute for Neuroscience of Castilla y León (INCYL), University of Salamanca, Salamanca, Spain.,Recognized Research Group - Molecular Bases of Development (Grupo de Investigación Reconocido - Bases Moleculares del Desarrollo - GIR-BMD), University of Salamanca, Salamanca, Spain
| | - Rafael Coveñas
- Laboratory of Neuroanatomy of the Peptidergic Systems, Institute for Neuroscience of Castilla y León (INCYL), University of Salamanca, Salamanca, Spain.,Recognized Research Group - Molecular Bases of Development (Grupo de Investigación Reconocido - Bases Moleculares del Desarrollo - GIR-BMD), University of Salamanca, Salamanca, Spain
| | - Ignacio Plaza
- Auditory Neuroplasticity Laboratory, Institute for Neuroscience of Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - José Alfonso Abecia
- Environmental Science Institute (IUCA), University of Zaragoza, Zaragoza, Spain
| | - Carlos Palacios
- Animal Production Area, Department of Construction and Agronomy, Faculty of Agricultural and Environmental Sciences, University of Salamanca, Salamanca, Spain
| |
Collapse
|
9
|
Exogenous Melatonin Regulates Puberty and the Hypothalamic GnRH-GnIH System in Female Mice. Brain Sci 2022; 12:brainsci12111550. [PMID: 36421874 PMCID: PMC9688274 DOI: 10.3390/brainsci12111550] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022] Open
Abstract
In recent years, the age of children entering puberty is getting lower and the incidence of central precocious puberty is increasing. It is known that melatonin plays an increasingly important role in regulating animal reproduction, but the specific role and mechanism of melatonin in regulating the initiation of puberty remain unclear. The purpose of the current study was to investigate the effect of subcutaneous melatonin injection on pubertal development in female mice and its mechanism of action. Female mice that were 22 days old received 1 mg/kg doses of melatonin subcutaneously every day for 10, 15 and 20 days. The vaginal opening was checked daily. Hematoxylin and eosin (HE) stain was used to determine the growth of the uterus and ovaries. Enzyme-linked immunosorbent assay (ELISA) was used to determine the levels of follicle-stimulating hormone (FSH), gonadotropin-inhibiting hormone (GnIH), and gonadotropin-releasing hormone (GnRH) in serum. By using RT-PCR and Western blotting, the mRNA and protein expression of the hypothalamus GnRH, GnIH, Kisspeptin (Kp), Proopiomelanocortin (POMC), Neuropeptide Y (NPY), as well as G protein-coupled receptor 147 (GPR147) were identified. The findings demonstrated that melatonin could suppress ovarian follicle and uterine wall growth as well as delay vaginal opening, decrease serum levels of GnRH and FSH and increase levels of GnIH. Melatonin increased GnIH and GPR147 expression in the hypothalamus in comparison to the saline group, while decreasing the expression of GnRH, Kisspeptin, POMC, and NPY. In conclusion, exogenous melatonin can inhibit the onset of puberty in female mice by modulating the expression of hypothalamic GnRH, GnIH, Kisspeptin, POMC and NPY neurons and suppressing the hypothalamic–pituitary–gonadal axis.
Collapse
|
10
|
Dardente H, Lomet D, Desmarchais A, Téteau O, Lasserre O, Gonzalez AA, Dubois E, Beltramo M, Elis S. Impact of food restriction on the medio-basal hypothalamus of intact ewes as revealed by a large-scale transcriptomics study. J Neuroendocrinol 2022; 34:e13198. [PMID: 36168278 DOI: 10.1111/jne.13198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 11/27/2022]
Abstract
In mammals, the medio-basal hypothalamus (MBH) integrates photoperiodic and food-related cues to ensure timely phasing of physiological functions, including seasonal reproduction. The current human epidemics of obesity and associated reproductive disorders exemplifies the tight link between metabolism and reproduction. Yet, how food-related cues impact breeding at the level of the MBH remains unclear. In this respect, the sheep, which is a large diurnal mammal with a marked dual photoperiodic/metabolic control of seasonal breeding, is a relevant model. Here, we present a large-scale study in ewes (n = 120), which investigated the impact of food restriction (FRes) on the MBH transcriptome using unbiased RNAseq, followed by RT-qPCR. Few genes (~100) were impacted by FRes and the transcriptional impact was very modest (<2-fold increase or < 50% decrease for most genes). As anticipated, FRes increased expression of Npy/AgRP/LepR and decreased expression of Pomc/Cartpt, while Kiss1 expression was not impacted. Of particular interest, Eya3, Nmu and Dio2, genes involved in photoperiodic decoding within the MBH, were also affected by FRes. Finally, we also identified a handful of genes not known to be regulated by food-related cues (e.g., RNase6, HspA6, Arrdc2). In conclusion, our transcriptomics study provides insights into the impact of metabolism on the MBH in sheep, which may be relevant to human, and identifies possible molecular links between metabolism and (seasonal) reproduction.
Collapse
Affiliation(s)
- Hugues Dardente
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Didier Lomet
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | | | - Ophélie Téteau
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | | | - Anne-Alicia Gonzalez
- MGX-Montpellier GenomiX, Université Montpellier, CNRS, INSERM, Montpellier, France
| | - Emeric Dubois
- MGX-Montpellier GenomiX, Université Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Sébastien Elis
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| |
Collapse
|
11
|
Dai Pra R, Mohr SM, Merriman DK, Bagriantsev SN, Gracheva EO. Ground squirrels initiate sexual maturation during hibernation. Curr Biol 2022; 32:1822-1828.e4. [PMID: 35245461 DOI: 10.1016/j.cub.2022.02.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/15/2021] [Accepted: 02/09/2022] [Indexed: 12/01/2022]
Abstract
Adequate nutrition is essential for normal reproductive function, which is vital for species to survive. In humans and other mammals, starvation and undernutrition deplete fat reserves and cause weight loss, attenuating the function of the reproductive axis and causing hypogonadism.1-4 Thirteen-lined ground squirrels (Ictidomys tridecemlineatus) spend 7 months of every year in hibernation without food and water. Hibernating squirrels alternate between periods of torpor and interbout arousal (IBA), when animals temporarily return to an active-like state.5 The physiological significance of IBA is unclear, but it is thought to be essential for hibernation in animals that drop their body temperature to 2°C-4°C during torpor. Here, we report that juvenile male ground squirrels initiate reproductive maturation during their first hibernation season, despite prolonged undernutrition and profound weight loss. We show that the hypothalamic reproductive axis undergoes activation during interbout arousals in the middle of hibernation, triggering production of luteinizing hormone and testosterone, and promoting testicular growth. Initiation of sexual maturation is circannually entrained and is independent of physiological state, ambient temperature, and food availability. Our study suggests a role for interbout arousals during hibernation and uncovers the neurophysiological mechanism of reproductive axis activation during conditions of extreme negative energy balance.
Collapse
Affiliation(s)
- Rafael Dai Pra
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Sarah M Mohr
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Dana K Merriman
- Department of Biology, University of Wisconsin-Oshkosh, 800 Algoma Boulevard, Oshkosh, WI 54901, USA
| | - Sviatoslav N Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA.
| | - Elena O Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA.
| |
Collapse
|
12
|
Khan S, Batool B, Zubair H, Bano R, Ahmad S, Shahab M. Expression and co-localization of RFRP-3 and kisspeptin during breeding and non-breeding season in the hypothalamus of male rhesus monkey ( Macaca mulatta). Reprod Med Biol 2022; 21:e12479. [PMID: 35847413 PMCID: PMC9270642 DOI: 10.1002/rmb2.12479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/12/2022] [Accepted: 06/21/2022] [Indexed: 12/01/2022] Open
Abstract
Propose The mechanism that underpins how RFRP-3 and kisspeptin interacts are not fully understood in higher primates. This study therefore set out to assess RFRP-3 and kisspeptin expression and their morphological interactions in the breeding, and in the non-breeding period in monkey hypothalamus. Methods Eight mature male macaques (Macaca mulatta) in the breeding season (February; n = 4) and non-breeding season (June; n = 4) were used. To reveal the expression and co-localization of RFRP-3 and kisspeptin, double-labeled immunohistochemistry was performed. Testicular volume, sperm count, and plasma testosterone level were also measured to validate the breeding and non-breeding paradigms. Results Testicular volume, plasma testosterone level, and sperm count showed a significant reduction during non-breeding season. The number of kisspeptin-positive cells was significantly increased during the breeding season (p < 0.05), whereas more RFRP-3-positive cell bodies were seen in the non-breeding season (p < 0.01). Close contacts of RFRP-3 fibers with kisspeptin cells showed no significant difference (p > 0.05) across seasons. However, co-localization of RFRP-3-ir cell bodies onto kisspeptin IR cell bodies showed a statistical increase (p < 0.01) in non-breeding season. Conclusion In higher primates, RFRP-3 decreases kisspeptin drives from the same cells to GnRH neurons in an autocrine manner causing suppression of the reproductive axis during the non-breeding period.
Collapse
Affiliation(s)
- Safdar Khan
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Bakhtwar Batool
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Hira Zubair
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Riffat Bano
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Shakil Ahmad
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Muhammad Shahab
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
- Shaheed Benazir Bhutto University, SheringalDir UpperPakistan
| |
Collapse
|
13
|
Gloria A, Contri A, Mele E, Fasano S, Pierantoni R, Meccariello R. Kisspeptin Receptor on the Sperm Surface Reflects Epididymal Maturation in the Dog. Int J Mol Sci 2021; 22:ijms221810120. [PMID: 34576283 PMCID: PMC8466692 DOI: 10.3390/ijms221810120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 11/16/2022] Open
Abstract
Alongside the well-known central modulatory role, the Kisspeptin system, comprising Kiss1, its cleavage products (Kisspeptins), and Kisspeptin receptor (Kiss1R), was found to regulate gonadal functions in vertebrates; however, its functional role in the male gamete and its localization during maturation have been poorly understood. The present study analyzed Kisspeptin system in dog testis and spermatozoa recovered from different segments of the epididymis, with focus on Kiss1R on sperm surface alongside the maturation during epididymal transit, demonstrated by modification in sperm kinetic, morphology, and protamination. The proteins Kiss1 and Kiss1R were detected in dog testis. The receptor Kiss1R only was detected in total protein extracts from epididymis spermatozoa, whereas dot blot revealed Kiss1 immunoreactivity in the epidydimal fluid. An increase of the Kiss1R protein on sperm surface along the length of the epididymis, with spermatozoa in the tail showing plasma membrane integrity and Kiss1R protein (p < 0.05 vs. epididymis head and body) was observed by flow cytometry and further confirmed by epifluorescence microscopy and Western blot carried on sperm membrane preparations. In parallel, during the transit in the epididymis spermatozoa significantly modified their ability to move and the pattern of motility; a progressive increase in protaminization also occurred. In conclusion, Kisspeptin system was detected in dog testis and spermatozoa. Kiss1R trafficking toward plasma membrane along the length of the epididymis and Kiss1 in epididymal fluid suggested a new functional role of the Kisspeptin system in sperm maturation and storage.
Collapse
Affiliation(s)
- Alessia Gloria
- Faculty of Veterinary Medicine, University of Teramo, Loc. Piano d’Accio, 64100 Teramo, Italy;
| | - Alberto Contri
- Faculty of Biosciences and Technologies for Agriculture Food and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
- Correspondence: (A.C.); (R.M.)
| | - Elena Mele
- Department of Movement Sciences and Wellbeing, Parthenope University of Naples, 80133 Naples, Italy;
| | - Silvia Fasano
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.F.); (R.P.)
| | - Riccardo Pierantoni
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.F.); (R.P.)
| | - Rosaria Meccariello
- Department of Movement Sciences and Wellbeing, Parthenope University of Naples, 80133 Naples, Italy;
- Correspondence: (A.C.); (R.M.)
| |
Collapse
|
14
|
Lagares MDA, Varago FC, Moustacas VS, Gheller VA, Nicolino RR, Borges I, Henry M. Effect of season and frequency of embryo collections on superovulatory response and embryo recovery in Santa Inês hair sheep. Small Rumin Res 2021. [DOI: 10.1016/j.smallrumres.2021.106441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
15
|
Butruille L, Vancamp P, Demeneix BA, Remaud S. Thyroid hormone regulation of adult neural stem cell fate: A comparative analysis between rodents and primates. VITAMINS AND HORMONES 2021; 116:133-192. [PMID: 33752817 DOI: 10.1016/bs.vh.2021.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Thyroid hormone (TH) signaling, a highly conserved pathway across vertebrates, is crucial for brain development and function throughout life. In the adult mammalian brain, including that of humans, multipotent neural stem cells (NSCs) proliferate and generate neuronal and glial progenitors. The role of TH has been intensively investigated in the two main neurogenic niches of the adult mouse brain, the subventricular and the subgranular zone. A key finding is that T3, the biologically active form of THs, promotes NSC commitment toward a neuronal fate. In this review, we first discuss the roles of THs in the regulation of adult rodent neurogenesis, as well as how it relates to functional behavior, notably olfaction and cognition. Most research uncovering these roles of TH in adult neurogenesis was conducted in rodents, whose genetic background, brain structure and rate of neurogenesis are considerably different from that of humans. To bridge the phylogenetic gap, we also explore the similarities and divergences of TH-dependent adult neurogenesis in non-human primate models. Lastly, we examine how photoperiodic length changes TH homeostasis, and how that might affect adult neurogenesis in seasonal species to increase fitness. Several aspects by which TH acts on adult NSCs seem to be conserved among mammals, while we only start to uncover the molecular pathways, as well as how other in- and extrinsic factors are intertwined. A multispecies approach delivering more insights in the matter will pave the way for novel NSC-based therapies to combat neurological disorders.
Collapse
Affiliation(s)
- Lucile Butruille
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Pieter Vancamp
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Barbara A Demeneix
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Sylvie Remaud
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France.
| |
Collapse
|
16
|
Lakhssassi K, Serrano M, Lahoz B, Sarto MP, Iguácel LP, Folch J, Alabart JL, Calvo JH. The LEPR Gene Is Associated with Reproductive Seasonality Traits in Rasa Aragonesa Sheep. Animals (Basel) 2020; 10:ani10122448. [PMID: 33371230 PMCID: PMC7766475 DOI: 10.3390/ani10122448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/07/2020] [Accepted: 12/18/2020] [Indexed: 01/23/2023] Open
Abstract
The aim of this study was to characterize and identify causative polymorphisms in the leptin receptor (LEPR) gene responsible for the seasonal variation of reproductive traits in sheep. Three reproductive seasonality traits were studied: the total days of anoestrous (TDA), the progesterone cycling months (P4CM) and the oestrous cycling months (OCM). In total, 18 SNPs were detected in 33 ewes with extreme values for TDA and OCM. Six SNPs were non-synonymous substitutions and two of them were predicted in silico as deleterious: rs596133197 and rs403578195. These polymorphisms were then validated in 239 ewes. The SNP rs403578195, located in exon 8 and leading to a change of alanine to glycine (Ala284Gly) in the extracellular domain of the protein, was associated with the OCM trait, being the G allele associated with a decrease of 12 percent of the OCM trait. Haplotype analyses also suggested the involvement of other non-synonymous SNP located in exon 20 (rs405459906). This SNP also produces an amino acid change (Lys1069Glu) in the intracellular domain of the protein and segregates independently of rs403578195. These results confirm for the first time the role of the LEPR gene in sheep reproductive seasonality.
Collapse
Affiliation(s)
- Kenza Lakhssassi
- Centro de Investigación y Tecnología Agroalimentaria de Aragón, Instituto Agroalimentario de Aragón (IA2), CITA–Zaragoza University, 50059 Zaragoza, Spain; (K.L.); (B.L.); (M.P.S.); (L.P.I.); (J.F.); (J.L.A.)
| | - Malena Serrano
- Departamento de Mejora Genética Animal INIA, 28040 Madrid, Spain;
| | - Belén Lahoz
- Centro de Investigación y Tecnología Agroalimentaria de Aragón, Instituto Agroalimentario de Aragón (IA2), CITA–Zaragoza University, 50059 Zaragoza, Spain; (K.L.); (B.L.); (M.P.S.); (L.P.I.); (J.F.); (J.L.A.)
| | - María Pilar Sarto
- Centro de Investigación y Tecnología Agroalimentaria de Aragón, Instituto Agroalimentario de Aragón (IA2), CITA–Zaragoza University, 50059 Zaragoza, Spain; (K.L.); (B.L.); (M.P.S.); (L.P.I.); (J.F.); (J.L.A.)
| | - Laura Pilar Iguácel
- Centro de Investigación y Tecnología Agroalimentaria de Aragón, Instituto Agroalimentario de Aragón (IA2), CITA–Zaragoza University, 50059 Zaragoza, Spain; (K.L.); (B.L.); (M.P.S.); (L.P.I.); (J.F.); (J.L.A.)
| | - José Folch
- Centro de Investigación y Tecnología Agroalimentaria de Aragón, Instituto Agroalimentario de Aragón (IA2), CITA–Zaragoza University, 50059 Zaragoza, Spain; (K.L.); (B.L.); (M.P.S.); (L.P.I.); (J.F.); (J.L.A.)
| | - José Luis Alabart
- Centro de Investigación y Tecnología Agroalimentaria de Aragón, Instituto Agroalimentario de Aragón (IA2), CITA–Zaragoza University, 50059 Zaragoza, Spain; (K.L.); (B.L.); (M.P.S.); (L.P.I.); (J.F.); (J.L.A.)
| | - Jorge Hugo Calvo
- Centro de Investigación y Tecnología Agroalimentaria de Aragón, Instituto Agroalimentario de Aragón (IA2), CITA–Zaragoza University, 50059 Zaragoza, Spain; (K.L.); (B.L.); (M.P.S.); (L.P.I.); (J.F.); (J.L.A.)
- ARAID, 50018 Zaragoza, Spain
- Correspondence: ; Tel.: +34-976-716-471
| |
Collapse
|
17
|
Cisint S, Crespo CA, Iruzubieta Villagra L, Fernández SN, Ramos I. Effect of nervous stimulation on ovarian steroid secretion in amphibians. JOURNAL OF EXPERIMENTAL ZOOLOGY PART 2020; 333:681-691. [PMID: 33058568 DOI: 10.1002/jez.2421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 11/09/2022]
Abstract
In the present study, the effect of nerve stimulation on the secretory activity of the ovary of adult females was analyzed for the first time in amphibians. Results revealed that in Rhinella arenarum the stimulation of nerves that supply the gonad induced an increase in estradiol and progesterone secretion, this response showing differences during the reproductive cycle of the species. During the postreproductive period, an increase in estradiol secretion was observed while, in the reproductive period, progesterone secretion increased. Our results suggest that the sympathetic division of the autonomic nervous system would be responsible for this increase, taking into account that, under our experimental conditions, acetylcholine did not affect the endocrine activity of the gonad, while adrenaline (epinephrine) was effective in inducing steroid secretion an effect that could be due to interaction with β receptors. On the other hand, our data show that the association of adrenaline with follicle-stimulating hormone increased estradiol secretion during the postreproductive period, while the association of catecholamine with LH or hCG increased progesterone secretion during the reproductive period. Our results would suggest that nerve stimulation, mediated by the release of adrenaline, would act synergistically with gonadotrophins to stimulate steroid secretion.
Collapse
Affiliation(s)
- Susana Cisint
- Institute of Biology, Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucumán, Chacabuco, Tucumán, Argentina
| | - Claudia A Crespo
- Institute of Biology, Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucumán, Chacabuco, Tucumán, Argentina.,Higher Institute of Biological Research, National Council for Scientific and Technical Research, National University of Tucumán, Chacabuco, Tucumán, Argentina
| | - Lucrecia Iruzubieta Villagra
- Institute of Biology, Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucumán, Chacabuco, Tucumán, Argentina
| | - Silvia N Fernández
- Institute of Biology, Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucumán, Chacabuco, Tucumán, Argentina
| | - Inés Ramos
- Institute of Biology, Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucumán, Chacabuco, Tucumán, Argentina.,Higher Institute of Biological Research, National Council for Scientific and Technical Research, National University of Tucumán, Chacabuco, Tucumán, Argentina
| |
Collapse
|
18
|
Zou K, Asiamah CA, Lu LL, Liu Y, Pan Y, Chen T, Zhao Z, Su Y. Ovarian transcriptomic analysis and follicular development of Leizhou black duck. Poult Sci 2020; 99:6173-6187. [PMID: 33142535 PMCID: PMC7647846 DOI: 10.1016/j.psj.2020.08.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 08/10/2020] [Accepted: 08/15/2020] [Indexed: 12/19/2022] Open
Abstract
This study investigated the factors that caused the differences in egg production during the development of ovarian follicles in Leizhou black ducks. Leizhou black ducks population was divided into 2 groups as high-yield group (HG) and low-yield group (LG). The number of eggs (NE), age at first egg (AFE), weight at first egg, and egg weight (EW) of both groups were recorded, and differences were analyzed using the t test. The logistic model was used to simulate the egg production curves to analyze the production rules. The ovarian follicles of both duck groups were collected to count the number of different grades sized follicles, weigh the ovaries, and observe follicular sections to analyze the developmental differences. Ovarian transcriptomic sequencing was performed to investigate differentially expressed genes and signal pathways in both duck groups. The results revealed a significant difference (P < 0.01) in the NE laid, AFE, and EW between both groups. Comparatively, HG had significantly more (P < 0.01) large yellow follicles (LYF) than LG. The density of medullary layer cells of the follicle section was greater in HG than LG ducks. Transcriptome sequencing revealed a total of 1,027 differentially expressed genes between the HG and LG ducks of which 495 genes were upregulated, and 532 genes were downregulated. Fifty genes were related to reproduction and reproductive processes. Kyoto Encyclopedia of Genes and Genomes–enriched signaling pathways revealed 274 signal pathways enriched in these differentially expressed genes of which the steroid biosynthesis pathway was significantly enriched. Analysis (Q < 0.05) showed that HSD3β → gonadotropin-releasing hormone (GnRH) and estrogen receptor (ESR) → LHβ/ERK1/2 were enriched in the steroid biosynthesis signal pathway. Follicle-stimulating hormone signal pathway mediated by HSD3β → GnRH and ESR → LHβ/ERK1/2 may be involved in ovarian follicle development to regulate LYF reserve process and affect its ovulation cycle, which in turn influence the egg production of Leizhou black ducks.
Collapse
Affiliation(s)
- Kun Zou
- College of Agriculture, Guangdong Ocean University, Zhanjiang 524025, PR China
| | | | - Li-Li Lu
- College of Agriculture, Guangdong Ocean University, Zhanjiang 524025, PR China
| | - Yuanbo Liu
- College of Agriculture, Guangdong Ocean University, Zhanjiang 524025, PR China
| | - Yiting Pan
- College of Agriculture, Guangdong Ocean University, Zhanjiang 524025, PR China
| | - Tongxin Chen
- College of Agriculture, Guangdong Ocean University, Zhanjiang 524025, PR China
| | - Zhihui Zhao
- College of Agriculture, Guangdong Ocean University, Zhanjiang 524025, PR China.
| | - Ying Su
- College of Agriculture, Guangdong Ocean University, Zhanjiang 524025, PR China.
| |
Collapse
|
19
|
Lomet D, Druart X, Hazlerigg D, Beltramo M, Dardente H. Circuit-level analysis identifies target genes of sex steroids in ewe seasonal breeding. Mol Cell Endocrinol 2020; 512:110825. [PMID: 32422398 DOI: 10.1016/j.mce.2020.110825] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/26/2020] [Accepted: 04/19/2020] [Indexed: 02/04/2023]
Abstract
Thyroid hormone (TH) and estradiol (E2) direct seasonal switches in ovine reproductive physiology. In sheep, as in other mammals and birds, control of thyrotropin (TSH) production by the pars tuberalis (PT) links photoperiod responsiveness to seasonal breeding. PT-derived TSH governs opposite seasonal patterns of the TH deiodinases Dio2/Dio3 expression in tanycytes of the neighboring medio-basal hypothalamus (MBH), which explain the key role of TH. We recently used RNA-Seq to identify seasonal markers in the MBH and define the impact of TH. This impact was found to be quite limited, in terms of number of target genes, and very restricted with regards to neuroanatomical location, as TH specifically impacts genes expressed in tanycytes and hypothalamus, not in the PT. Here we address the impact of E2 on these seasonal markers, which are specifically expressed in either PT, tanycytes or hypothalamus. We also investigate if progesterone (P4) may be involved in timing the seasonal transition to anestrus. Our analysis provides circuit-level insights into the impact of sex steroids on the ewe seasonal breeding cycle. First, seasonal gene expression in the PT is independent of the sex steroid status. The fact that seasonal gene expression in the PT is also TH-independent strengthens the view that the PT is a circannual timer. Second, select tanycytic markers display some level of responsiveness to E2 and P4, which indicates another potential level of feedback control by sex steroids. Third, Kiss1 neurons of the arcuate nucleus are responsive to both TH and E2, which places them at the crossroads of photoperiodic transduction pathway and sex steroid feedback. This provides strong support to the concept that these Kiss1 neurons are pivotal to the long-recognized "seasonal switch in the ability of E2 to exert negative feedback", which drives seasonal breeding.
Collapse
Affiliation(s)
- Didier Lomet
- Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Xavier Druart
- Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - David Hazlerigg
- Department of Arctic and Marine Biology, University of Tromsø, 9037, Tromsø, Norway
| | - Massimiliano Beltramo
- Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Hugues Dardente
- Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France.
| |
Collapse
|
20
|
Andreatta G, Tessmar-Raible K. The Still Dark Side of the Moon: Molecular Mechanisms of Lunar-Controlled Rhythms and Clocks. J Mol Biol 2020; 432:3525-3546. [PMID: 32198116 PMCID: PMC7322537 DOI: 10.1016/j.jmb.2020.03.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/18/2020] [Accepted: 03/09/2020] [Indexed: 12/22/2022]
Abstract
Starting with the beginning of the last century, a multitude of scientific studies has documented that the lunar cycle times behaviors and physiology in many organisms. It is plausible that even the first life forms adapted to the different rhythms controlled by the moon. Consistently, many marine species exhibit lunar rhythms, and also the number of documented "lunar-rhythmic" terrestrial species is increasing. Organisms follow diverse lunar geophysical/astronomical rhythms, which differ significantly in terms of period length: from hours (circalunidian and circatidal rhythms) to days (circasemilunar and circalunar cycles). Evidence for internal circatital and circalunar oscillators exists for a range of species based on past behavioral studies, but those species with well-documented behaviorally free-running lunar rhythms are not typically used for molecular studies. Thus, the underlying molecular mechanisms are largely obscure: the dark side of the moon. Here we review findings that start to connect molecular pathways with moon-controlled physiology and behaviors. The present data indicate connections between metabolic/endocrine pathways and moon-controlled rhythms, as well as interactions between circadian and circatidal/circalunar rhythms. Moreover, recent high-throughput analyses provide useful leads toward pathways, as well as molecular markers. However, for each interpretation, it is important to carefully consider the, partly substantially differing, conditions used in each experimental paradigm. In the future, it will be important to use lab experiments to delineate the specific mechanisms of the different solar- and lunar-controlled rhythms, but to also start integrating them together, as life has evolved equally long under rhythms of both sun and moon.
Collapse
Affiliation(s)
- Gabriele Andreatta
- Max Perutz Labs, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9/4, A-1030 Vienna, Austria; Research Platform "Rhythms of Life", University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9/4, A-1030 Vienna, Austria
| | - Kristin Tessmar-Raible
- Max Perutz Labs, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9/4, A-1030 Vienna, Austria; Research Platform "Rhythms of Life", University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9/4, A-1030 Vienna, Austria.
| |
Collapse
|
21
|
Corazonin signaling integrates energy homeostasis and lunar phase to regulate aspects of growth and sexual maturation in Platynereis. Proc Natl Acad Sci U S A 2019; 117:1097-1106. [PMID: 31843923 PMCID: PMC6969523 DOI: 10.1073/pnas.1910262116] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gonadotropin Releasing Hormone (GnRH) acts as a key regulator of sexual maturation in vertebrates, and is required for the integration of environmental stimuli to orchestrate breeding cycles. Whether this integrative function is conserved across phyla remains unclear. We characterized GnRH-type signaling systems in the marine worm Platynereis dumerilii, in which both metabolic state and lunar cycle regulate reproduction. We find gnrh-like (gnrhl) genes upregulated in sexually mature animals, after feeding, and in specific lunar phases. Animals in which the corazonin1/gnrhl1 gene has been disabled exhibit delays in growth, regeneration, and maturation. Molecular analyses reveal glycoprotein turnover/energy homeostasis as targets of CRZ1/GnRHL1. These findings point at an ancestral role of GnRH superfamily signaling in coordinating energy demands dictated by environmental and developmental cues. The molecular mechanisms by which animals integrate external stimuli with internal energy balance to regulate major developmental and reproductive events still remain enigmatic. We investigated this aspect in the marine bristleworm, Platynereis dumerilii, a species where sexual maturation is tightly regulated by both metabolic state and lunar cycle. Our specific focus was on ligands and receptors of the gonadotropin-releasing hormone (GnRH) superfamily. Members of this superfamily are key in triggering sexual maturation in vertebrates but also regulate reproductive processes and energy homeostasis in invertebrates. Here we show that 3 of the 4 gnrh-like (gnrhl) preprohormone genes are expressed in specific and distinct neuronal clusters in the Platynereis brain. Moreover, ligand–receptor interaction analyses reveal a single Platynereis corazonin receptor (CrzR) to be activated by CRZ1/GnRHL1, CRZ2/GnRHL2, and GnRHL3 (previously classified as AKH1), whereas 2 AKH-type hormone receptors (GnRHR1/AKHR1 and GnRHR2/AKHR2) respond only to a single ligand (GnRH2/GnRHL4). Crz1/gnrhl1 exhibits a particularly strong up-regulation in sexually mature animals, after feeding, and in specific lunar phases. Homozygous crz1/gnrhl1 knockout animals exhibit a significant delay in maturation, reduced growth, and attenuated regeneration. Through a combination of proteomics and gene expression analysis, we identify enzymes involved in carbohydrate metabolism as transcriptional targets of CRZ1/GnRHL1 signaling. Our data suggest that Platynereis CRZ1/GnRHL1 coordinates glycoprotein turnover and energy homeostasis with growth and sexual maturation, integrating both metabolic and developmental demands with the worm’s monthly cycle.
Collapse
|
22
|
Li Z, Guo R, Gu Z, Wang X, Wang Y, Xu H, Wang C, Liu X. Identification of a promoter element mediating kisspeptin-induced increases in GnRH gene expression in sheep. Gene 2019; 699:1-7. [PMID: 30853631 DOI: 10.1016/j.gene.2019.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 01/04/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) plays an important role in regulating the activities of other components downstream of the hypothalamic-pituitary-gonadal (HPG) axis and maintaining the normal reproductive cycle of animals. However, the molecular mechanisms by which GnRH synthesis and secretion are regulated in sheep remains unclear. In this study, a series of eight recombinant vectors with deletion fragments were constructed and cotransfected with pGL3-Basic and pRL-SV40 into sheep hypothalamic neuronal cells. After treatment with 1 nM kisspeptin, the core promoter of the sheep GnRH gene was identified to be in the region of -1912 bp to -1461 bp by dual-luciferase reporter assay. Bioinformatics analysis showed that there was a binding site for the transcription factor Otx-2 in the core promoter region (-1786 to -1770 bp) that was highly conserved among different species. The expression patterns of Kiss-1, Otx-2 and GnRH in the sheep hypothalamus were the same, and the expression of Kiss-1, Otx-2 and GnRH was significantly higher in the breeding season than in nonbreeding season (P < 0.01). In addition, when hypothalamic neurons were cultured in vitro with kisspeptin, kisspeptin induced the expression of GnRH and Otx-2. In conclusion, these results provide evidence that the core promoter region (-1786 to -1770 bp) of the GnRH gene is involved in the regulation of hypothalamic activity by kisspeptin and that binding of the transcription factor Otx-2 mediates this activation.
Collapse
Affiliation(s)
- Zhuanjian Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Ruoting Guo
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, China; Center for Animal Disease Control and Prevention, Changji 831100, Xinjiang, China
| | - Zhenzhen Gu
- Key Laboratory of Genetics, Breeding & Reproduction of Grass-Feeding Livestock, Ministry of Agriculture, Xinjiang Academy of Animal Science, Urumqi 830026, Xinjiang, China
| | - Xiangnan Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Yongcai Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Huifen Xu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Chunxiu Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Xiaojun Liu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China; College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, China.
| |
Collapse
|
23
|
Ahmad Pampori Z, Ahmad Sheikh A, Aarif O, Hasin D, Ahmad Bhat I. Physiology of reproductive seasonality in sheep – an update. BIOL RHYTHM RES 2018. [DOI: 10.1080/09291016.2018.1548112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Zahoor Ahmad Pampori
- Division of Veterinary Physiology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, India
| | - Aasif Ahmad Sheikh
- Division of Veterinary Physiology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, India
| | - Ovais Aarif
- Division of Veterinary Physiology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, India
| | - Dilruba Hasin
- Division of Veterinary Physiology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, India
| | - Irfan Ahmad Bhat
- Division of Veterinary Physiology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, India
| |
Collapse
|
24
|
Muñoz AL, Chesneau D, Hernández H, Bedos M, Duarte G, Vielma J, Zarazaga LA, Chemineau P, Keller M, Delgadillo JA. Sexually active bucks counterbalance the seasonal negative feedback of estradiol on LH in ovariectomized goats. Domest Anim Endocrinol 2017; 60:42-49. [PMID: 28437622 DOI: 10.1016/j.domaniend.2017.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/21/2017] [Accepted: 03/21/2017] [Indexed: 11/28/2022]
Abstract
We showed previously that the permanent presence of bucks rendered sexually active by photoperiodic treatments, thereafter called photostimulated bucks, prevents the occurrence of seasonal anovulation; also, the introduction of these sexually active bucks induces ovulations during seasonal anestrus. Here, we studied the response of ovariectomized goats bearing 12-mm subcutaneous implants filled or not with estradiol to sexually active males to determine (1) whether the permanent presence of such bucks prevents the decrease of LH despite the presence of a negative feedback by estradiol mimicking that of seasonal anestrus (experiment 1) and (2) whether the introduction of photostimulated bucks increases the plasma LH concentrations in spite of this negative feedback (experiment 2). In experiment 1, one group of goats remained in contact with sexually active bucks, whereas the other group remained in contact with control bucks. Plasma LH concentrations were high and did not differ with time or between groups of females from November to February (P > 0.05), when both types of bucks were sexually active. Afterward, in goats in contact with control and sexually inactive bucks, LH concentrations decreased from March (P ≤ 0.01) and remained low until May, whereas LH levels remained high from March to May in goats in contact with the photostimulated bucks (P > 0.05). In experiment 2, 2 groups of females bearing empty subcutaneous implants, and 2 groups of goats bearing subcutaneous implants filled with estradiol, were exposed to control or photostimulated bucks. Plasma LH concentrations did not increase in goats bearing empty implants, when exposed to control or photostimulated bucks (from 2.01 ± 0.26 to 1.98 ± 0.31 ng/mL, and from 2.45 ± 0.29 to 2.42 ± 0.21 ng/mL respectively; P > 0.05). In contrast, plasma LH concentrations increased from 0.97 ± 0.41 to 2.80 ± 0.62 ng/mL in goats exposed to the photostimulated bucks and bearing estradiol implants (P < 0.05). Thus, the permanent presence of sexually active bucks prevented the decrease of plasma LH concentration in OVX + E2 goats during the seasonal anestrus, and the introduction of the photostimulated bucks increased the plasma LH concentrations in OVX + E2 goats during the seasonal anestrus. Therefore, we conclude that in both cases, the photostimulated bucks are able to reduce or counterbalance the seasonal negative feedback of estradiol on LH secretion.
Collapse
Affiliation(s)
- A L Muñoz
- Centro de Investigación en Reproducción Caprina, Universidad Autónoma Agraria Antonio Narro, Periférico Raúl López Sánchez y Carretera a Santa Fe, C.P. 27054 Torreón, Coahuila, Mexico
| | - D Chesneau
- PRC, INRA, CNRS, Université de Tours, IFCE, Agreenium, Nouzilly, France
| | - H Hernández
- Centro de Investigación en Reproducción Caprina, Universidad Autónoma Agraria Antonio Narro, Periférico Raúl López Sánchez y Carretera a Santa Fe, C.P. 27054 Torreón, Coahuila, Mexico
| | - M Bedos
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - G Duarte
- Centro de Investigación en Reproducción Caprina, Universidad Autónoma Agraria Antonio Narro, Periférico Raúl López Sánchez y Carretera a Santa Fe, C.P. 27054 Torreón, Coahuila, Mexico
| | - J Vielma
- Centro de Investigación en Reproducción Caprina, Universidad Autónoma Agraria Antonio Narro, Periférico Raúl López Sánchez y Carretera a Santa Fe, C.P. 27054 Torreón, Coahuila, Mexico
| | - L A Zarazaga
- Departamento de Ciencias Agroforestales, Universidad de Huelva, Carretera de Palos de la Frontera s/n, 21819 Palos de la Frontera, Huelva, Spain
| | - P Chemineau
- PRC, INRA, CNRS, Université de Tours, IFCE, Agreenium, Nouzilly, France
| | - M Keller
- PRC, INRA, CNRS, Université de Tours, IFCE, Agreenium, Nouzilly, France
| | - J A Delgadillo
- Centro de Investigación en Reproducción Caprina, Universidad Autónoma Agraria Antonio Narro, Periférico Raúl López Sánchez y Carretera a Santa Fe, C.P. 27054 Torreón, Coahuila, Mexico.
| |
Collapse
|
25
|
Zhu Z, Miao Z, Chen H, Xin Q, Li L, Lin R, Huang Q, Zheng N. Ovarian transcriptomic analysis of Shan Ma ducks at peak and late stages of egg production. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2016; 30:1215-1224. [PMID: 28111447 PMCID: PMC5582276 DOI: 10.5713/ajas.16.0470] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/06/2016] [Accepted: 12/17/2016] [Indexed: 02/08/2023]
Abstract
Objective To assess the differences in ovarian transcriptomes in Shan Ma ducks between their peak and late stages of egg production, and to obtain new transcriptomic data of these egg-producing ducks. Methods The Illumina HiSeq 2000 system was used for high throughput sequencing of ovarian transcriptomes from Shan Ma ducks at their peak or late stages of egg production. Results Greater than 93% of the sequencing data had a base quality score (Q score) that was not less than 20 (Q20). From ducks at their peak stage of egg production, 42,782,676 reads were obtained, with 4,307,499,083 bp sequenced. From ducks at their late stage of egg production, 45,316,166 reads were obtained, with 4,562,063,363 bp sequenced. A comparison of the two datasets identified 2,002 differentially expressed genes, with 790 upregulated and 1,212 downregulated. Further analysis showed that 1,645 of the 2,002 differentially expressed genes were annotated in the non-redundant (NR) database, with 646 upregulated and 999 downregulated. Among the differentially expressed genes with annotations in the NR database, 696 genes were functionally annotated in the clusters of orthologous groups of proteins database, involving 25 functional categories. One thousand two hundred four of the differentially expressed genes with annotations in the NR database were functionally annotated in the gene ontology (GO) database, and could be divided into three domains and 56 categories. The three domains were cellular component, molecular function, and biological process. Among the genes identified in the GO database, 451 are involved in development and reproduction. Analysis of the differentially expressed genes with annotations in the NR database against the Kyoto encyclopedia of genes and genomes database revealed that 446 of the genes could be assigned to 175 metabolic pathways, of which the peroxisome proliferator-activated receptor signaling pathway, insulin signaling pathway, fructose and mannose metabolic pathways, gonadotropin releasing hormone signaling pathway and transforming growth factor beta signaling pathway were significantly enriched. Conclusion The differences in ovarian transcriptomes in Shan Ma ducks between their peak and late stages of egg production were elucidated, which greatly enriched the ovarian transcriptomic information of egg-producing ducks.
Collapse
Affiliation(s)
- ZhiMing Zhu
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China
| | - ZhongWei Miao
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China
| | - HongPing Chen
- Longyan Original Breeder's Farm of Shan Ma Duck, LongYan 364000, China
| | - QingWu Xin
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China
| | - Li Li
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China
| | - RuLong Lin
- Longyan Original Breeder's Farm of Shan Ma Duck, LongYan 364000, China
| | - QinLou Huang
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China
| | - NenZhu Zheng
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China
| |
Collapse
|
26
|
Yeo S, Kyle V, Morris PG, Jackman S, Sinnett‐Smith LC, Schacker M, Chen C, Colledge WH. Visualisation of Kiss1 Neurone Distribution Using a Kiss1-CRE Transgenic Mouse. J Neuroendocrinol 2016; 28:10.1111/jne.12435. [PMID: 27663274 PMCID: PMC5091624 DOI: 10.1111/jne.12435] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 08/25/2016] [Accepted: 09/20/2016] [Indexed: 12/11/2022]
Abstract
Kisspeptin neuropeptides are encoded by the Kiss1 gene and play a critical role in the regulation of the mammalian reproductive axis. Kiss1 neurones are found in two locations in the rodent hypothalamus: one in the arcuate nucleus (ARC) and another in the RP3V region, which includes the anteroventral periventricular nucleus (AVPV). Detailed mapping of the fibre distribution of Kiss1 neurones will help with our understanding of the action of these neurones in other regions of the brain. We have generated a transgenic mouse in which the Kiss1 coding region is disrupted by a CRE-GFP transgene so that expression of the CRE recombinase protein is driven from the Kiss1 promoter. As expected, mutant mice of both sexes are sterile with hypogonadotrophic hypogonadism and do not show the normal rise in luteinising hormone after gonadectomy. Mutant female mice do not develop mature Graafian follicles or form corpora lutea consistent with ovulatory failure. Mutant male mice have low blood testosterone levels and impaired spermatogenesis beyond the meiosis stage. Breeding Kiss-CRE heterozygous mice with CRE-activated tdTomato reporter mice allows fluorescence visualisation of Kiss1 neurones in brain slices. Approximately 80-90% of tdTomato positive neurones in the ARC were co-labelled with kisspeptin and expression of tdTomato in the AVPV region was sexually dimorphic, with higher expression in females than males. A small number of tdTomato-labelled neurones was also found in other locations, including the lateral septum, the anterodorsal preoptic nucleus, the amygdala, the dorsomedial and ventromedial hypothalamic nuclei, the periaquaductal grey, and the mammillary nucleus. Three dimensional visualisation of Kiss1 neurones and fibres by CLARITY processing of whole brains showed an increase in ARC expression during puberty and higher numbers of Kiss1 neurones in the caudal region of the ARC compared to the rostral region. ARC Kiss1 neurones sent fibre projections to several hypothalamic regions, including rostrally to the periventricular and pre-optic areas and to the lateral hypothalamus.
Collapse
Affiliation(s)
- S.‐H. Yeo
- Reproductive Physiology GroupDepartment of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - V. Kyle
- Reproductive Physiology GroupDepartment of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - P. G. Morris
- Reproductive Physiology GroupDepartment of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - S. Jackman
- Reproductive Physiology GroupDepartment of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - L. C. Sinnett‐Smith
- Reproductive Physiology GroupDepartment of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - M. Schacker
- Reproductive Physiology GroupDepartment of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - C. Chen
- School of Biomedical SciencesUniversity of QueenslandSt LuciaAustralia
| | - W. H. Colledge
- Reproductive Physiology GroupDepartment of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| |
Collapse
|
27
|
Clarke SA, Dhillo WS. Kisspeptin across the human lifespan:evidence from animal studies and beyond. J Endocrinol 2016; 229:R83-98. [PMID: 27340201 DOI: 10.1530/joe-15-0538] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 03/10/2016] [Indexed: 11/08/2022]
Abstract
Since its first description in 1996, the KISS1 gene and its peptide products, kisspeptins, have increasingly become recognised as key regulators of reproductive health. With kisspeptins acting as ligands for the kisspeptin receptor KISS1R (previously known as GPR54 or KPR54), recent work has consistently shown that administration of kisspeptin across a variety of species stimulates gonadotrophin release through influencing gonadotrophin-releasing hormone secretion. Evidence from both animal and human studies supports the finding that kisspeptins are crucial for ensuring healthy development, with knockout animal models, as well as proband genetic testing in human patients affected by abnormal pubertal development, corroborating the notion that a functional kisspeptin receptor is required for appropriate gonadotrophin secretion. Given the large body of evidence that exists surrounding the influence of kisspeptin in a variety of settings, this review summarises our physiological understanding of the role of these important peptides and their receptors, before proceeding to describe the varying role they play across the reproductive lifespan.
Collapse
Affiliation(s)
- Sophie A Clarke
- Department of Investigative MedicineImperial College London, Hammersmith Hospital, London, UK
| | - Waljit S Dhillo
- Department of Investigative MedicineImperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
28
|
Clarke H, Dhillo WS, Jayasena CN. Comprehensive Review on Kisspeptin and Its Role in Reproductive Disorders. Endocrinol Metab (Seoul) 2015; 30:124-41. [PMID: 26194072 PMCID: PMC4508256 DOI: 10.3803/enm.2015.30.2.124] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/05/2015] [Accepted: 01/12/2015] [Indexed: 02/05/2023] Open
Abstract
Kisspeptin has recently emerged as a key regulator of the mammalian reproductive axis. It is known that kisspeptin, acting centrally via the kisspeptin receptor, stimulates secretion of gonadotrophin releasing hormone (GnRH). Loss of kisspeptin signaling causes hypogonadotrophic hypogonadism in humans and other mammals. Kisspeptin interacts with other neuropeptides such as neurokinin B and dynorphin, to regulate GnRH pulse generation. In addition, a growing body of evidence suggests that kisspeptin signaling be regulated by nutritional status and stress. Kisspeptin may also represent a novel potential therapeutic target in the treatment of fertility disorders. Early human studies suggest that peripheral exogenous kisspeptin administration stimulates gonadotrophin release in healthy adults and in patients with certain forms of infertility. This review aims to concisely summarize what is known about kisspeptin as a regulator of reproductive function, and provide an update on recent advances within this field.
Collapse
Affiliation(s)
- Holly Clarke
- Department of Investigative Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Waljit S Dhillo
- Department of Investigative Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Channa N Jayasena
- Department of Investigative Medicine, Hammersmith Hospital, Imperial College London, London, UK.
| |
Collapse
|
29
|
Kriegsfeld LJ, Ubuka T, Bentley GE, Tsutsui K. Seasonal control of gonadotropin-inhibitory hormone (GnIH) in birds and mammals. Front Neuroendocrinol 2015; 37:65-75. [PMID: 25511257 PMCID: PMC4405439 DOI: 10.1016/j.yfrne.2014.12.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/05/2014] [Accepted: 12/07/2014] [Indexed: 12/14/2022]
Abstract
Animals inhabiting temperate and boreal latitudes experience marked seasonal changes in the quality of their environments and maximize reproductive success by phasing breeding activities with the most favorable time of year. Whereas the specific mechanisms driving seasonal changes in reproductive function vary across species, converging lines of evidence suggest gonadotropin-inhibitory hormone (GnIH) serves as a key component of the neuroendocrine circuitry driving seasonal changes in reproduction and sexual motivation in some species. In addition to anticipating environmental change through transduction of photoperiodic information and modifying reproductive state accordingly, GnIH is also positioned to regulate acute changes in reproductive status should unpredictable conditions manifest throughout the year. The present overview summarizes the role of GnIH in avian and mammalian seasonal breeding while considering the similarities and disparities that have emerged from broad investigations across reproductively photoperiodic species.
Collapse
Affiliation(s)
- Lance J Kriegsfeld
- Department of Psychology and Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, CA 94720-1650, USA.
| | - Takayoshi Ubuka
- Laboratory of Integrative Brain Sciences, Department of Biology, Waseda University, Center for Medical Life Science of Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - George E Bentley
- Department of Integrative Biology and Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, CA 94720-3140, USA
| | - Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences, Department of Biology, Waseda University, Center for Medical Life Science of Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| |
Collapse
|
30
|
Tanaka T, Ohkura S, Wakabayashi Y, Kuroiwa T, Nagai K, Endo N, Tanaka A, Matsui H, Kusaka M, Okamura H. Differential effects of continuous exposure to the investigational metastin/kisspeptin analog TAK-683 on pulsatile and surge mode secretion of luteinizing hormone in ovariectomized goats. J Reprod Dev 2013; 59:563-8. [PMID: 24047956 PMCID: PMC3934154 DOI: 10.1262/jrd.2013-060] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The aim of the present study was to determine if the estradiol-induced luteinizing
hormone (LH) surge is influenced by the constant exposure to TAK-683, an
investigational metastin/kisspeptin analog, that had been established to depress the
pulsatile gonadotropin-releasing hormone (GnRH) and LH secretion in goats.
Ovariectomized goats subcutaneously received TAK-683 (TAK-683 group, n=6) or vehicle
(control group, n=6) constantly via subcutaneous implantation of an osmotic pump.
Five days after the start of the treatment, estradiol was infused intravenously in
both groups to evaluate the effects on the LH surge. Blood samples were collected at
6-min intervals for 4 h prior to the initiation of either the TAK-683 treatment or
the estradiol infusion, to determine the profiles of pulsatile LH secretion. They
were also collected at 2-h intervals from –4 h to 32 h after the start of estradiol
infusion for analysis of LH surges. The frequency and mean concentrations of LH
pulses in the TAK-683 group were remarkably suppressed 5 days after the start of
TAK-683 treatment compared with those of the control group (P<0.05). On the other
hand, a clear LH surge was observed in all animals of both groups. There were no
significant differences in the LH concentrations for surge peak and the peak time of
the LH surge between the TAK-683 and control groups. These findings suggest that the
effects of continuous exposure to kisspeptin or its analog on the mechanism(s) that
regulates the pulsatile and surge mode secretion of GnRH/LH are different in
goats.
Collapse
Affiliation(s)
- Tomomi Tanaka
- Laboratory of Veterinary Reproduction, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Chai K, Liu X, Zhang Y, Lin H. Day-night and reproductive cycle profiles of melatonin receptor, kiss
, and gnrh
expression in orange-spotted grouper (Epinephelus coioides
). Mol Reprod Dev 2013; 80:535-48. [DOI: 10.1002/mrd.22191] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Accepted: 05/02/2013] [Indexed: 12/15/2022]
Affiliation(s)
- Ke Chai
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals; School of Life Sciences, Sun Yat-Sen University; Guangzhou China
- Material and Chemical Engineering College, Hainan University; Haikou China
| | - Xiaochun Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals; School of Life Sciences, Sun Yat-Sen University; Guangzhou China
| | - Yong Zhang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals; School of Life Sciences, Sun Yat-Sen University; Guangzhou China
| | - Haoran Lin
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals; School of Life Sciences, Sun Yat-Sen University; Guangzhou China
- College of Ocean, Hainan University; Haikou China
| |
Collapse
|
32
|
Klosen P, Sébert M, Rasri K, Laran‐Chich M, Simonneaux V. TSH restores a summer phenotype in photoinhibited mammals
via
the RF‐amides RFRP3 and kisspeptin. FASEB J 2013; 27:2677-86. [DOI: 10.1096/fj.13-229559] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Paul Klosen
- Département de Neurobiologie des RythmesInstitut des Neurosciences Cellulaires et IntégrativesCentre National de la Recherche Scientifique (CNRS) Unité Propres de Recherche (UPR) 3212Université de StrasbourgStrasbourgFrance
| | - Marie‐Emilie Sébert
- Département de Neurobiologie des RythmesInstitut des Neurosciences Cellulaires et IntégrativesCentre National de la Recherche Scientifique (CNRS) Unité Propres de Recherche (UPR) 3212Université de StrasbourgStrasbourgFrance
| | - Kamontip Rasri
- Department of Pre‐Clinical ScienceFaculty of MedicineThammasart UniversityRangsitThailand
| | - Marie‐Pierre Laran‐Chich
- Département de Neurobiologie des RythmesInstitut des Neurosciences Cellulaires et IntégrativesCentre National de la Recherche Scientifique (CNRS) Unité Propres de Recherche (UPR) 3212Université de StrasbourgStrasbourgFrance
| | - Valérie Simonneaux
- Département de Neurobiologie des RythmesInstitut des Neurosciences Cellulaires et IntégrativesCentre National de la Recherche Scientifique (CNRS) Unité Propres de Recherche (UPR) 3212Université de StrasbourgStrasbourgFrance
| |
Collapse
|
33
|
Meza-Herrera CA, Vargas-Beltran F, Tena-Sempere M, González-Bulnes A, Macias-Cruz U, Veliz-Deras FG. Short-term beta-carotene-supplementation positively affects ovarian activity and serum insulin concentrations in a goat model. J Endocrinol Invest 2013; 36:185-9. [PMID: 22572738 DOI: 10.3275/8410] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In early October 2010, adult goats (no.=22, 3.5 yr old, 7/8 Sannen-Alpine, 26° N, 103° W, at 1117 m), were randomly assigned to: i) beta-carotene group (BC) [no.=10; live weight (LW)=45.9±1.97 kg, body condition score (BCS) =3.04±0.08; orally supplemented with 50 mg of BC per goat per day]; ii) control group (CONT) (no.=12; LW=46.2±2.04 kg, BCS=3.0±0.08). Animals received a basal diet of alfalfa hay, corn silage, and corn grain, having free access to water, shade, and mineral salts. During the second half of October, estrus was synchronized by using intravaginal sponges. Thereafter, by mid-follicular phase, an intensive blood sampling (6 h × 60 min) was performed to evaluate serum insulin concentrations (INS) by radioimmunoassay. By the end of the luteal phase, an ultrasonographic scanning was performed to evaluate total ovarian activity (TOA) [TOA=total follicles (TF) + total corpus luteum (TCL)]. The whole experimental period consisted of 34 days pre- and 17 days post-ovulation, for a total of 52 days. Average LW and BCS did not differ (p>0.05) during the experimental period. Nonetheless, increases in TF no. (5.0 vs 3.4±0.6 units; p=0.05), TCL no. (3.4 vs 2.8±0.2 units; p=0.05), TOA (8.1 vs 6.2±0.6 units; p=0.05) and INS (4.6 vs 3.9±0.4 ng ml-1; p=0.05) favored to the BC-supplemented group. A positive correlation between LW (r(2)=0.42; p=0.04) and BCS (r(2)=0.47; p=0.02) with respect to ovulation rate, was detected. BC-supplementation increased ovarian activity in the female goat while positively affected the release pattern of insulin, suggesting a potential role of BC as a central and/or pancreas-activating molecule in adult goats; such results may hold not only physiologic but also clinical significance.
Collapse
Affiliation(s)
- C A Meza-Herrera
- Unidad Regional Universitaria de Zonas Áridas, Universidad Autónoma Chapingo, Bermejillo, Mexico.
| | | | | | | | | | | |
Collapse
|
34
|
De Bond JAP, Li Q, Millar RP, Clarke IJ, Smith JT. Kisspeptin signaling is required for the luteinizing hormone response in anestrous ewes following the introduction of males. PLoS One 2013; 8:e57972. [PMID: 23469121 PMCID: PMC3585258 DOI: 10.1371/journal.pone.0057972] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 01/29/2013] [Indexed: 11/18/2022] Open
Abstract
The introduction of a novel male stimulates the hypothalamic-pituitary-gonadal axis of female sheep during seasonal anestrus, leading to the resumption of follicle maturation and ovulation. How this pheromone cue activates pulsatile secretion of gonadotropin releasing hormone (GnRH)/luteinizing hormone (LH) is unknown. We hypothesised that pheromones activate kisspeptin neurons, the product of which is critical for the stimulation of GnRH neurons and fertility. During the non-breeding season, female sheep were exposed to novel males and blood samples collected for analysis of plasma LH profiles. Females without exposure to males served as controls. In addition, one hour before male exposure, a kisspeptin antagonist (P-271) or vehicle was infused into the lateral ventricle and continued for the entire period of male exposure. Introduction of a male led to elevated mean LH levels, due to increased LH pulse amplitude and pulse frequency in females, when compared to females not exposed to a male. Infusion of P-271 abolished this effect of male exposure. Brains were collected after the male effect stimulus and we observed an increase in the percentage of kisspeptin neurons co-expressing Fos, by immunohistochemistry. In addition, the per-cell expression of Kiss1 mRNA was increased in the rostral and mid (but not the caudal) arcuate nucleus (ARC) after male exposure in both aCSF and P-271 treated ewes, but the per-cell content of neurokinin B mRNA was decreased. There was also a generalized increase in Fos positive cells in the rostral and mid ARC as well as the ventromedial hypothalamus of females exposed to males. We conclude that introduction of male sheep to seasonally anestrous female sheep activates kisspeptin neurons and other cells in the hypothalamus, leading to increased GnRH/LH secretion.
Collapse
Affiliation(s)
| | - Qun Li
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Robert P. Millar
- Mammal Research Institute, University of Pretoria, Pretoria, Gauteng, South Africa
- UCT/MRC Receptor Biology Unit, University of Cape Town, Cape Town, Western Cape, South Africa
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Scotland
| | - Iain J. Clarke
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Jeremy T. Smith
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
35
|
Nocillado JN, Mechaly AS, Elizur A. In silico analysis of the regulatory region of the Yellowtail Kingfish and Zebrafish Kiss and Kiss receptor genes. FISH PHYSIOLOGY AND BIOCHEMISTRY 2013; 39:59-63. [PMID: 22527613 DOI: 10.1007/s10695-012-9642-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 03/30/2012] [Indexed: 05/31/2023]
Abstract
We have cloned and analysed the partial putative promoter sequences of the Yellowtail Kingfish (Seriola lalandi) Kiss2 and Kiss2r genes (380 and 420 bp, respectively). We obtained in silico 1.5 kb of the zebrafish (Danio rerio) Kiss1, Kiss2, Kiss1r and zfKiss2r sequences upstream of the putative transcriptional initiation site. Bioinformatic analysis revealed promoter regulatory elements including AP-1, Sp1, GR, ER, PR, AR, GATA-1, TTF-1, YY1 and C/EBP. These regulatory elements may mediate novel roles of the Kiss genes and their receptors in addition to their established role in reproductive function.
Collapse
Affiliation(s)
- J N Nocillado
- School of Science, Education and Engineering, University of the Sunshine Coast, 90 Sippy Downs Drive, Sippy Downs, QLD, 4556, Australia
| | | | | |
Collapse
|
36
|
Rønnekleiv OK, Kelly MJ. Kisspeptin excitation of GnRH neurons. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 784:113-31. [PMID: 23550004 PMCID: PMC4019505 DOI: 10.1007/978-1-4614-6199-9_6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Kisspeptin binding to its cognate G protein-coupled receptor (GPR54, aka Kiss1R) in gonadotropin-releasing hormone (GnRH) neurons stimulates peptide release and activation of the reproductive axis in mammals. Kisspeptin has pronounced pre- and postsynaptic effects, with the latter dominating the excitability of GnRH neurons. Presynaptically, kisspeptin increases the excitatory drive (both GABA-A and glutamate) to GnRH neurons and postsynaptically, kisspeptin inhibits an A-type and inwardly rectifying K(+) (Kir 6.2 and GIRK) currents and activates nonselective cation (TRPC) currents to cause long-lasting depolarization and increased action potential firing. The signaling cascades and the multiple intracellular targets of kisspeptin actions in native GnRH neurons are continuing to be elucidated. This review summarizes our current state of knowledge about kisspeptin signaling in GnRH neurons.
Collapse
Affiliation(s)
- Oline K Rønnekleiv
- Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA.
| | | |
Collapse
|
37
|
Goodman RL, Maltby MJ, Millar RP, Hileman SM, Nestor CC, Whited B, Tseng AS, Coolen LM, Lehman MN. Evidence that dopamine acts via kisspeptin to hold GnRH pulse frequency in check in anestrous ewes. Endocrinology 2012; 153:5918-27. [PMID: 23038740 PMCID: PMC3512065 DOI: 10.1210/en.2012-1611] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent work has implicated stimulatory kisspeptin neurons in the arcuate nucleus (ARC) as important for seasonal changes in reproductive function in sheep, but earlier studies support a role for inhibitory A15 dopaminergic (DA) neurons in the suppression of GnRH (and LH) pulse frequency in the nonbreeding (anestrous) season. Because A15 neurons project to the ARC, we performed three experiments to test the hypothesis that A15 neurons act via ARC kisspeptin neurons to inhibit LH in anestrus: 1) we used dual immunocytochemistry to determine whether these ARC neurons contain D2 dopamine receptor (D2-R), the receptor responsible for inhibition of LH in anestrus; 2) we tested the ability of local administration of sulpiride, a D2-R antagonist, into the ARC to increase LH secretion in anestrus; and 3) we determined whether an antagonist to the kisspeptin receptor could block the increase in LH secretion induced by sulpiride in anestrus. In experiment 1, 40% of this ARC neuronal subpopulation contained D2-R in breeding season ewes, but this increased to approximately 80% in anestrus. In experiment 2, local microinjection of the two highest doses (10 and 50 nmol) of sulpiride into the ARC significantly increased LH pulse frequency to levels 3 times that seen with vehicle injections. Finally, intracerebroventricular infusion of a kisspeptin receptor antagonist completely blocked the increase in LH pulse frequency induced by systemic administration of sulpiride to anestrous ewes. These results support the hypothesis that DA acts to inhibit GnRH (and LH) secretion in anestrus by suppressing the activity of ARC kisspeptin neurons.
Collapse
Affiliation(s)
- Robert L Goodman
- Department of Physiology and Pharmacology, Robert C. Byrd Health Sciences Center, Morgantown, WV 26506, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Bosch MA, Xue C, Rønnekleiv OK. Kisspeptin expression in guinea pig hypothalamus: effects of 17β-estradiol. J Comp Neurol 2012; 520:2143-62. [PMID: 22173890 DOI: 10.1002/cne.23032] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Kisspeptin is essential for reproductive functions in humans. As a model for the human we have used the female guinea pig, which has a long ovulatory cycle similar to that of primates. Initially, we cloned a guinea pig kisspeptin cDNA sequence and subsequently explored the distribution and 17β-estradiol (E2) regulation of kisspeptin mRNA (Kiss1) and protein (kisspeptin) by using in situ hybridization, real-time PCR and immunocytochemistry. In ovariectomized females, Kiss1 neurons were scattered throughout the preoptic periventricular areas (PV), but the vast majority of Kiss1 neurons were localized in the arcuate nucleus (Arc). An E2 treatment that first inhibits (negative feedback) and then augments (positive feedback) serum luteinizing hormone (LH) increased Kiss1 mRNA density and number of cells expressing Kiss1 in the PV at both time points. Within the Arc, Kiss1 mRNA density was reduced at both time points. Quantitative real-time PCR confirmed the in situ hybridization results during positive feedback. E2 reduced the number of immunoreactive kisspeptin cells in the PV at both time points, perhaps an indication of increased release. Within the Arc, the kisspeptin immunoreactivity was decreased during negative feedback but increased during positive feedback. Therefore, it appears that in guinea pig both the PV and the Arc kisspeptin neurons act cooperatively to excite gonadotropin-releasing hormone (GnRH) neurons during positive feedback. We conclude that E2 regulation of negative and positive feedback may reflect a complex interaction of the kisspeptin circuitry, and both the PV and the Arc respond to hormone signals to encode excitation of GnRH neurons during the ovulatory cycle.
Collapse
Affiliation(s)
- Martha A Bosch
- Department of Physiology/Pharmacology, Oregon Health and Science University, Portland, Oregon 97239, USA
| | | | | |
Collapse
|
39
|
Williams GL, Thorson JF, Prezotto LD, Velez IC, Cardoso RC, Amstalden M. Reproductive seasonality in the mare: neuroendocrine basis and pharmacologic control. Domest Anim Endocrinol 2012; 43:103-15. [PMID: 22579068 DOI: 10.1016/j.domaniend.2012.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 04/11/2012] [Accepted: 04/11/2012] [Indexed: 11/23/2022]
Abstract
Reproductive seasonality in the mare is characterized by a marked decline in adenohypophyseal synthesis and secretion of LH beginning near the autumnal equinox. Thus, ovarian cycles have ceased in most mares by the time of the winter solstice. Endogenous reproductive rhythms in seasonal species are entrained or synchronized as a result of periodic environmental cues. In the horse, this cue is primarily day length. Hence, supplemental lighting schemes have been used managerially for decades to modify the annual timing of reproduction in the mare. Although a full characterization of the cellular and molecular bases of seasonal rhythms has not been realized in any species, many of their synaptic and humoral signaling pathways have been defined. In the mare, neuroendocrine-related studies have focused primarily on the roles of GnRH and interneuronal signaling pathways that subserve the GnRH system in the regulatory cascade. Recent studies have considered the role of a newly discovered neuropeptide, RF-related peptide 3 that could function to inhibit GnRH secretion or gonadotrope responsiveness. Although results that used native peptide sequences have been negative in the mare and mixed in all mammalian females, new studies that used an RFRP3 antagonist (RF9) in sheep are encouraging. Importantly, despite continuing deficits in some fundamental areas, the knowledge required to control seasonal anovulation pharmacologically has been available for >20 yr. Specifically, the continuous infusion of native GnRH is both reliable and efficient for accelerating reproductive transition and is uniquely applicable to the horse. However, its practical exploitation continues to await the development of a commercially acceptable delivery vehicle.
Collapse
Affiliation(s)
- G L Williams
- Animal Reproduction Laboratory, Texas AgriLife Research, Beeville, TX 78102, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Stevenson TJ, Hahn TP, MacDougall-Shackleton SA, Ball GF. Gonadotropin-releasing hormone plasticity: a comparative perspective. Front Neuroendocrinol 2012; 33:287-300. [PMID: 23041619 PMCID: PMC3484179 DOI: 10.1016/j.yfrne.2012.09.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 09/04/2012] [Accepted: 09/05/2012] [Indexed: 11/16/2022]
Abstract
Gonadotropin-releasing hormone 1 (GnRH1) is a key regulator of the reproductive neuroendocrine system in vertebrates. Recent developments have suggested that GnRH1 neurons exhibit far greater plasticity at the cellular and molecular levels than previously thought. Furthermore, there is growing evidence that sub-populations of GnRH1 neurons in the preoptic area are highly responsive to specific environmental and hormonal conditions. In this paper we discuss findings that reveal large variation in GnRH1 mRNA and protein expression that are regulated by social cues, photoperiod, and hormonal feedback. We draw upon studies using histochemistry and immediate early genes (e.g., c-FOS/ZENK) to illustrate that specific groups of GnRH1 neurons are topographically organized. Based on data from diverse vertebrate species, we suggest that GnRH1 expression within individuals is temporally dynamic and this plasticity may be evolutionarily conserved. We suggest that the plasticity observed in other neuropeptide systems (i.e. kisspeptin) may have evolved in a similar manner.
Collapse
Affiliation(s)
- T J Stevenson
- Institute for Mind and Biology, University of Chicago, Chicago, IL 60637, USA.
| | | | | | | |
Collapse
|
41
|
Analysis on DNA sequence of goat RFRP gene and its possible association with average daily sunshine duration. Mol Biol Rep 2012; 39:9167-77. [PMID: 22733487 DOI: 10.1007/s11033-012-1789-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 06/09/2012] [Indexed: 10/28/2022]
Abstract
Goat RFRP gene was cloned and its mutations were detected in thirteen goat breeds whose reproductive seasonality and litter size were different. Then sequence characteristics were analyzed and association analyses were performed to reveal the relationships between mutations of RFRP gene and average daily sunshine duration, reproductive seasonality as well as litter size in goats. A 4,862 bp DNA fragment of goat RFRP gene was obtained and the complete CDS of 591 bp encodes 196 amino acids, having high homology with that of other mammals. The protein was predicted to be a secreted protein with a signal peptide of 21 amino acids. Moreover, two mutations (A712G, T1493C) in 5' regulatory region and one mutation (A3438T) in exon 2 were detected. The test of genotype distribution in six selective goat breeds showed that there was no uniform significant association between the three polymorphisms and seasonal reproduction. The association just existed in some goat breeds for each locus. Interestingly, however, there was a strong positive correlation (r = 0.830, P = 0.003) between the G allele frequency of the A712G locus and average daily sunshine duration in ten local goat breeds, suggesting that RFRP gene has undergone a selective pressure in sunshine duration and may have indirect relationship with reproductive seasonality in goats. Additionally, no significant difference was found in litter size between genotypes in prolific Jining Grey goats.
Collapse
|
42
|
[The neuroendocrine regulatory mechanisms of mammalian seasonal reproduction]. YI CHUAN = HEREDITAS 2012; 34:281-8. [PMID: 22425946 DOI: 10.3724/sp.j.1005.2012.00281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The seasonal reproduction of mammal means the reproduction experiences an annual period from quiescence to renaissance. Studies have shown that kisspeptin and RFRP play an important role in the reproductive seasonality. The non-breeding season is characterized by an increase in the negative feedback effect of estrogen on GnRH, and this effect is transmitted by kisspeptin neurons, which may be an important factor affecting the reproduction activities. The expression of RFRP depends on melatonin secretion, and shows an apparent inhibition on reproduction in non-breeding season. In addition, thyroid hormones influence termination of the breeding season. Dopaminergic neuron A14/A15 also contributes to the seasonal changes in estrogen negative feedback. These neural systems may synergistically modulate the seasonal changes of reproductive function with the photoperiod. This review makes a systematic expatiation on the relationship between seasonal reproduction and these neural systems.
Collapse
|
43
|
Dardente H. Melatonin-dependent timing of seasonal reproduction by the pars tuberalis: pivotal roles for long daylengths and thyroid hormones. J Neuroendocrinol 2012; 24:249-66. [PMID: 22070540 DOI: 10.1111/j.1365-2826.2011.02250.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Most mammals living at temperate latitudes exhibit marked seasonal variations in reproduction. In long-lived species, it is assumed that timely physiological alternations between a breeding season and a period of sexual rest depend upon the ability of day length (photoperiod) to synchronise an endogenous timing mechanism called the circannual clock. The sheep has been extensively used to characterise the time-measurement mechanisms of seasonal reproduction. Melatonin, secreted only during the night, acts as the endocrine transducer of the photoperiodic message. The present review is concerned with the endocrine mechanisms of seasonal reproduction in sheep and the evidence that long day length and thyroid hormones are mandatory to their proper timing. Recent evidence for a circadian-based molecular mechanism within the pars tuberalis of the pituitary, which ties the short duration melatonin signal reflecting long day length to the hypothalamic increase of triiodothyronine (T3) through a thyroid-stimulating hormone/deiodinase2 paracrine mechanism is presented and evaluated in this context. A parallel is also drawn with the golden hamster, a long-day breeder, aiming to demonstrate that features of seasonality appear to be phylogenetically conserved. Finally, potential mechanisms of T3 action within the hypothalamus/median eminence in relationship to seasonal timing are examined.
Collapse
Affiliation(s)
- Hugues Dardente
- Physiologie de la Reproduction et des Comportements, INRA UMR85, CNRS UMR6175, Université de Tours, Nouzilly, Haras Nationaux France.
| |
Collapse
|
44
|
Simonneaux V, Bur I, Ancel C, Ansel L, Klosen P. A kiss for daily and seasonal reproduction. PROGRESS IN BRAIN RESEARCH 2012; 199:423-437. [DOI: 10.1016/b978-0-444-59427-3.00024-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
45
|
Li Q, Rao A, Pereira A, Clarke IJ, Smith JT. Kisspeptin cells in the ovine arcuate nucleus express prolactin receptor but not melatonin receptor. J Neuroendocrinol 2011; 23:871-82. [PMID: 21793946 DOI: 10.1111/j.1365-2826.2011.02195.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Melatonin is secreted at night by the pineal gland and governs the reproductive system in seasonal breeders, such as sheep. The mechanism by which melatonin regulates reproduction is not known. The circannual rhythmicity of other factors, including prolactin, is also regulated by photoperiod via changes in melatonin secretion. In sheep, plasma prolactin levels are higher in the nonbreeding season than the breeding season. Kisspeptin, synthesised by neurones in the ovine arcuate nucleus (ARC) and preoptic area, is a key regulator of reproduction through stimulation of gonadotrophin-releasing hormone secretion and its expression in the ARC is reduced during the nonbreeding season. We hypothesised that kisspeptin expression is directly, or indirectly, regulated by melatonin and/or prolactin. We first examined the expression of melatonin receptor (MTNR1A) in kisspeptin (Kiss1 mRNA) neurones in the ARC of ovariectomised (OVX) sheep using double-label in situ hybridisation. MTNR1A mRNA was not expressed by kisspeptin neurones, whereas strong expression was detected in the pars tuberalis. We then examined the expression of the long-form prolactin receptor (PRLR-L) in ARC kisspeptin neurones. In OVX ewes, approximately 60% of kisspeptin neurones expressed PRLR-L mRNA at similar levels in the breeding and nonbreeding seasons. We then aimed to determine whether prolactin treatment during the breeding season regulates kisspeptin expression in the ARC. Continuous central infusion of prolactin (20 μg/h for 7 days) in oestradiol-treated OVX sheep did not alter Kiss1 mRNA expression or luteinising hormone secretion, although it induced substantial phosphorylated signal transducer and activator of transcription 5-immunoreactive nuclei staining in the mediobasal hypothalamus. We conclude that the seasonal change in kisspeptin neurones cannot be regulated directly by melatonin, although it may be a result of changes in prolactin levels. Despite this, kisspeptin expression was unchanged after exogenous prolactin treatment in breeding season ewes.
Collapse
Affiliation(s)
- Q Li
- Department of Physiology, Monash University, Clayton, Victoria, Australia
| | | | | | | | | |
Collapse
|
46
|
Uenoyama Y, Inoue N, Pheng V, Homma T, Takase K, Yamada S, Ajiki K, Ichikawa M, Okamura H, Maeda KI, Tsukamura H. Ultrastructural evidence of kisspeptin-gonadotrophin-releasing hormone (GnRH) interaction in the median eminence of female rats: implication of axo-axonal regulation of GnRH release. J Neuroendocrinol 2011; 23:863-70. [PMID: 21815953 DOI: 10.1111/j.1365-2826.2011.02199.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The present study was conducted to determine the morphological and functional interaction between kisspeptin and gonadotrophin-releasing hormone (GnRH) neuronal elements at the median eminence in female rats to clarify a possibility that kisspeptin directly stimulates GnRH release at the nerve end. A dual immunoelectron microscopic study of kisspeptin and GnRH showed that the kisspeptin-immunoreactive nerve element directly abutted the GnRH-immunoreactive nerve element, although no obvious synaptic structure was found between kisspeptin and GnRH neurones in the median eminence. The current retrograde tracing study with FluoroGold (FG) indicates that kisspeptin neurones are not in contact with fenestrated capillaries because no FG signal was found in kisspeptin neurones when the FG was injected peripherally. This peripheral FG injection revealed the neuroendocrine neurones projecting to the median eminence because FG-positive GnRH neuronal cell bodies were found in the preoptic area. Synthetic rat kisspeptin (1-52)-amide stimulated GnRH release from the median eminence tissues in a dose-dependent manner. Thus, the present results suggest that kisspeptin at least partly exerts stimulatory effects on GnRH release from the neuronal terminals of GnRH neurones by axo-axonal nonsynaptic interaction in the median eminence.
Collapse
Affiliation(s)
- Y Uenoyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Research progress in molecular mechanism of animal seasonal reproduction. YI CHUAN = HEREDITAS 2011; 33:695-706. [DOI: 10.3724/sp.j.1005.2011.00695] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
48
|
Stevenson TJ, Ball GF. Information theory and the neuropeptidergic regulation of seasonal reproduction in mammals and birds. Proc Biol Sci 2011; 278:2477-85. [PMID: 21208957 PMCID: PMC3125617 DOI: 10.1098/rspb.2010.2181] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 12/09/2010] [Indexed: 11/12/2022] Open
Abstract
Seasonal breeding in the temperate zone is a dramatic example of a naturally occurring change in physiology and behaviour. Cues that predict periods of environmental amelioration favourable for breeding must be processed by the brain so that the appropriate responses in reproductive physiology can be implemented. The neural integration of several environmental cues converges on discrete hypothalamic neurons in order to regulate reproductive physiology. Gonadotrophin-releasing hormone-1 (GnRH1) and Kisspeptin (Kiss1) neurons in avian and mammalian species, respectively, show marked variation in expression that is positively associated with breeding state. We applied the constancy/contingency model of predictability to investigate how GnRH1 and Kiss1 integrate different environmental cues to regulate reproduction. We show that variation in GnRH1 from a highly seasonal avian species exhibits a predictive change that is primarily based on contingency information. Opportunistic species have low measures of predictability and exhibit a greater contribution of constancy information that is sex-dependent. In hamsters, Kiss1 exhibited a predictive change in expression that was predominantly contingency information and is anatomically localized. The model applied here provides a framework for studies geared towards determining the impact of variation in climate patterns to reproductive success in vertebrate species.
Collapse
Affiliation(s)
- Tyler J Stevenson
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD 21218, USA.
| | | |
Collapse
|
49
|
Krohmer R, Lutterschmidt D. Environmental and Neuroendorcrine Control of Reproduction in Snakes. REPRODUCTIVE BIOLOGY AND PHYLOGENY OF SNAKES 2011. [DOI: 10.1201/b10879-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
50
|
Wahab F, Ullah F, Chan YM, Seminara SB, Shahab M. Decrease in hypothalamic Kiss1 and Kiss1r expression: a potential mechanism for fasting-induced suppression of the HPG axis in the adult male rhesus monkey (Macaca mulatta). Horm Metab Res 2011; 43:81-5. [PMID: 21154197 PMCID: PMC4119764 DOI: 10.1055/s-0030-1269852] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Fasting suppresses functioning of the hypothalamic-pituitary-gonadal (HPG) axis by mechanisms that are incompletely understood. In 2003, hypothalamic kisspeptin-Kiss1r signaling was discovered to play a significant role in regulating the HPG axis. We have recently shown that in adult male macaques, short-term fasting attenuates the response of the HPG axis to an exogenous kisspeptin challenge. In the present study, we explored the mechanism underlying this attenuated response by examining the modulation of the hypothalamic expression of KISS1 and KISS1R under short-term fasting and normal feeding conditions in the adult male macaques. Hypothalamic mRNA was extracted from normal fed (n=3) and 48-h fasted (n=3) monkeys. KISS1, KISS1R, and GNRH1 mRNA were quantified by reverse transcription followed by real-time polymerase chain reaction. In addition, blood samples were collected for measurement of plasma concentrations of glucose, cortisol, leptin, and testosterone. In contrast to fed animals, plasma glucose, leptin, and testosterone levels decreased and cortisol levels increased in fasted animals. The hypothalamic expression of KISS1 and KISS1R mRNA was significantly lower (p<0.05) in fasted monkeys compared to fed monkeys while hypothalamic GNRH1 mRNA expression was comparable between the 2 groups. Thus, our results demonstrate that expression of hypothalamic KISS1 and KISS1R decrease after a short-term fasting in monkeys. This decrease may contribute to the suppression of the HPG axis during fasting conditions in primates. In addition, our finding of lower expression of KISS1R in fasted monkeys provides an explanation for the attenuation in the HPG axis response to peripheral kisspeptin challenge during short-term fasting.
Collapse
Affiliation(s)
- Fazal Wahab
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
- Reproductive Endocrine Unit and Harvard Reproductive Sciences Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Farhad Ullah
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Yee-Ming Chan
- Reproductive Endocrine Unit and Harvard Reproductive Sciences Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Stephanie B. Seminara
- Reproductive Endocrine Unit and Harvard Reproductive Sciences Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Muhammad Shahab
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| |
Collapse
|