1
|
Coutant R, Zaegel N, Illouz F, Levaillant L, Magnin F, Perrot J, Ait Tayeb AEK, Briet C, Kaiser UB, Bouligand J, Young J, Maione L. Reversal of congenital hypogonadotropic hypogonadism in two brothers carrying a homozygous KISS1 frameshift mutation. Lancet Diabetes Endocrinol 2025; 13:550-552. [PMID: 40480241 DOI: 10.1016/s2213-8587(25)00121-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/07/2025] [Accepted: 04/18/2025] [Indexed: 06/22/2025]
Affiliation(s)
- Régis Coutant
- Department of Pediatric Endocrinology, Reference Center for Rare Pituitary Diseases (CRMR-HYPO), Angers University Hospital, Angers, France
| | - Nadia Zaegel
- Paris-Saclay University, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Department of Endocrinology and Reproductive Diseases, Reference Center for Rare Pituitary Diseases (CRMR-HYPO), Paris 94275, France; INSERM UMRS-1185 PHYSENDO, Physiologie et Physiopathologie Endocriniennes, Paris, France
| | - Frédéric Illouz
- Department of Endocrinology, Diabetology and Nutrition, Reference Center for Rare Pituitary Diseases HYPO, Angers University Hospital, Angers, France
| | - Lucie Levaillant
- Department of Pediatric Endocrinology, Reference Center for Rare Pituitary Diseases (CRMR-HYPO), Angers University Hospital, Angers, France
| | - Françoise Magnin
- INSERM UMRS-1185 PHYSENDO, Physiologie et Physiopathologie Endocriniennes, Paris, France
| | - Julie Perrot
- INSERM UMRS-1185 PHYSENDO, Physiologie et Physiopathologie Endocriniennes, Paris, France
| | - Abd El Kader Ait Tayeb
- INSERM UMRS-1185 PHYSENDO, Physiologie et Physiopathologie Endocriniennes, Paris, France; Department of Endocrinology, Diabetology and Nutrition, Reference Center for Rare Pituitary Diseases HYPO, Angers University Hospital, Angers, France; Paris-Saclay University, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Department of Molecular Genetics, Pharmacogenomics, and Hormonology, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Paris, France
| | - Claire Briet
- Department of Endocrinology, Diabetology and Nutrition, Reference Center for Rare Pituitary Diseases HYPO, Angers University Hospital, Angers, France
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jérôme Bouligand
- INSERM UMRS-1185 PHYSENDO, Physiologie et Physiopathologie Endocriniennes, Paris, France; Department of Endocrinology, Diabetology and Nutrition, Reference Center for Rare Pituitary Diseases HYPO, Angers University Hospital, Angers, France; Paris-Saclay University, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Department of Molecular Genetics, Pharmacogenomics, and Hormonology, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Paris, France
| | - Jacques Young
- Paris-Saclay University, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Department of Endocrinology and Reproductive Diseases, Reference Center for Rare Pituitary Diseases (CRMR-HYPO), Paris 94275, France; INSERM UMRS-1185 PHYSENDO, Physiologie et Physiopathologie Endocriniennes, Paris, France
| | - Luigi Maione
- Paris-Saclay University, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Department of Endocrinology and Reproductive Diseases, Reference Center for Rare Pituitary Diseases (CRMR-HYPO), Paris 94275, France; INSERM UMRS-1185 PHYSENDO, Physiologie et Physiopathologie Endocriniennes, Paris, France.
| |
Collapse
|
2
|
Nied E, Simonneaux M, Simonneaux V. The ticking clock sets the pace for female fertility. ANNALES D'ENDOCRINOLOGIE 2025; 86:101785. [PMID: 40339688 DOI: 10.1016/j.ando.2025.101785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
At a time when an increasing number of men and women face fertility issues, it is necessary to understand the basic mechanisms involved in mammalian reproductive activity in order to propose adapted therapeutic tools. This review describes how endogenous circadian clocks take part in the timing of reproductive cycles in female mammals and, consequently, how exposure to circadian disruption may impair female fertility. In female mammals, the master circadian clock, located in the hypothalamic suprachiasmatic nuclei (SCN), uses a vasopressinergic output to knock on preoptic kisspeptin (Kp) neurons each day at the onset of the active period. Kp is a potent activator of neurons producing the gonadotropin-releasing hormone (GnRH) controlling the release of the pituitary gonadotropins luteinizing (LH) and follicle-stimulating (FSH) hormones, which in turn promote ovarian gameto- and steroido-genesis. Estradiol, produced as oocytes mature, exerts positive feedback on Kp neurons. This dual control of Kp neuronal activity by the clock-driven vasopressin output and the elevated circulating estradiol allows a large increase in GnRH-induced LH release at the onset of the waking period, at the end of the follicular phase, triggering the release of mature oocytes. Additionally, different parts of the reproductive axis also host secondary circadian clocks that participate in the daily and ovarian regulation of female reproductive cycles. Different experiments revealed the functional significance of the circadian regulation of female reproduction. Indeed, exposure of female rodents to different protocols of circadian disruption impairs estrous cycle robustness, LH surge timing, and gestational success. Additionally, epidemiological studies indicate that women working non-standard schedules face increased risks of reproductive issues. Therefore, when women seek medical assistance for infertility, lifestyle factors, including work schedule organization, should be assessed. Chronotherapeutic interventions could then be considered to enhance the robustness of female reproductive cycles and, as a result, improve their reproductive health.
Collapse
Affiliation(s)
- Elisa Nied
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, 8 allée du Général Rouvillois, 67000 Strasbourg, France
| | - Marine Simonneaux
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, 8 allée du Général Rouvillois, 67000 Strasbourg, France
| | - Valérie Simonneaux
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, 8 allée du Général Rouvillois, 67000 Strasbourg, France.
| |
Collapse
|
3
|
Cutigni M, Cucina G, Galante E, Cerri M, Bizzarri M. Microgravity impairs endocrine signaling and reproductive health of women. A narrative review. Front Physiol 2025; 16:1558711. [PMID: 40432927 PMCID: PMC12106020 DOI: 10.3389/fphys.2025.1558711] [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: 01/10/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
During space exploration missions the organism is subjected to several challenges. Most of the studies have been performed on male health in space, leaving the focus on sex differences behind. With the development of new biological technologies, attention is now being paid more to how spaceflight conditions affect human reproductive health. In this review, the focus is on how weightlessness disrupts ovarian function and endocrine signaling by affecting the hypothalamic-pituitary-gonadal axis. Emerging evidence suggests that microgravity can impair estrogen production through the suppression of aromatase expression in granulosa cells. This condition leads to a hypo-estrogenic condition that harms the ovulation and the menstrual cycle. Likely, due to reduced estrogen availability, bone density, and cardiovascular health can consequently be severely involved. New studies focus on how space-related deregulation involving ovarian steroidogenesis look like the picture observed in the Polycystic Ovary Syndrome. These similarities open the perspective to counteract pharmacologically the observed abnormalities. However, our knowledge is severely constrained by the limited data available as well by the lack of proper experimental models of investigation. Indeed, much is required in order to acquire a full understanding of endocrine and functional changes occurring during microgravity exposure, including the joint effect of radiation and weightlessness that deserve to be thoroughly investigated to recognize the respective contribution of each one as well as the eventual synergies.
Collapse
Affiliation(s)
- Michela Cutigni
- Department of Experimental Medicine, Space Biomedicine Laboratory, University Sapienza, Rome, Italy
| | - Giorgia Cucina
- Department of Experimental Medicine, Space Biomedicine Laboratory, University Sapienza, Rome, Italy
| | - Emanuele Galante
- Department of Experimental Medicine, Space Biomedicine Laboratory, University Sapienza, Rome, Italy
| | - Matteo Cerri
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Mariano Bizzarri
- Department of Experimental Medicine, Space Biomedicine Laboratory, University Sapienza, Rome, Italy
| |
Collapse
|
4
|
Hernández VS, Zetter MA, Hernández‐Pérez OR, Hernández‐González R, Camacho‐Arroyo I, Millar RP, Eiden LE, Zhang L. Comprehensive chemoanatomical mapping, and the gonadal regulation, of seven kisspeptin neuronal populations in the mouse brain. J Neuroendocrinol 2025; 37:e70019. [PMID: 40102056 PMCID: PMC12045674 DOI: 10.1111/jne.70019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/30/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025]
Abstract
Kisspeptinergic signaling is well-established as crucial for the regulation of reproduction, but its potential broader role in brain function is less understood. This study investigates the distribution and chemotyping of kisspeptin-expressing neurons within the mouse brain. RNAscope single, dual, and multiplex in situ hybridization methods were used to assess kisspeptin mRNA (Kiss1) expression and its co-expression with other neuropeptides, excitatory and inhibitory neurotransmitter markers, and sex steroid receptors in wild-type intact and gonadectomized young adult mice. Seven distinct kisspeptin neuronal chemotypes were characterized, including two novel kisspeptin-expressing groups described for the first time, that is, the Kiss1 population in the ventral premammillary nucleus and the nucleus of the solitary tract. Kiss1 mRNA was also observed to localize in both somatic and dendritic compartments of hypothalamic neurons. High androgen receptor expression and changes in medial amygdala and septo-hypothalamic Kiss1 expression following GDX in males, but not in females, suggest a role for androgen receptors in regulating kisspeptin signaling. This study provides a detailed chemoanatomical map of kisspeptin-expressing neurons, highlighting their potential functional diversity. The discovery of a new kisspeptin-expressing group and gonadectomy-induced changes in Kiss1 expression patterns suggest broader roles for kisspeptin in brain functions beyond those of reproduction.
Collapse
Affiliation(s)
- Vito S. Hernández
- Department of Physiology, School of MedicineNational Autonomous University of Mexico (UNAM)Mexico CityMexico
- Section on Molecular NeuroscienceNIMH‐IRP, NIHBethesdaMarylandUSA
| | - Mario A. Zetter
- Department of Physiology, School of MedicineNational Autonomous University of Mexico (UNAM)Mexico CityMexico
- Department of Medicine and HealthUniversity of La SalleMexico CityMexico
| | - Oscar R. Hernández‐Pérez
- Department of Physiology, School of MedicineNational Autonomous University of Mexico (UNAM)Mexico CityMexico
| | | | - Ignacio Camacho‐Arroyo
- Research Unit in Human ReproductionNational Institute of Perinatology‐Faculty of Chemistry, UNAMMexico CityMexico
| | - Robert P. Millar
- Department of Physiology, School of MedicineNational Autonomous University of Mexico (UNAM)Mexico CityMexico
- Centre for NeuroendocrinologyUniversity of PretoriaPretoriaSouth Africa
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular MedicineUniversity of Cape TownCape TownSouth Africa
| | - Lee E. Eiden
- Section on Molecular NeuroscienceNIMH‐IRP, NIHBethesdaMarylandUSA
| | - Limei Zhang
- Department of Physiology, School of MedicineNational Autonomous University of Mexico (UNAM)Mexico CityMexico
- Section on Molecular NeuroscienceNIMH‐IRP, NIHBethesdaMarylandUSA
| |
Collapse
|
5
|
Igler A, Amodei R, Roselli CE. Anatomic distribution of kisspeptin neurons in the adult sheep amygdala: Associations with sex, estrogen receptor alpha, androgen receptor, and sexual partner preference. J Neuroendocrinol 2025; 37:e70011. [PMID: 40033683 PMCID: PMC12045730 DOI: 10.1111/jne.70011] [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: 09/18/2024] [Revised: 02/07/2025] [Accepted: 02/16/2025] [Indexed: 03/05/2025]
Abstract
Kisspeptin neurons are primarily known for regulating reproductive function by stimulating hormone release that controls puberty and fertility. While typically associated with the hypothalamus, recent research suggests their presence in other brain regions, including the amygdala. The amygdala, crucial for emotional processing and social behaviors, consists of various nuclei. However, the specific distribution and potential functional implications of kisspeptin neurons within this region remain unclear. Understanding kisspeptin neuron distribution in the sheep amygdala could provide insights into their roles in modulating reproductive functions, emotional, and social behaviors in a species closely related to humans. This study employed immunohistochemistry and RNAscope™ fluorescent in situ hybridization to map the distribution of kisspeptin fibers and cells in the amygdala of intact adult male and luteal-phase female sheep. The research also investigated the co-expression of Kiss1 with estrogen receptor-α (ESR1) and androgen receptor (AR) mRNA, as well as the presence of kisspeptin receptor (Kiss1r) mRNA-containing cells. Kisspeptin immunoreactive fibers were most dense in the medial amygdala, while Kiss1 mRNA-containing cells were abundant in the medial, cortical, and basal nuclei. Extensive co-expression of Kiss1 with ESR1 and AR mRNA was observed. In the posterior medial nucleus, 80% of kisspeptin neurons co-expressed ESR1, and 40% co-expressed AR. Kiss1r mRNA-containing cells were found in the medial, cortical, and basal nuclei and co-localized within cells expressing Kiss1 mRNA. No differences in kisspeptin cell numbers were found between rams and ewes or between rams with different sexual partner preferences. This study provides a foundational map of the kisspeptin system in the sheep amygdala, offering insights into its potential roles in reproductive, emotional, and social behaviors. The extensive co-expression of Kiss1 mRNA with ESR1 and AR mRNA suggests possible regulation by sex steroids, while the presence of Kiss1r mRNA-containing cells indicates potential autocrine or paracrine signaling. These findings contribute to our understanding of kisspeptin neurons' distribution and potential functions beyond the hypothalamus, particularly in the amygdala.
Collapse
Affiliation(s)
- Anna Igler
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Rebecka Amodei
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Charles E Roselli
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
6
|
Zhang L, Hernández VS, Zetter MA, Hernández‐Pérez OR, Hernández‐González R, Camacho‐Arroyo I, Eiden LE, Millar RP. Kisspeptin fiber and receptor distribution analysis suggests its potential role in central sensorial processing and behavioral state control. J Neuroendocrinol 2025; 37:e70007. [PMID: 40065551 PMCID: PMC12045677 DOI: 10.1111/jne.70007] [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: 09/13/2024] [Revised: 01/01/2025] [Accepted: 02/03/2025] [Indexed: 03/19/2025]
Abstract
Kisspeptin (KP) signaling in the brain is defined by the anatomical distribution of KP-producing neurons, their fibers, receptors, and connectivity. Technological advances have prompted a re-evaluation of these chemoanatomical aspects, originally studied in the early years after the discovery of KP and its receptor Kiss1r. Previously, we characterized (Hernández et al. bioRxiv 2024) seven KP neuronal populations in the mouse brain at the mRNA level, including two novel populations, and examined their response to gonadectomy. In this study, we mapped KP fiber distribution in rats and mice using immunohistochemistry under intact as well as short- and long-term post-gonadectomy conditions. Kiss1r mRNA expression was examined via RNAscope, in relation to vesicular GABA transporter (Slc32a1) in whole mouse brain, and to KP and vesicular glutamate transporter 2 (Slc17a6), Kiss1, and Slc32a1 in hypothalamic RP3V and arcuate regions. We identified KP fibers in 118 brain regions, primarily in extra-hypothalamic areas associated with sensorial processing and behavioral state control. KP-immunoreactive fiber density and distribution were largely unchanged by gonadectomy. Kiss1r was expressed prominently in sensorial and state control regions such as the septal nuclei, the suprachiasmatic nucleus, locus coeruleus, hippocampal layers, thalamic nuclei, and cerebellar structures. Co-expression of Kiss1r and Kiss1 was observed in hypothalamic neurons, suggesting both autocrine and paracrine KP signaling mechanisms. These findings enhance our understanding of KP signaling beyond reproductive functions, particularly in sensorial processing and behavioral state regulation. This study opens new avenues for investigating KP's role in controlling complex physiological processes, including those unrelated to reproduction.
Collapse
Affiliation(s)
- Limei Zhang
- Department of Physiology, School of MedicineNational Autonomous University of MexicoMexico CityMexico
- Section on Molecular Neuroscience, NIMH‐IRP, NIHBethesdaMarylandUSA
| | - Vito Salvador Hernández
- Department of Physiology, School of MedicineNational Autonomous University of MexicoMexico CityMexico
- Section on Molecular Neuroscience, NIMH‐IRP, NIHBethesdaMarylandUSA
| | - Mario Alberto Zetter
- Department of Physiology, School of MedicineNational Autonomous University of MexicoMexico CityMexico
- Department of Medicine and HealthUniversity of La SalleMexico CityMexico
| | | | | | - Ignacio Camacho‐Arroyo
- Research Unit in Human Reproduction, National Institute of Perinatology‐Faculty of ChemistryNational Autonomous University of MexicoMexico CityMexico
| | - Lee E. Eiden
- Section on Molecular Neuroscience, NIMH‐IRP, NIHBethesdaMarylandUSA
| | - Robert P. Millar
- Department of Physiology, School of MedicineNational Autonomous University of MexicoMexico CityMexico
- Centre for NeuroendocrinologyUniversity of PretoriaPretoriaSouth Africa
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular MedicineUniversity of Cape TownCape TownSouth Africa
| |
Collapse
|
7
|
Koysombat K, Tsoutsouki J, Patel AH, Comninos AN, Dhillo WS, Abbara A. Kisspeptin and neurokinin B: roles in reproductive health. Physiol Rev 2025; 105:707-764. [PMID: 39813600 DOI: 10.1152/physrev.00015.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 10/17/2024] [Accepted: 11/13/2024] [Indexed: 01/18/2025] Open
Abstract
Kisspeptin and neurokinin B (NKB) play a key role in several physiological processes including in puberty, adult reproductive function including the menstrual cycle, as well as mediating the symptoms of menopause. Infundibular kisspeptin neurons, which coexpress NKB, regulate the activity of gonadotropin-releasing hormone (GnRH) neurons and thus the physiological pulsatile secretion of GnRH from the hypothalamus. Outside of their hypothalamic reproductive roles, these peptides are implicated in several physiological functions including sexual behavior and attraction, placental function, and bone health. Over the last two decades, research findings have considerably enhanced our understanding of the physiological regulation of the hypothalamic-pituitary-gonadal (HPG) axis and identified potential therapeutic applications. For example, recognition of the role of kisspeptin as the natural inductor of ovulation has led to research investigating its use as a safer, more physiological trigger of oocyte maturation in in vitro fertilization (IVF) treatment. Moreover, the key role of NKB in the pathophysiology of menopausal hot flashes has led to the development of pharmacological antagonism of this pathway. Indeed, fezolinetant, a neurokinin 3 receptor antagonist, has recently received Food and Drug Administration (FDA) approval for clinical use to treat menopausal vasomotor symptoms. Here, we discuss the roles of kisspeptin and NKB in human physiology, including in the regulation of puberty, menstrual cyclicity, reproductive behavior, pregnancy, menopause, and bone homeostasis. We describe how perturbations of these key physiological processes can result in disease states and consider how kisspeptin and NKB could be exploited diagnostically as well as therapeutically to treat reproductive disorders.
Collapse
Affiliation(s)
- Kanyada Koysombat
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Jovanna Tsoutsouki
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Aaran H Patel
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Alexander N Comninos
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Waljit S Dhillo
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Ali Abbara
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
8
|
Amodei R, Jonker SS, Lazen E, Nestor CC, Estill CT, Roselli CE. KNDy Neurons and the Control of the Gonadotropic Axis in the Midgestation Fetal Sheep. Endocrinology 2025; 166:bqaf049. [PMID: 40048705 PMCID: PMC12168063 DOI: 10.1210/endocr/bqaf049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Indexed: 03/25/2025]
Abstract
KNDy neurons, located in the hypothalamic arcuate nucleus, coexpress kisspeptin (Kiss), neurokinin B, and dynorphin and play a crucial role in regulating GnRH/LH secretion in midgestation sheep fetuses. We hypothesize that KNDy-GnRH signaling is established during midgestation, with negative feedback acting through KNDy neurons regulating testosterone levels needed for brain masculinization in male fetuses. We used immunofluorescence histochemistry to assess the effect of chemical castration with the GnRH antagonist degarelix on arcuate KNDy neurons in fetal sheep. Fluorescent in situ hybridization demonstrated the presence of steroid receptors in untreated midgestation fetal kisspeptin neurons. Additionally, unanesthetized cannulated midgestation fetal sheep were used to examine the effects of KNDy peptides on LH secretion and characterize receptor specificity. Treatment of male lamb fetuses with degarelix on day 62 of gestation resulted in significantly decreased plasma LH and testosterone concentrations (P < .05), accompanied by a significant increase in arcuate Kiss neurons (P < .05). In unanesthetized cannulated fetuses, bolus administration of KP-10 (a Kiss receptor agonist) and senktide (NK3 receptor agonist) elicited robust LH release within 15 minutes. Pretreatment with the NK3 receptor antagonist SB222200 blocked the LH response to senktide, whereas P271 (Kiss receptor antagonist) did not affect basal LH or block the LH response to KP-10. Blocking κ-opiate receptor with PF4455242 significantly increased LH release. These results support the hypothesis that KNDy neurons regulate GnRH and gonadotropin secretion in midgestation sheep fetuses, acting as targets for negative feedback to maintain a stable androgen environment crucial for brain masculinization.
Collapse
Affiliation(s)
- Rebecka Amodei
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239-3098, USA
| | - Sonnet S Jonker
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR 97239-3098, USA
| | - Evelyn Lazen
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239-3098, USA
| | - Casey C Nestor
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Charles T Estill
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331-4501, USA
- College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331-4501, USA
| | - Charles E Roselli
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239-3098, USA
| |
Collapse
|
9
|
Phillips CD, DeFazio RA, Moenter SM. Sex and Time of Day Alter the Interactions Between Hypothalamic Glia and the Neural Circuits Controlling Reproduction. Endocrinology 2025; 166:bqaf057. [PMID: 40111184 PMCID: PMC11968336 DOI: 10.1210/endocr/bqaf057] [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: 01/30/2025] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 03/22/2025]
Abstract
An upstream network, including glia and arcuate nucleus (ARC) kisspeptin neurons, controls hormone secretion from preoptic area (POA) gonadotropin-releasing hormone (GnRH) neurons, which form the final common pathway for the central control of fertility. In males, chemogenetic activation of Gq-mediated signaling in POA glia activated GnRH neurons and downstream luteinizing hormone (LH) release, while chemogenetic activation of ARC glia had no effect on ARC kisspeptin neurons. We characterized sex differences and time-of-day effects in these critical circuits to understand their effects on reproduction. Chemogenetic activation of glial fibrillary acidic protein (GFAP)-expressing cells increased intracellular calcium concentrations regardless of sex, brain region, or time of day. Activation of POA glia or treatment with the gliotransmitter analog dimethyl prostaglandin E2 (dmPGE2) increased GnRH neuron firing rate, and these responses were dependent upon sex and time of day. In contrast, ARC kisspeptin neuron firing rate was unresponsive to ARC glia activation or dmPGE2 regardless of sex or time of day. POA glial activation increased LH levels in males and females but the response in males was more rapid. ARC glia activation had no effect on LH in females and the response in males was delayed compared to POA glia activation. A similar LH response persisted after ARC kisspeptin neuron ablation, suggesting it is not mediated by those neurons. GnRH neurons, rather than arcuate kisspeptin neurons, are thus the main target of glial regulation of reproductive neuroendocrine output and this regulation is dependent on sex and time of day.
Collapse
Affiliation(s)
- Chrystian D Phillips
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - R Anthony DeFazio
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Suzanne M Moenter
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
10
|
Bonaldo B, Casile A, Bettarelli M, Marraudino M, Gotti S. Perinatal exposure to bisphenol A or S alters differently sexual behavior and kisspeptin system in mice. ENVIRONMENTAL RESEARCH 2025; 269:120888. [PMID: 39828186 DOI: 10.1016/j.envres.2025.120888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/09/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
The effects of bisphenol A (BPA), a highly diffused endocrine-disrupting chemical found mainly in plastics, on neural circuits and behaviors are well-known. However, the effects of its substitutes have not been fully investigated. Thus, in the present study, we compare the effects of perinatal exposure to bisphenol A or S (BPS) on reproductive behaviors and related hypothalamic kisspeptin system in mice. C57BL/6J dams were orally treated with 4 μg/kg body weight/day of BPA, BPS, or vehicle from mating until the weaning of the offspring. In the adult offspring, we performed the two-bedding T-Maze test, and we observed the spontaneous sexual behavior. Exposure to BPA caused a delay in puberty onset in females, while BPS caused anticipation in males, and both altered the estrous cycle in females. The sexual and sexual-related behaviors were partially altered in males, especially in the BPA-exposed ones. Regarding the kisspeptin immunoreactivity in the analyzed hypothalamic nuclei, in BPA- or BPS-treated females, we observed an increase within the rostral periventricular area, while BPA led to an increase in the paraventricular nucleus, and BPS induced a reduction compared to control females. Among males, we observed a significant increase in the arcuate nucleus of BPA-treated males and a significant decrease in the paraventricular nucleus of BPS-treated ones. These results support the idea that perinatal exposure to low doses of either BPA or BPS is altering, in a sexually differentiated way, some reproductive-relevant parameters, sexual behaviors, and kisspeptin hypothalamic nuclei.
Collapse
Affiliation(s)
- Brigitta Bonaldo
- Department of Health Sciences and Research Center on Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy; Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043, Orbassano, Turin, Italy.
| | - Antonino Casile
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043, Orbassano, Turin, Italy; School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 9, Camerino, 62032, Italy
| | - Martina Bettarelli
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043, Orbassano, Turin, Italy
| | - Marilena Marraudino
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043, Orbassano, Turin, Italy; Department of Neuroscience "Rita Levi-Montalcini", University of Turin, Via Cherasco 15, 10126, Turin, Italy
| | - Stefano Gotti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043, Orbassano, Turin, Italy; Department of Neuroscience "Rita Levi-Montalcini", University of Turin, Via Cherasco 15, 10126, Turin, Italy
| |
Collapse
|
11
|
Yang Q, Jia S, Tao J, Zhang J, Fan Z. Multiple effects of kisspeptin on neuroendocrine, reproduction, and metabolism in polycystic ovary syndrome. J Neuroendocrinol 2025; 37:e13482. [PMID: 39694850 DOI: 10.1111/jne.13482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a highly prevalent and heterogeneous disease characterized by a combination of reproductive and endocrine abnormalities, often associated with metabolic and mental health disorders. The etiology and pathogenesis of PCOS remain unclear, but recent research has increasingly focused on the upstream mechanisms underlying its development. Among these, kisspeptin (KISS) signaling has emerged as a pivotal component in the regulation of the hypothalamic-pituitary-gonadal axis, with significant roles in reproductive function, energy regulation, and metabolism. Women with PCOS commonly exhibit disruptions in gonadotropin secretion, including elevated luteinizing hormone (LH) levels, imbalanced LH/follicle-stimulating hormone (FSH) ratios, and increased androgen levels, all of which are usually parallel with abnormal KISS signaling. Furthermore, alterations in the KISS/KISS1R system within the central and circulatory systems, as well as peripheral tissues, have been implicated in the development of PCOS. These changes affect multiple pathophysiological domains, including reproductive function, energy regulation, metabolic homeostasis, inflammatory response, and emotional disorders, and are further influenced by lifestyle and environmental factors. This review aims to comprehensively summarize the existing experimental and clinical evidence supporting these roles of KISS in PCOS, with the goal of establishing a foundation for future research and potential clinical applications.
Collapse
Affiliation(s)
- Qiaorui Yang
- Department of Gynecology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shengxiao Jia
- Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Jing Tao
- Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Jinfu Zhang
- Department of Gynecology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Gynecology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai, China
| | - Zhenliang Fan
- Nephrology Department, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Zhejiang, China
| |
Collapse
|
12
|
Curi TZ, Passoni MT, Tolouei SEL, de Araújo Ramos AT, de Almeida SCF, Romano RM, de Oliveira JM, Dalsenter PR, Martino-Andrade AJ. In Utero and Lactational Exposure to an Environmentally Relevant Mixture of Phthalates Alters Hypothalamic Gene Expression and Sexual Preference in Rats. ENVIRONMENTAL TOXICOLOGY 2025; 40:54-65. [PMID: 39248502 DOI: 10.1002/tox.24414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/19/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024]
Abstract
Several phthalates, mainly used as plasticizers, are known for their adverse effects on the male genital system. Previously, we demonstrated that an environmentally relevant mixture of six antiandrogenic phthalates (PMix), derived from a biomonitoring study in pregnant Brazilian women, was able to disrupt the reproductive development in male rats. Experimental groups (control, 0.1, 0.5, and 500 mg PMix/kg/day) were established starting from the extrapolated human dose (0.1 mg/kg/day), followed by doses 5 times and 5000 times higher. Pregnant rats received daily oral gavage administration of either vehicle (control) or PMix from gestational day 13 to postnatal day 10. Here, we examined male and female offspring regarding changes in gene expression of key reproductive factors in the hypothalamus and pituitary gland at adulthood and conducted a battery of behavioral tests in males, including partner preference, sexual behavior, and male attractiveness tests. PMix induced some changes in mating-related behavior in males, as demonstrated by the absence of preference for females against males and a higher number of penetrations up to ejaculation in the 0.5 dose group. PMix decreased Esr2 expression in the male hypothalamus across all three doses, and in females at mid and high doses in both the hypothalamus and pituitary. In male hypothalamus, we also observed decreased Kiss1 transcripts in these groups and a reduction in AR at the 0.5 dose group. In summary, our results provide further evidence that phthalates in a mixture, even at low doses, may exert cumulative effects on the structures underlying sexual behavior, which seems to be more sensitive than reproductive endpoints for the same experimental design.
Collapse
Affiliation(s)
- Tatiana Zauer Curi
- Reproductive Toxicology Laboratory, Department of Pharmacology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Marcella Tapias Passoni
- Reproductive Toxicology Laboratory, Department of Pharmacology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Sara Emilia Lima Tolouei
- Reproductive Toxicology Laboratory, Department of Pharmacology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Anderson Tadeu de Araújo Ramos
- Animal Endocrine and Reproductive Physiology Laboratory, Department of Physiology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Samara Christina França de Almeida
- Animal Endocrine and Reproductive Physiology Laboratory, Department of Physiology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Renata Marino Romano
- Reproductive Toxicology Laboratory, Department of Pharmacy, State University of Centro-Oeste, Guarapuava, Brazil
| | - Jeane Maria de Oliveira
- Reproductive Toxicology Laboratory, Department of Pharmacy, State University of Centro-Oeste, Guarapuava, Brazil
| | - Paulo Roberto Dalsenter
- Reproductive Toxicology Laboratory, Department of Pharmacology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Anderson Joel Martino-Andrade
- Reproductive Toxicology Laboratory, Department of Pharmacology, Federal University of Paraná (UFPR), Curitiba, Brazil
- Animal Endocrine and Reproductive Physiology Laboratory, Department of Physiology, Federal University of Paraná (UFPR), Curitiba, Brazil
| |
Collapse
|
13
|
Zhang L, Hernández VS, Zetter MA, Hernández-Pérez OR, Hernández-González R, Camacho-Arroyo I, Eiden LE, Millar RP. Kisspeptin fiber and receptor distribution analysis suggests its potential role in central sensorial processing and behavioral state control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.05.556375. [PMID: 39651138 PMCID: PMC11623528 DOI: 10.1101/2023.09.05.556375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Background Kisspeptin (KP) signaling in the brain is defined by the anatomical distribution of KP-producing neurons, their fibers, receptors, and connectivity. Technological advances have prompted a re-evaluation of these chemoanatomical aspects, originally studied in the early years after the discovery of KP and its receptor Kiss1r. We have previously characterized(1) seven KP neuronal populations in the mouse brain at the mRNA level, including two novel populations, and examined their short-term response to gonadectomy. Methods In this study, we mapped KP fiber distribution in rats and mice using immunohistochemistry under intact and short- and long-term post-gonadectomy conditions. Kiss1r mRNA expression was examined via RNAscope, in relation to vesicular GABA transporter ( Slc32a1 ) in whole mouse brain and to KP and vesicular glutamate transporter 2 ( Kiss1 and Slc17a6 ) in hypothalamic RP3V and arcuate regions. Results We identified KP fibers in 118 brain regions, primarily in extra-hypothalamic areas associated with sensorial processing and behavioral state control. KP-immunoreactive fiber density and distribution were largely unchanged by gonadectomy. Kiss1r was expressed prominently in sensorial and state control regions such as septal nuclei, the suprachiasmatic nucleus, locus coeruleus, hippocampal layers, thalamic nuclei, and cerebellar structures. Co-expression of Kiss1r and Kiss1 was observed in hypothalamic neurons, suggesting both autocrine and paracrine KP signaling mechanisms. Conclusion These findings enhance our understanding of KP signaling beyond reproductive functions, particularly in sensorial and behavioral state regulation. This study opens new avenues for investigating KP's role in controlling complex physiological processes, including those not related to reproduction.
Collapse
|
14
|
Hernandez VS, Zetter MA, Hernandez-Perez OR, Hernandez-Gonzalez R, Camacho-Arroyo IS, Millar RP, Eiden LE, Zhang L. Comprehensive chemotyping, and the gonadal regulation, of seven kisspeptinergic neuronal populations in the mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.23.604881. [PMID: 39211104 PMCID: PMC11361108 DOI: 10.1101/2024.07.23.604881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
BACKGROUND Kisspeptinergic signaling is well-established as crucial for regulation of reproduction, but its potential broader role in brain function is less understood. This study investigates the distribution and chemotyping of kisspeptin-expressing neurons within the mouse brain. METHODS RNAscope singleplex, duplex and multiplex in situ hybridization methods were used to assess kisspeptin mRNA (Kiss1) expression and its co-expression with other neuropeptides, excitatory and inhibitory neurotransmitter markers, and sex steroid receptors in intact and gonadectomized young adult mice. RESULTS Seven distinct kisspeptin neuronal chemotypes were characterized, including within two novel Kiss1-expressing groups described here for the first time: the ventral premammillary nucleus, and the nucleus of the solitary tract. Kiss1 mRNA was also localized in the soma, and within the dendritic compartment, of hypothalamic neurons. Altered Kiss1 expression following gonadectomy suggests a previously unappreciated role for androgen receptors in regulating kisspeptin signaling. CONCLUSION This study provides a detailed chemoanatomical map of kisspeptin-expressing neurons in the brain, highlighting their potential functional diversity. The discovery of new kisspeptin-expressing neuronal populations, and gonadectomy-induced changes in Kiss1 expression patterns, provide a basis for further exploration of non-endocrine roles for kisspeptin in brain function.
Collapse
|
15
|
Pfau DR, Schwartz AR, Dela Cruz C, Padmanabhan V, Moravek MB, Shikanov A. A Mouse Model to Investigate the Impact of Gender Affirming Hormone Therapy with Estradiol on Reproduction. Adv Biol (Weinh) 2024; 8:e2300126. [PMID: 37688350 PMCID: PMC10920391 DOI: 10.1002/adbi.202300126] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/21/2023] [Indexed: 09/10/2023]
Abstract
Gender-affirming hormone therapy (GAHT) can help transgender and/or gender diverse (TGD) individuals achieve emobidment goals that align with their transition needs. Clinical evidence from estradiol (E)-GAHT patients indicate widespread changes in tissues sensitive to E and testosterone (T), particularly in the reproductive system. Notably, E-GAHTs effects on hormones and reproduction vary greatly between patients. With the goal of informing clinical research and practice for TGD individuals taking E, this study examines intact male mice implanted with capsules containing one of three different E doses (low 1.25 mg; mid 2.5 mg; high 5 mg), or a blank control capsule. All E-GAHT doses suppress T and follicle stimulating hormone levels while elevating E levels. Only the high E-GAHT dose significantly supresses luteinizing hormone levels. All E-GAHT doses affect epididymis tubule size similarly while seminiferous tubule morphology and bladder weight changes are dose-dependent. E-GAHT does not alter the presence of mature sperm, though E-exposed sperm have altered motility. These data represent the first evidence that mouse models offer an effective tool to understand E-GAHTs impact on reproductive health and the dose-dependent effects of this model permit examinations of diverse patient outcomes.
Collapse
Affiliation(s)
- DR Pfau
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor MI 48109, USA
| | - AR Schwartz
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor MI 48109, USA
| | - C Dela Cruz
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor MI 48109, USA
| | - V Padmanabhan
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor MI 48109, USA
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109, USA
| | - MB Moravek
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor MI 48109, USA
- Division of Reproductive Endocrinology and Infertility, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Urology, University of Michigan, Ann Arbor, MI 48109, USA
| | - A Shikanov
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor MI 48109, USA
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
- Biomedical Engineering Department, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
16
|
Kauffman AS. Androgen Inhibition of Reproductive Neuroendocrine Function in Females and Transgender Males. Endocrinology 2024; 165:bqae113. [PMID: 39207217 PMCID: PMC11393496 DOI: 10.1210/endocr/bqae113] [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: 07/05/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Ovarian function is controlled by pituitary secretion of luteinizing hormone (LH) and follicle stimulating hormone (FSH), which in turn are governed by gonadotropin releasing hormone (GnRH) secreted from the brain. A fundamental principle of reproductive axis regulation is negative feedback signaling by gonadal sex steroids back to the brain to fine-tune GnRH and gonadotropin secretion. Endogenous negative feedback effects can be mimicked by exogenous steroid treatments, including androgens, in both sexes. Indeed, a growing number of clinical and animal studies indicate that high levels of exogenous androgens, in the typically male physiological range, can inhibit LH secretion in females, as occurs in males. However, the mechanisms by which male-level androgens inhibit GnRH and LH secretion still remain poorly understood, and this knowledge gap is particularly pronounced in transgender men (individuals designated female at birth but identifying as male). Indeed, many transgender men take long-term gender-affirming hormone therapy that mimics male-level testosterone levels. The impact of such gender-affirming testosterone on the reproductive axis, both at the ovarian and neuroendocrine level, is a long-understudied area that still requires further investigation. Importantly, the few concepts of androgen actions in females mostly come from studies of polycystic ovary syndrome, which does not recapitulate a similar androgen milieu or a pathophysiology of inhibited LH secretion as occurs in testosterone-treated transgender men. This review summarizes clinical evidence indicating that exogenous androgens can impair neuroendocrine reproductive function in both female individuals and transgender men and highlights emerging experimental data supporting this in recently developed transgender rodent models.
Collapse
Affiliation(s)
- Alexander S Kauffman
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
17
|
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
|
18
|
Hess RA, Park CJ, Soto S, Reinacher L, Oh JE, Bunnell M, Ko CJ. Male animal sterilization: history, current practices, and potential methods for replacing castration. Front Vet Sci 2024; 11:1409386. [PMID: 39027909 PMCID: PMC11255590 DOI: 10.3389/fvets.2024.1409386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
Sterilization and castration have been synonyms for thousands of years. Making an animal sterile meant to render them incapable of producing offspring. Castration or the physical removal of the testes was discovered to be the most simple but reliable method for managing reproduction and sexual behavior in the male. Today, there continues to be global utilization of castration in domestic animals. More than six hundred million pigs are castrated every year, and surgical removal of testes in dogs and cats is a routine practice in veterinary medicine. However, modern biological research has extended the meaning of sterilization to include methods that spare testis removal and involve a variety of options, from chemical castration and immunocastration to various methods of vasectomy. This review begins with the history of sterilization, showing a direct link between its practice in man and animals. Then, it traces the evolution of concepts for inducing sterility, where research has overlapped with basic studies of reproductive hormones and the discovery of testicular toxicants, some of which serve as sterilizing agents in rodent pests. Finally, the most recent efforts to use the immune system and gene editing to block hormonal stimulation of testis function are discussed. As we respond to the crisis of animal overpopulation and strive for better animal welfare, these novel methods provide optimism for replacing surgical castration in some species.
Collapse
Affiliation(s)
- Rex A. Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Epivara, Inc, Champaign, IL, United States
| | - Chan Jin Park
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Epivara, Inc, Champaign, IL, United States
| | | | | | - Ji-Eun Oh
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Mary Bunnell
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - CheMyong J. Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Epivara, Inc, Champaign, IL, United States
| |
Collapse
|
19
|
van Rosmalen L, Deota S, Maier G, Le HD, Lin T, Ramasamy RK, Hut RA, Panda S. Energy balance drives diurnal and nocturnal brain transcriptome rhythms. Cell Rep 2024; 43:113951. [PMID: 38508192 PMCID: PMC11330649 DOI: 10.1016/j.celrep.2024.113951] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/30/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Plasticity in daily timing of activity has been observed in many species, yet the underlying mechanisms driving nocturnality and diurnality are unknown. By regulating how much wheel-running activity will be rewarded with a food pellet, we can manipulate energy balance and switch mice to be nocturnal or diurnal. Here, we present the rhythmic transcriptome of 21 tissues, including 17 brain regions, sampled every 4 h over a 24-h period from nocturnal and diurnal male CBA/CaJ mice. Rhythmic gene expression across tissues comprised different sets of genes with minimal overlap between nocturnal and diurnal mice. We show that non-clock genes in the suprachiasmatic nucleus (SCN) change, and the habenula was most affected. Our results indicate that adaptive flexibility in daily timing of behavior is supported by gene expression dynamics in many tissues and brain regions, especially in the habenula, which suggests a crucial role for the observed nocturnal-diurnal switch.
Collapse
Affiliation(s)
- Laura van Rosmalen
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Shaunak Deota
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Geraldine Maier
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Hiep D Le
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Terry Lin
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ramesh K Ramasamy
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Roelof A Hut
- Chronobiology Unit, Groningen Institute for Evolutionary Life Sciences, University of Groningen, 9700 CC Groningen, the Netherlands.
| | - Satchidananda Panda
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
20
|
Yacca SS, Kanasaki H, Tumurbaatar T, Cairang Z, Oride A, Okada H, Kyo S. Changes in pituitary gonadotropin subunits and hypothalamic Kiss-1 gene expression by administration of sex steroids in ovary-intact female rats. Endocrine 2024; 83:733-746. [PMID: 37966704 DOI: 10.1007/s12020-023-03596-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 11/03/2023] [Indexed: 11/16/2023]
Abstract
OBJECTIVE We examined how the sex steroids influence the synthesis of gonadotropins. MATERIALS AND METHODS The effects of sex steroids estradiol (E2), progesterone (P4), and dihydrotestosterone (DHT) in pituitary gonadotroph cell model (LβT2 cells) in vitro and ovary-intact rats in vivo were examined. The effects of sex steroids on Kiss1 gene expression in the hypothalamus were also examined in ovary-intact rats. RESULTS In LβT2 cells, E2 increased common glycoprotein alpha (Cga) and luteinizing hormone beta (Lhb) subunit promoter activity as well as their mRNA expression. Although gonadotropin subunit promoter activity was not modulated by P4, Cga and Lhb mRNA expression was increased by P4. DHT inhibited Cga and Lhb mRNA expression with a concomitant decrease in their promoter activity. During the 2-week administration of exogenous E2 to ovary-intact rats, the estrous cycle determined by vaginal smears was disrupted. P4 or DHT administration completely eliminated the estrous cycle. Protein expression of all three gonadotropin subunits within the pituitary gland was inhibited by E2 or P4 treatment in vivo; however, DHT reduced Cga expression but did not modulate Lhb or follicle-stimulating hormone beta subunit expression. E2 administration significantly repressed Kiss1 mRNA expression in a posterior hypothalamic region that included the arcuate nucleus. P4 and DHT did not modulate Kiss1 mRNA expression in this region. In contrast, P4 administration significantly inhibited Kiss1 mRNA expression in the anterior region of the hypothalamus that included the anteroventral periventricular nucleus. The expression of gonadotropin-releasing hormone (Gnrh) mRNA in the anterior hypothalamic region, where the preoptic area is located, appeared to be decreased by treatment with E2 and P4. CONCLUSION Our findings suggest that sex steroids have different effects in the hypothalamus and pituitary gland.
Collapse
Affiliation(s)
- Susdiaman S Yacca
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Haruhiko Kanasaki
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, 693-8501, Japan.
| | - Tuvshintugs Tumurbaatar
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Zhuoma Cairang
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Aki Oride
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Hiroe Okada
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Satoru Kyo
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, 693-8501, Japan
| |
Collapse
|
21
|
Moore AM, Novak AG, Lehman MN. KNDy Neurons of the Hypothalamus and Their Role in GnRH Pulse Generation: an Update. Endocrinology 2023; 165:bqad194. [PMID: 38170643 PMCID: PMC10768882 DOI: 10.1210/endocr/bqad194] [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: 10/23/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024]
Abstract
There is considerable evidence that synchronized activity within a reciprocally connected population of cells in the arcuate nucleus (ARC) coexpressing kisspeptin, neurokinin B (NKB), and dynorphin (KNDy cells) is crucial for the generation of gonadotrophin-releasing hormone (GnRH) pulses in mammals. The initial "KNDy hypothesis" proposed that pulsatile GnRH secretion is elicited by episodic kisspeptin release from KNDy cells following synchronized activation and termination of the population by NKB and dynorphin, respectively. Since then, the role of KNDy cells as a critical component of the pulse generator has been further supported by studies at the single-cell level, demonstrating that the population is both necessary and sufficient for pulsatility. In addition, there have been considerable modifications and expansion of the original hypothesis, including work demonstrating the critical role of glutamate in synchronization of the KNDy cell network, functional interactions with other ARC subpopulations, and the existence of species differences in the role of dynorphin in pulse generation. Here we review these recent changes and discuss how the translation of these findings has led to the development of new therapies for disorders related to pulse generation. We also outline critical gaps in knowledge that are currently limiting the application of KNDy research in the clinic, particularly regarding the role of dynorphin in pulse generation in primates.
Collapse
Affiliation(s)
- Aleisha M Moore
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| | - Alyssa G Novak
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| | - Michael N Lehman
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
22
|
Morishita M, Higo S, Iwata K, Ishii H. Sex and interspecies differences in ESR2-expressing cell distributions in mouse and rat brains. Biol Sex Differ 2023; 14:89. [PMID: 38111056 PMCID: PMC10726529 DOI: 10.1186/s13293-023-00574-z] [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: 07/07/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND ESR2, a nuclear estrogen receptor also known as estrogen receptor β, is expressed in the brain and contributes to the actions of estrogen in various physiological phenomena. However, its expression profiles in the brain have long been debated because of difficulties in detecting ESR2-expressing cells. In the present study, we aimed to determine the distribution of ESR2 in rodent brains, as well as its sex and interspecies differences, using immunohistochemical detection with a well-validated anti-ESR2 antibody (PPZ0506). METHODS To determine the expression profiles of ESR2 protein in rodent brains, whole brain sections from mice and rats of both sexes were subjected to immunostaining for ESR2. In addition, to evaluate the effects of circulating estrogen on ESR2 expression profiles, ovariectomized female mice and rats were treated with low or high doses of estrogen, and the resulting numbers of ESR2-immunopositive cells were analyzed. Welch's t-test was used for comparisons between two groups for sex differences, and one-way analysis of variance followed by the Tukey-Kramer test were used for comparisons among multiple groups with different estrogen treatments. RESULTS ESR2-immunopositive cells were observed in several subregions of mouse and rat brains, including the preoptic area, extended amygdala, hypothalamus, mesencephalon, and cerebral cortex. Their distribution profiles exhibited sex and interspecies differences. In addition, low-dose estrogen treatment in ovariectomized female mice and rats tended to increase the numbers of ESR2-immunopositive cells, whereas high-dose estrogen treatment tended to decrease these numbers. CONCLUSIONS Immunohistochemistry using the well-validated PPZ0506 antibody revealed a more localized expression of ESR2 protein in rodent brains than has previously been reported. Furthermore, there were marked sex and interspecies differences in its distribution. Our histological analyses also revealed estrogen-dependent changes in ESR2 expression levels in female brains. These findings will be helpful for understanding the ESR2-mediated actions of estrogen in the brain.
Collapse
Affiliation(s)
- Masahiro Morishita
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Shimpei Higo
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Kinuyo Iwata
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Hirotaka Ishii
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan.
| |
Collapse
|
23
|
Grant B, Campbell J, Pradeep A, Burns AD, Bassett P, Abbara A, Saket P, Minhas S, Dhillo WS, McVeigh J, Bhasin S, Jayasena CN. Factors predicting normalization of reproductive hormones after cessation of anabolic-androgenic steroids in men: a single center retrospective study. Eur J Endocrinol 2023; 189:601-610. [PMID: 38102386 DOI: 10.1093/ejendo/lvad164] [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: 07/23/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 12/17/2023]
Abstract
OBJECTIVE Symptomatic hypogonadism discourages men from stopping anabolic-androgenic steroids (AAS). Some men illicitly take drugs temporarily stimulating endogenous testosterone following AAS cessation (post-cycle therapy; PCT) to lessen hypogonadal symptoms. We investigated whether prior PCT use was associated with the normalization of reproductive hormones following AAS cessation. METHODS Retrospective analysis of 641 men attending a clinic between 2015-2022 for a single, nonfasting, random blood test <36 months following AAS cessation, with or without PCT. Normalized reproductive hormones (ie, a combination of reference range serum luteinizing hormone, follicle-stimulating hormone, and total testosterone levels) were the surrogate marker of biochemical recovery. RESULTS Normalization of reproductive hormones was achieved in 48.2% of men. PCT use was associated with faster biochemical recovery (13.0 (IQR8.0-19.0) weeks, PCT; 26.0 (IQR10.5-52) weeks, no-PCT; P < .001). Odds of biochemical recovery during multivariable analysis were: (1) higher with PCT (OR3.80) vs no-PCT (P = .001), in men stopping AAS ≤3 months previously; (2) reduced when 2 (OR0.55), 3 (OR0.46), or 4 (OR0.25) AAS were administered vs 1 drug (P = .009); (3) lower with AAS >6 vs ≤3 months previously (OR0.34, P = .01); (4) higher with last reported AAS >3 months (OR 5.68) vs ≤3 months (P = .001). PCT use was not associated with biochemical recovery in men stopping AAS >3 months previously. CONCLUSION Without evidence-based withdrawal protocols, men commonly try avoiding post-AAS hypogonadism with PCT, which is illicit, ill-defined, and not recommended. Only half of men had complete biochemical testicular recovery after stopping AAS. The surprising association of self-reported PCT use with short-term biochemical recovery from AAS-induced hypogonadism warrants further investigation.
Collapse
Affiliation(s)
- Bonnie Grant
- Section of Investigative Medicine, Imperial College London, Commonwealth Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, United Kingdom
| | - John Campbell
- Glasgow Alcohol & Drug Recovery Services, NHS Greater Glasgow & Clyde, Glasgow G5 8BG, United Kingdom
| | - Anjali Pradeep
- Section of Investigative Medicine, Imperial College London, Commonwealth Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, United Kingdom
| | - Angela D Burns
- Department of Clinical Biochemistry, Queen Elizabeth University Hospital, 1345 Govan Road, Glasgow G51 4TF, United Kingdom
| | - Paul Bassett
- Statsconsultancy Ltd, 40 Longwood Lane, Amersham, Bucks HP7 9EN, United Kingdom
| | - Ali Abbara
- Section of Investigative Medicine, Imperial College London, Commonwealth Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, United Kingdom
| | - Priyadarshi Saket
- Glasgow Alcohol & Drug Recovery Services, NHS Greater Glasgow & Clyde, Glasgow G5 8BG, United Kingdom
| | - Sukhbinder Minhas
- Department of Urology, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, United Kingdom
| | - Waljit S Dhillo
- Section of Investigative Medicine, Imperial College London, Commonwealth Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, United Kingdom
| | - James McVeigh
- Department of Sociology, Manchester Metropolitan University, 4 Rosamund Street West, Manchester M15 6LL, United Kingdom
| | - Shalender Bhasin
- Brigham and Women's Hospital, Division of Endocrinology, Diabetes and Hypertension, 221 Longwood Avenue, Boston, MA 02115, United States
| | - Channa N Jayasena
- Section of Investigative Medicine, Imperial College London, Commonwealth Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, United Kingdom
| |
Collapse
|
24
|
Boyd HM, Frick KM, Kwapis JL. Connecting the Dots: Potential Interactions Between Sex Hormones and the Circadian System During Memory Consolidation. J Biol Rhythms 2023; 38:537-555. [PMID: 37464775 PMCID: PMC10615791 DOI: 10.1177/07487304231184761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Both the circadian clock and sex hormone signaling can strongly influence brain function, yet little is known about how these 2 powerful modulatory systems might interact during complex neural processes like memory consolidation. Individually, the molecular components and action of each of these systems have been fairly well-characterized, but there is a fundamental lack of information about how these systems cooperate. In the circadian system, clock genes function as timekeeping molecules that convey time-of-day information on a well-stereotyped cycle that is governed by the suprachiasmatic nucleus. Keeping time is particularly important to synchronize various physiological processes across the brain and body, including those that regulate memory consolidation. Similarly, sex hormones are powerful modulators of memory, with androgens, estrogens, and progestins, all influencing memory consolidation within memory-relevant brain regions like the hippocampus. Despite clear evidence that each system can influence memory individually, exactly how the circadian and hormonal systems might interact to impact memory consolidation remains unclear. Research investigating either sex hormone action or circadian gene function within memory-relevant brain regions has unveiled several notable places in which the two systems could interact to control memory. Here, we bring attention to known interactions between the circadian clock and sex hormone signaling. We then review sex hormone-mediated control of memory consolidation, highlighting potential nodes through which the circadian system might interact during memory formation. We suggest that the bidirectional relationship between these two systems is essential for proper control of memory formation based on an animal's hormonal and circadian state.
Collapse
Affiliation(s)
- Hannah M. Boyd
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, Pennsylvania
| | - Karyn M. Frick
- Department of Psychology, University of Wisconsin–Milwaukee, Milwaukee, Wisconsin
| | - Janine L. Kwapis
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
25
|
Yamada K, Nagae M, Mano T, Tsuchida H, Hazim S, Goto T, Sanbo M, Hirabayashi M, Inoue N, Uenoyama Y, Tsukamura H. Sex difference in developmental changes in visualized Kiss1 neurons in newly generated Kiss1-Cre rats. J Reprod Dev 2023; 69:227-238. [PMID: 37518187 PMCID: PMC10602768 DOI: 10.1262/jrd.2023-019] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023] Open
Abstract
Hypothalamic kisspeptin neurons are master regulators of mammalian reproduction via direct stimulation of gonadotropin-releasing hormone and consequent gonadotropin release. Here, we generated novel Kiss1 (kisspeptin gene)-Cre rats and investigated the developmental changes and sex differences in visualized Kiss1 neurons of Kiss1-Cre-activated tdTomato reporter rats. First, we validated Kiss1-Cre rats by generating Kiss1-expressing cell-specific Kiss1 knockout (Kiss1-KpKO) rats, which were obtained by crossing the current Kiss1-Cre rats with Kiss1-floxed rats. The resulting male Kiss1-KpKO rats lacked Kiss1 expression in the brain and exhibited hypogonadotropic hypogonadism, similar to the hypogonadal phenotype of global Kiss1 KO rats. Histological analysis of Kiss1 neurons in Kiss1-Cre-activated tdTomato reporter rats revealed that tdTomato signals in the anteroventral periventricular nucleus (AVPV) and arcuate nucleus (ARC) were not affected by estrogen, and that tdTomato signals in the ARC, AVPV, and medial amygdala (MeA) were sexually dimorphic. Notably, neonatal AVPV tdTomato signals were detected only in males, but a larger number of tdTomato-expressing cells were detected in the AVPV and ARC, and a smaller number of cells in the MeA was detected in females than in males at postpuberty. These findings suggest that Kiss1-visualized rats can be used to examine the effect of estrogen feedback mechanisms on Kiss1 expression in the AVPV and ARC. Moreover, the Kiss1-Cre and Kiss1-visualized rats could be valuable tools for further detailed analyses of sexual differentiation in the brain and the physiological role of kisspeptin neurons across the brain in rats.
Collapse
Affiliation(s)
- Koki Yamada
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Mayuko Nagae
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Tetsuya Mano
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Hitomi Tsuchida
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Safiullah Hazim
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Teppei Goto
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
- Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Aichi 444-8787, Japan
| | - Makoto Sanbo
- Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Aichi 444-8787, Japan
| | - Masumi Hirabayashi
- Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Aichi 444-8787, Japan
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| |
Collapse
|
26
|
Dong J, Rees DA. Polycystic ovary syndrome: pathophysiology and therapeutic opportunities. BMJ MEDICINE 2023; 2:e000548. [PMID: 37859784 PMCID: PMC10583117 DOI: 10.1136/bmjmed-2023-000548] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023]
Abstract
Polycystic ovary syndrome is characterised by excessive levels of androgens and ovulatory dysfunction, and is a common endocrine disorder in women of reproductive age. Polycystic ovary syndrome arises as a result of polygenic susceptibility in combination with environmental influences that might include epigenetic alterations and in utero programming. In addition to the well recognised clinical manifestations of hyperandrogenism and ovulatory dysfunction, women with polycystic ovary syndrome have an increased risk of adverse mental health outcomes, pregnancy complications, and cardiometabolic disease. Unlicensed treatments have limited efficacy, mostly because drug development has been hampered by an incomplete understanding of the underlying pathophysiological processes. Advances in genetics, metabolomics, and adipocyte biology have improved our understanding of key changes in neuroendocrine, enteroendocrine, and steroidogenic pathways, including increased gonadotrophin releasing hormone pulsatility, androgen excess, insulin resistance, and changes in the gut microbiome. Many patients with polycystic ovary syndrome have high levels of 11-oxygenated androgens, with high androgenic potency, that might mediate metabolic risk. These advances have prompted the development of new treatments, including those that target the neurokinin-kisspeptin axis upstream of gonadotrophin releasing hormone, with the potential to lessen adverse clinical sequelae and improve patient outcomes.
Collapse
Affiliation(s)
- Jiawen Dong
- Neuroscience and Mental Health Innovation Institute, School of Medicine, Cardiff University, Cardiff, UK
| | - D Aled Rees
- Neuroscience and Mental Health Innovation Institute, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
27
|
Starrett JR, Moenter SM. Hypothalamic kisspeptin neurons as potential mediators of estradiol negative and positive feedback. Peptides 2023; 163:170963. [PMID: 36740189 PMCID: PMC10516609 DOI: 10.1016/j.peptides.2023.170963] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/09/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Gonadal steroid feedback regulates the brain's patterned secretion of gonadotropin-releasing hormone (GnRH). Negative feedback, which occurs in males and during the majority of the female cycle, modulates the amplitude and frequency of GnRH pulses. Positive feedback occurs in females when high estradiol induces a surge pattern of GnRH release. These two forms of feedback and their corresponding patterns of GnRH secretion are thought to be mediated by kisspeptin-expressing neurons in two hypothalamic areas: the arcuate nucleus and the anteroventral periventricular area. In this review, we present evidence for this theory and remaining questions to be addressed.
Collapse
Affiliation(s)
- J Rudolph Starrett
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Suzanne M Moenter
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Obstetrics & Gynecology, University of Michigan, Ann Arbor, MI 48109, USA; The Reproductive Sciences Program, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
28
|
Effects of Group Size on Behavior, Reproduction, and mRNA Expression in Brains of Brandt's Voles. Brain Sci 2023; 13:brainsci13020311. [PMID: 36831854 PMCID: PMC9954483 DOI: 10.3390/brainsci13020311] [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: 11/08/2022] [Revised: 01/24/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
For social animals, a moderate group size is greatly important to maintain their reproductive success. However, the underlying neurobiological mechanism of group size on behavior and reproduction has rarely been investigated. In this study, we examined the effects of group size (1, 2, 4 pairs of adult male and female voles raised per cage) on behavior and reproduction. Meanwhile, the mRNA expression of stress and reproduction response-related genes in male brains was detected. We found that Brandt's voles (Lasiopodomys brandtii) in the large-sized group fight more severely than those in the small-sized group. Meanwhile, male voles were more anxious than females. The average number of embryos and litters per female in the medium-sized group was significantly higher than that of large-sized group. In male voles, stress- or reproduction-response mRNA expressions were more related to final group size or final density due to death caused by fighting. Our results indicated that a moderate group size was beneficial to the reproductive output of Brandt's voles. Our study highlights the combined effects of stress- or reproduction-related gene expression or behavior in regulating the fitness of voles with different group sizes.
Collapse
|
29
|
Hatcher KM, Costanza L, Kauffman AS, Stephens SBZ. The molecular phenotype of kisspeptin neurons in the medial amygdala of female mice. Front Endocrinol (Lausanne) 2023; 14:1093592. [PMID: 36843592 PMCID: PMC9951589 DOI: 10.3389/fendo.2023.1093592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/13/2023] [Indexed: 02/12/2023] Open
Abstract
Reproduction is regulated through the hypothalamic-pituitary-gonadal (HPG) axis, largely via the action of kisspeptin neurons in the hypothalamus. Importantly, Kiss1 neurons have been identified in other brain regions, including the medial amygdala (MeA). Though the MeA is implicated in regulating aspects of both reproductive physiology and behavior, as well as non-reproductive processes, the functional roles of MeA Kiss1 neurons are largely unknown. Additionally, besides their stimulation by estrogen, little is known about how MeA Kiss1 neurons are regulated. Using a RiboTag mouse model in conjunction with RNA-seq, we examined the molecular profile of MeA Kiss1 neurons to identify transcripts that are co-expressed in MeA Kiss1 neurons of female mice and whether these transcripts are modulated by estradiol (E2) treatment. RNA-seq identified >13,800 gene transcripts co-expressed in female MeA Kiss1 neurons, including genes for neuropeptides and receptors implicated in reproduction, metabolism, and other neuroendocrine functions. Of the >13,800 genes co-expressed in MeA Kiss1 neurons, only 45 genes demonstrated significantly different expression levels due to E2 treatment. Gene transcripts such as Kiss1, Gal, and Oxtr increased in response to E2 treatment, while fewer transcripts, such as Esr1 and Cyp26b1, were downregulated by E2. Dual RNAscope and immunohistochemistry was performed to validate co-expression of MeA Kiss1 with Cck and Cartpt. These results are the first to establish a profile of genes actively expressed by MeA Kiss1 neurons, including a subset of genes regulated by E2, which provides a useful foundation for future investigations into the regulation and function of MeA Kiss1 neurons.
Collapse
Affiliation(s)
- Katherine M. Hatcher
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Leah Costanza
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Alexander S. Kauffman
- Department of OBGYN and Reproductive Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Shannon B. Z. Stephens
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
- Department of OBGYN and Reproductive Sciences, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
30
|
Xu G, Zhao X, Li Z, Hu J, Li X, Li J, Chen Y. Effects of electroacupuncture on the kisspeptin-gonadotropin-releasing hormone (GnRH) /luteinizing hormone (LH) neural circuit abnormalities and androgen receptor expression of kisspeptin/neurokinin B/dynorphin neurons in PCOS rats. J Ovarian Res 2023; 16:15. [PMID: 36650561 PMCID: PMC9844031 DOI: 10.1186/s13048-022-01078-x] [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: 08/24/2022] [Accepted: 12/13/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is characterized by hyperandrogenism, anovulation, and polycystic ovaries. Electroacupuncture (EA) can effectively improve hyperandrogenism and increase ovulation frequency in patients with PCOS. Pieces of suggest that androgen activity in the brain is associated with impaired steroid negative feedback in such patients. Studies have shown that EA regulated androgen receptor (AR) expression and local factor levels (such as anti-Müllerian hormone and inhibin B) in the ovary of PCOS rats. However, few studies have explored the effect of EA on androgen activity in the brain. OBJECTIVE This study investigated the effect of EA on the kisspeptin-gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) neural circuit and sex hormone receptor expression in the hypothalamus of PCOS rats. METHODS PCOS signs were induced by letrozole administration, and the induced rats were treated with low-frequency EA at Guan Yuan acupoint (CV4). The effect of EA on PCOS-like signs was evaluated by observing changes in the body weight, ovarian quality, ovarian morphology, and serum sex hormone levels in rats. To explore the mechanism of the effect of EA on PCOS-like signs, the neuropeptide content of the kisspeptin-GnRH/LH neural circuit was assessed using enzyme-linked immunosorbent assay(ELISA); AR and estrogen receptor α (ERα) coexpression on kisspeptin/neurokinin B/dynorphin (KNDy) neurons was determined via triple-label immunofluorescence; and protein and mRNA expression of Kiss1, Ar, Esr1, and kisspeptin receptor (Kiss1r) was evaluated via western blotting and Reverse Transcription-Polymerase Chain Reaction (RT-PCR). RESULTS The results revealed that the estrous cycle of rats in the EA treatment group recovered, and their body and ovary weight reduced; ovarian morphology improved; serum testosterone and LH levels significantly decreased; and kisspeptin, GnRH, and dynorphin levels in hypothalamic arcuate nucleus significantly decreased. Compared with controls, the number of AR/Kiss1-positive cells increased, number of ERα/Kiss1-positive cells decreased, and protein and mRNA expression of Kiss1, Ar, and Kiss1r significantly increased in PCOS rats. However, EA treatment reversed these changes and reduced the expression of Kiss1, Ar, and Kiss1r significantly. CONCLUSION Improvement in the reproductive hallmarks of PCOS rats via EA may be achieved by regulating the kisspeptin-GnRH/LH circuit via androgen activity attenuation. Thus, the results provide an experimental basis for acupuncture as an adjuvant medical therapy on PCOS.
Collapse
Affiliation(s)
- Ge Xu
- grid.411480.80000 0004 1799 1816Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China ,grid.412540.60000 0001 2372 7462Shanghai University of Traditional Chinese Medicine, Shanghai, 201210 China
| | - Xuedan Zhao
- grid.411480.80000 0004 1799 1816Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China ,grid.412540.60000 0001 2372 7462Shanghai University of Traditional Chinese Medicine, Shanghai, 201210 China
| | - Zhihao Li
- grid.411480.80000 0004 1799 1816Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
| | - Junwei Hu
- grid.411480.80000 0004 1799 1816Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
| | - Xu Li
- grid.411480.80000 0004 1799 1816Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
| | - Juanjuan Li
- grid.411480.80000 0004 1799 1816Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
| | - Yuelai Chen
- grid.411480.80000 0004 1799 1816Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
| |
Collapse
|
31
|
King'uyu DN, Stephens SBZ, Kopec AM. Immune signaling in sex-specific neural and behavioral development: Adolescent opportunity. Curr Opin Neurobiol 2022; 77:102647. [PMID: 36332416 PMCID: PMC9893405 DOI: 10.1016/j.conb.2022.102647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
Sex differences in neural and behavioral development are integral to understanding neurodevelopmental, mental health, and neurodegenerative disorders. Much of the literature has focused on late prenatal and early postnatal life as a critical juncture for establishing sex-specific developmental trajectories, and data are now clear that immune signaling plays a central role in establishing sex differences early in life. Adolescence is another developmental period during which sex differences arise. However, we know far less about how immune signaling plays a role in establishing sex differences during adolescence. Herein, we review well-defined examples of sex differences during adolescence and then survey the literature to speculate how immune signaling might be playing a role in defining sex-specific adolescent outcomes. We discuss open questions in the literature and propose experimental design tenets that may assist in better understanding adolescent neurodevelopment.
Collapse
Affiliation(s)
- David N King'uyu
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, USA
| | - Shannon B Z Stephens
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, USA. https://twitter.com/Stephens_Lab
| | - Ashley M Kopec
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, USA.
| |
Collapse
|
32
|
Constantin S. Targeting KNDy neurons to control GnRH pulses. Curr Opin Pharmacol 2022; 67:102316. [PMID: 36347163 PMCID: PMC9772270 DOI: 10.1016/j.coph.2022.102316] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is the final output of the central nervous system that drives fertility. A characteristic of GnRH secretion is its pulsatility, which is driven by a pulse generator. Each GnRH pulse triggers a luteinizing hormone (LH) pulse. However, the puzzle has been to reconcile the synchronicity of GnRH neurons with the scattered hypothalamic distribution of their cell bodies. A leap toward understanding GnRH pulses was the discovery of kisspeptin neurons near the distal processes of GnRH neurons, which secrete kisspeptins, potent excitatory neuropeptides on GnRH neurons, and equipped with dual, but opposite, self-modulatory neuropeptides, neurokinin B and dynorphin. Over the last decade, this cell-to-cell communication has been dissected in animal models. Today the 50-year quest for the basic mechanism of GnRH pulse generation may be over, but questions about its physiological tuning remain. Here is an overview of recent basic research that frames translational research.
Collapse
Affiliation(s)
- Stephanie Constantin
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
33
|
Bulut F, Kacar E, Bilgin B, Hekim MG, Keleştemur MM, Sahin Z, Ayar A, Ozcan M. Crosstalk between kisspeptin and gonadotropin-inhibitory hormone in the silence of puberty: preclinical evidence from a calcium signaling study. J Recept Signal Transduct Res 2022; 42:608-613. [PMID: 36137227 DOI: 10.1080/10799893.2022.2125014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Kisspeptin and gonadotropin-inhibitory hormone (GnIH) are among suggested neuroendocrine modulators of reproductive function. Intracellular calcium signaling is a critical component in the regulation of a variety of physiological and pathological processes including neurotransmitter release, and, therefore, can be used as signaling indicator for investigating the involvement of kisspeptin, GnIH, and gonadotropin-releasing hormone (GnRH) release. Hence, this study investigated the effects of kisspeptin and GnIH on calcium signaling using immortalized hypothalamic cells (rHypoE-8) as a model. Kisspeptin neurons were loaded with the ratiometric calcium dye (Fura-2 AM, 1 μmol) and intracellular free calcium ([Ca2+]i) responses were quantified using digital fluorescence imaging system. Kisspeptin-10 (100, 300, and 1000 nM) caused a significant increase in [Ca2+]i in rHypoE-8 cells (n = 58, n = 64, and n = 49, respectively, p < 0.001). The kisspeptin receptor antagonist, P234, inhibited the calcium responses to kisspeptin (p < 0.001, n = 32). GnIH (100 and 1000 nM), alone, did not cause any significant change in the mean basal [Ca2+]i levels in kisspeptin cells, but GnIH attenuated the kisspeptin-evoked [Ca2+]i transients (n = 47, p < 0.001). This novel findings of [Ca2+]i signaling in in vitro setting implicate that kisspeptin and GnIH may exert their effects on hypothalamus-pituitary-gonadal (HPG) axis by modulating kisspeptin neurons. These results also implicate that kisspeptin neurons may have an autocrine regulation.
Collapse
Affiliation(s)
- Ferah Bulut
- Department of Biophysics, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Emine Kacar
- Department of Physiology, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Batuhan Bilgin
- Department of Biophysics, Faculty of Medicine, Firat University, Elazig, Turkey
| | | | | | - Zafer Sahin
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Ahmet Ayar
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Mete Ozcan
- Department of Biophysics, Faculty of Medicine, Firat University, Elazig, Turkey
| |
Collapse
|
34
|
Kim HD, Kim YJ, Jang M, Bae SG, Yun SH, Lee MR, Seo YR, Cho JK, Kim SJ, Lee WJ. Heat Stress during Summer Attenuates Expression of the Hypothalamic Kisspeptin, an Upstream Regulator of the Hypothalamic-Pituitary-Gonadal Axis, in Domestic Sows. Animals (Basel) 2022; 12:2967. [PMID: 36359090 PMCID: PMC9657376 DOI: 10.3390/ani12212967] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 11/16/2023] Open
Abstract
The release of reproductive hormones in the hypothalamic-pituitary-gonadal (HPG) axis is regulated by its upstream regulator, kisspeptin, and influenced by external stresses, including heat stress. Since the effect of heat stress (summer infertility) on hypothalamic kisspeptin expression in domestic sows is not yet understood, the present study attempted to identify changes in kisspeptin expression in different seasons (summer and spring). The high atmospheric temperature in summer decreased the pregnancy rate and litter size and increased stress-related hormones as a chronic stressor to domestic sows. The hypothalamic kisspeptin expression in summer was decreased regardless of the estrus phase and negatively correlated with atmospheric temperature, indicating that high temperature decreased kisspeptin. When the activity of hypothalamic kisspeptin neurons in the follicular phase was assessed using c-Fos staining, a decreased number of kisspeptin neurons coexpressing c-Fos was observed in domestic sows in summer. Accordingly, lower expression of kisspeptin induced decreased levels of HPG axis-related reproductive hormones, such as gonadotropins and estrogen, and fewer large ovarian follicles. In conclusion, the present study demonstrated that reduced kisspeptin expression and its neuronal activity in the hypothalamus under heat stress in summer induced downregulation of the HPG axis and caused summer infertility in domestic sows.
Collapse
Affiliation(s)
- Hwan-Deuk Kim
- College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
- Department of Veterinary Research, Daegu Metropolitan City Institute of Health & Environment, Daegu 42183, Korea
| | - Young-Jong Kim
- College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Min Jang
- College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Seul-Gi Bae
- College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Sung-Ho Yun
- College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Mi-Ree Lee
- Department of Veterinary Research, Daegu Metropolitan City Institute of Health & Environment, Daegu 42183, Korea
| | - Yong-Ryul Seo
- Department of Veterinary Research, Daegu Metropolitan City Institute of Health & Environment, Daegu 42183, Korea
| | - Jae-Keun Cho
- Department of Veterinary Research, Daegu Metropolitan City Institute of Health & Environment, Daegu 42183, Korea
| | - Seung-Joon Kim
- College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
- Institute of Equine Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Won-Jae Lee
- College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
- Institute of Equine Medicine, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
35
|
Moeller JS, Bever SR, Finn SL, Phumsatitpong C, Browne MF, Kriegsfeld LJ. Circadian Regulation of Hormonal Timing and the Pathophysiology of Circadian Dysregulation. Compr Physiol 2022; 12:4185-4214. [PMID: 36073751 DOI: 10.1002/cphy.c220018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Circadian rhythms are endogenously generated, daily patterns of behavior and physiology that are essential for optimal health and disease prevention. Disruptions to circadian timing are associated with a host of maladies, including metabolic disease and obesity, diabetes, heart disease, cancer, and mental health disturbances. The circadian timing system is hierarchically organized, with a master circadian clock located in the suprachiasmatic nucleus (SCN) of the anterior hypothalamus and subordinate clocks throughout the CNS and periphery. The SCN receives light information via a direct retinal pathway, synchronizing the master clock to environmental time. At the cellular level, circadian rhythms are ubiquitous, with rhythms generated by interlocking, autoregulatory transcription-translation feedback loops. At the level of the SCN, tight cellular coupling maintains rhythms even in the absence of environmental input. The SCN, in turn, communicates timing information via the autonomic nervous system and hormonal signaling. This signaling couples individual cellular oscillators at the tissue level in extra-SCN brain loci and the periphery and synchronizes subordinate clocks to external time. In the modern world, circadian disruption is widespread due to limited exposure to sunlight during the day, exposure to artificial light at night, and widespread use of light-emitting electronic devices, likely contributing to an increase in the prevalence, and the progression, of a host of disease states. The present overview focuses on the circadian control of endocrine secretions, the significance of rhythms within key endocrine axes for typical, homeostatic functioning, and implications for health and disease when dysregulated. © 2022 American Physiological Society. Compr Physiol 12: 1-30, 2022.
Collapse
Affiliation(s)
- Jacob S Moeller
- Graduate Group in Endocrinology, University of California, Berkeley, California, USA
| | - Savannah R Bever
- Department of Psychology, University of California, Berkeley, California, USA
| | - Samantha L Finn
- Department of Psychology, University of California, Berkeley, California, USA
| | | | - Madison F Browne
- Department of Psychology, University of California, Berkeley, California, USA
| | - Lance J Kriegsfeld
- Graduate Group in Endocrinology, University of California, Berkeley, California, USA.,Department of Psychology, University of California, Berkeley, California, USA.,Department of Integrative Biology, University of California, Berkeley, California, USA.,The Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA
| |
Collapse
|
36
|
Socs3 ablation in kisspeptin cells partially prevents lipopolysaccharide-induced body weight loss. Cytokine 2022; 158:155999. [PMID: 35985175 DOI: 10.1016/j.cyto.2022.155999] [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: 03/15/2022] [Revised: 07/25/2022] [Accepted: 08/02/2022] [Indexed: 11/22/2022]
Abstract
Many cytokines have been proposed to regulate reproduction due to their actions on hypothalamic kisspeptin cells, the main modulators of gonadotropin-releasing hormone (GnRH) neurons. Hormones such as leptin, prolactin and growth hormone are good examples of cytokines that lead to Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway activation, consequently exerting effects in kisspeptin neurons. Different studies have investigated how specific components of the JAK/STAT signaling pathway affect the functions of kisspeptin cells, but the role of the suppressor of cytokine signaling 3 (SOCS3) in mediating cytokine actions in kisspeptin cells remains unknown. Cre-Loxp technology was used in the present study to ablate Socs3 expression in kisspeptin cells (Kiss1/Socs3-KO). Then, male and female control and Kiss1/Socs3-KO mice were evaluated for sexual maturation, energy homeostasis features, and fertility. It was found that hypothalamic Kiss1 mRNA expression is significantly downregulated in Kiss1/Socs3-KO mice. Despite reduced hypothalamic Kiss1 mRNA content, these mice did not present any sexual maturation or fertility impairments. Additionally, body weight gain, leptin sensitivity and glucose homeostasis were similar to control mice. Interestingly, Kiss1/Socs3-KO mice were partially protected against lipopolysaccharide (LPS)-induced body weight loss. Our results suggest that Socs3 ablation in kisspeptin cells partially prevents the sickness behavior induced by LPS, suggesting that kisspeptin cells can modulate energy metabolism in mice in certain situations.
Collapse
|
37
|
Kauffman AS. Neuroendocrine mechanisms underlying estrogen positive feedback and the LH surge. Front Neurosci 2022; 16:953252. [PMID: 35968365 PMCID: PMC9364933 DOI: 10.3389/fnins.2022.953252] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/08/2022] [Indexed: 01/26/2023] Open
Abstract
A fundamental principle in reproductive neuroendocrinology is sex steroid feedback: steroid hormones secreted by the gonads circulate back to the brain to regulate the neural circuits governing the reproductive neuroendocrine axis. These regulatory feedback loops ultimately act to modulate gonadotropin-releasing hormone (GnRH) secretion, thereby affecting gonadotropin secretion from the anterior pituitary. In females, rising estradiol (E2) during the middle of the menstrual (or estrous) cycle paradoxically "switch" from being inhibitory on GnRH secretion ("negative feedback") to stimulating GnRH release ("positive feedback"), resulting in a surge in GnRH secretion and a downstream LH surge that triggers ovulation. While upstream neural afferents of GnRH neurons, including kisspeptin neurons in the rostral hypothalamus, are proposed as critical loci of E2 feedback action, the underlying mechanisms governing the shift between E2 negative and positive feedback are still poorly understood. Indeed, the precise cell targets, neural signaling factors and receptors, hormonal pathways, and molecular mechanisms by which ovarian-derived E2 indirectly stimulates GnRH surge secretion remain incompletely known. In many species, there is also a circadian component to the LH surge, restricting its occurrence to specific times of day, but how the circadian clock interacts with endocrine signals to ultimately time LH surge generation also remains a major gap in knowledge. Here, we focus on classic and recent data from rodent models and discuss the consensus knowledge of the neural players, including kisspeptin, the suprachiasmatic nucleus, and glia, as well as endocrine players, including estradiol and progesterone, in the complex regulation and generation of E2-induced LH surges in females.
Collapse
|
38
|
Jamieson BB, Piet R. Kisspeptin neuron electrophysiology: Intrinsic properties, hormonal modulation, and regulation of homeostatic circuits. Front Neuroendocrinol 2022; 66:101006. [PMID: 35640722 DOI: 10.1016/j.yfrne.2022.101006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 05/05/2022] [Accepted: 05/19/2022] [Indexed: 11/04/2022]
Abstract
The obligatory role of kisspeptin (KISS1) and its receptor (KISS1R) in regulating the hypothalamic-pituitary-gonadal axis, puberty and fertility was uncovered in 2003. In the few years that followed, an impressive body of work undertaken in many species established that neurons producing kisspeptin orchestrate gonadotropin-releasing hormone (GnRH) neuron activity and subsequent GnRH and gonadotropin hormone secretory patterns, through kisspeptin-KISS1R signaling, and mediate many aspects of gonadal steroid hormone feedback regulation of GnRH neurons. Here, we review knowledge accrued over the past decade, mainly in genetically modified mouse models, of the electrophysiological properties of kisspeptin neurons and their regulation by hormonal feedback. We also discuss recent progress in our understanding of the role of these cells within neuronal circuits that control GnRH neuron activity and GnRH secretion, energy balance and, potentially, other homeostatic and reproductive functions.
Collapse
Affiliation(s)
| | - Richard Piet
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, USA.
| |
Collapse
|
39
|
Moralia MA, Quignon C, Simonneaux M, Simonneaux V. Environmental disruption of reproductive rhythms. Front Neuroendocrinol 2022; 66:100990. [PMID: 35227765 DOI: 10.1016/j.yfrne.2022.100990] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/06/2022] [Accepted: 02/21/2022] [Indexed: 12/17/2022]
Abstract
Reproduction is a key biological function requiring a precise synchronization with annual and daily cues to cope with environmental fluctuations. Therefore, humans and animals have developed well-conserved photoneuroendocrine pathways to integrate and process daily and seasonal light signals within the hypothalamic-pituitary-gonadal axis. However, in the past century, industrialization and the modern 24/7 human lifestyle have imposed detrimental changes in natural habitats and rhythms of life. Indeed, exposure to an excessive amount of artificial light at inappropriate timing because of shift work and nocturnal urban lighting, as well as the ubiquitous environmental contamination by endocrine-disrupting chemicals, threaten the integrity of the daily and seasonal timing of biological functions. Here, we review recent epidemiological, field and experimental studies to discuss how light and chemical pollution of the environment can disrupt reproductive rhythms by interfering with the photoneuroendocrine timing system.
Collapse
Affiliation(s)
- Marie-Azélie Moralia
- Université de Strasbourg, Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Clarisse Quignon
- Université de Strasbourg, Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Marine Simonneaux
- Université de Strasbourg, Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Valérie Simonneaux
- Université de Strasbourg, Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France.
| |
Collapse
|
40
|
Sex-specific hypothalamic expression of kisspeptin, gonadotropin releasing hormone, and kisspeptin receptor in progressive demyelination model. J Chem Neuroanat 2022; 123:102120. [PMID: 35718292 DOI: 10.1016/j.jchemneu.2022.102120] [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: 03/25/2022] [Revised: 05/24/2022] [Accepted: 06/13/2022] [Indexed: 11/21/2022]
Abstract
Demyelinating diseases, such as multiple sclerosis, decrease the quality of life of patients and can affect reproduction. Assisted reproductive therapies are available, which although effective, aggravate motor symptoms. For this reason, it is important to determine how the control of the hypothalamus-pituitary-gonadal axis is affected in order to develop better strategies for these patients. One way to determine this is using animal models such as the taiep rat, which shows progressive demyelination of the central nervous system, and was used in the present study to characterize the expression of gonadotrophin-releasing hormone (GnRH), Kisspeptin, and kisspeptin receptor (Kiss1R) and luteinizing hormone (LH) secretion. The expression of kisspeptin, GnRH, and Kiss1R was determined at the hypothalamic level by immunofluorescence and serum LH levels were determined by ELISA. The expression of kisspeptin at the hypothalamic level showed sexual dimorphism, where there was an increase in males and a decrease in females during oestrus. There was no change in the expression of GnRH or kisspeptin receptor, regardless of sex. However, a decrease in serum LH concentration was observed in both sexes. The taiep rat showed changes in the expression of kisspeptin at the hypothalamic level. These changes are different from those reported in the literature with the use of animals with experimental allergic encephalomyelitis, this is because both animal models represent different degrees of progression of multiple sclerosis. Our results suggest that the effects on the hypothalamus-pituitary-gonadal axis depend on the differences between the demyelinating processes, their progression, and even individual factors, and it is thus important that fertility treatments are individualized to maximize therapeutic effects.
Collapse
|
41
|
Vigil P, Meléndez J, Soto H, Petkovic G, Bernal YA, Molina S. Chronic Stress and Ovulatory Dysfunction: Implications in Times of COVID-19. Front Glob Womens Health 2022; 3:866104. [PMID: 35677754 PMCID: PMC9168655 DOI: 10.3389/fgwh.2022.866104] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/04/2022] [Indexed: 12/11/2022] Open
Abstract
Stress is known to be associated with adverse health outcomes. The COVID-19 pandemic and its associated lockdowns are examples of chronic stressors. Lockdown measures inadvertently caused significant psychological distress and became a powerful source of anxiety/stress, sleep disturbances, nutritional changes and weight gain. Stress is known to impact women's health specifically, through hypothalamic-pituitary-gonadal (HPG) axis dysfunction and resultant ovulatory dysfunction. Such dysfunction may manifest in menstrual irregularities and/or infertility due to hypothalamic hypogonadism. Here, we review the key physiological mediators of stress and associated ovulatory dysfunction. The kisspeptinergic system is comprised of sets of neurons located in the hypothalamus, the rostral periventricular region of the third ventricle (RP3V) and the arcuate nucleus (ARC). This system links nutrition, reproductive signals and stress. It plays a key role in the function of the HPG axis. During chronic stress, the kisspeptinergic system affects the HPG axis, GnRH pulsatility, and, therefore, ovulation. Leptin, insulin and corticotrophin-releasing hormone (CRH) are thought to be additional key modulators in the behavioral responses to chronic stress and may contribute to stress-related ovulatory dysfunction. This mini-review also summarizes and appraises the available evidence on the negative impact of chronic stress as a result of the COVID-19 pandemic lockdowns. It proposes physiological mechanisms to explain the observed effects on women's reproductive health and well-being. The review suggests areas for future research.
Collapse
Affiliation(s)
- Pilar Vigil
- Reproductive Health Research Institute (RHRI), Santiago, Chile
- Fundación Médica San Cristóbal, Santiago, Chile
- *Correspondence: Pilar Vigil
| | - Jaime Meléndez
- Reproductive Health Research Institute (RHRI), Santiago, Chile
| | - Hugo Soto
- Reproductive Health Research Institute (RHRI), Santiago, Chile
- Facultad de Medicina, Universidad San Sebastián, Santiago, Chile
| | | | - Yanara A. Bernal
- Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Santiago Molina
- Tallahassee Community College, Tallahassee, FL, United States
| |
Collapse
|
42
|
Sheep as a model for neuroendocrinology research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:1-34. [PMID: 35595346 DOI: 10.1016/bs.pmbts.2022.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Animal models remain essential to understand the fundamental mechanisms of physiology and pathology. Particularly, the complex and dynamic nature of neuroendocrine cells of the hypothalamus make them difficult to study. The neuroendocrine systems of the hypothalamus are critical for survival and reproduction, and are highly conserved throughout vertebrate evolution. Their roles in controlling body metabolism, growth and body composition, stress, electrolyte balance, and reproduction, have been intensively studied, and have yielded groundbreaking discoveries. Many of these discoveries would not have been feasible without the use of the domestic sheep (Ovis aries). The sheep has been used for decades to study the neuroendocrine systems of the hypothalamus and has become a model for human neuroendocrinology. The aim of this chapter is to review some of the profound biomedical discoveries made possible by the use of sheep. The advantages and limitations of sheep as a neuroendocrine model will be discussed. While no animal model can perfectly recapitulate a human disease or condition, sheep are invaluable for enabling manipulations not possible in human subjects and isolating physiologic variables to garner insight into neuroendocrinology and associated pathologies.
Collapse
|
43
|
Delcour C, Khawaja N, Gonzalez-Duque S, Lebon S, Talbi A, Drira L, Chevenne D, Ajlouni K, de Roux N. Estrogen Receptor α Inactivation in 2 Sisters: Different Phenotypic Severities for the Same Pathogenic Variant. J Clin Endocrinol Metab 2022; 107:e2553-e2562. [PMID: 35134944 DOI: 10.1210/clinem/dgac065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Estrogens play an essential role in reproduction. Their action is mediated by nuclear α and β receptors (ER) and by membrane receptors. Only 3 females and 2 males, from 3 families, with a loss of ERα function have been reported to date. OBJECTIVE We describe here a new family, in which 2 sisters display endocrine and ovarian defects of different severities despite carrying the same homozygous rare variant of ESR1. METHODS A 36-year-old woman from a consanguineous Jordanian family presented with primary amenorrhea and no breast development, with high plasma levels of 17β-estradiol (E2), follicle-stimulating hormone and luteinizing hormone, and enlarged multifollicular ovaries, strongly suggesting estrogen resistance. Her 18-year-old sister did not enter puberty and had moderately high levels of E2, high plasma gonadotropin levels, and normal ovaries. RESULTS Genetic analysis identified a homozygous variant of ESR1 leading to the replacement of a highly conserved glutamic acid with a valine (ERα-E385V). The transient expression of ERα-E385V in HEK293A and MDA-MB231 cells revealed highly impaired ERE-dependent transcriptional activation by E2. The analysis of the KISS1 promoter activity revealed that the E385V substitution induced a ligand independent activation of ERα. Immunofluorescence analysis showed that less ERα-E385V than ERα-WT was translocated into the nucleus in the presence of E2. CONCLUSION These 2 new cases are remarkable given the difference in the severity of their ovarian and hormonal phenotypes. This phenotypic discrepancy may be due to a mechanism partially compensating for the ERα loss of function.
Collapse
Affiliation(s)
- Clémence Delcour
- Université de Paris, INSERM UMR 1141 NeuroDiderot, 75019 Paris, France
| | - Nahla Khawaja
- National Center for Diabetes, Endocrinology and Genetics, Amman 11942, Jordan
| | - Sergio Gonzalez-Duque
- Biochemistry-Hormonology Laboratory, AP-HP, Robert Debré Hospital, 75019 Paris, France
| | - Sophie Lebon
- Université de Paris, INSERM UMR 1141 NeuroDiderot, 75019 Paris, France
| | - Abir Talbi
- Biochemistry-Hormonology Laboratory, AP-HP, Robert Debré Hospital, 75019 Paris, France
| | - Leila Drira
- Biochemistry-Hormonology Laboratory, AP-HP, Robert Debré Hospital, 75019 Paris, France
| | - Didier Chevenne
- Biochemistry-Hormonology Laboratory, AP-HP, Robert Debré Hospital, 75019 Paris, France
| | - Kamel Ajlouni
- National Center for Diabetes, Endocrinology and Genetics, Amman 11942, Jordan
| | - Nicolas de Roux
- Université de Paris, INSERM UMR 1141 NeuroDiderot, 75019 Paris, France
- Biochemistry-Hormonology Laboratory, AP-HP, Robert Debré Hospital, 75019 Paris, France
| |
Collapse
|
44
|
Goodman RL, Herbison AE, Lehman MN, Navarro VM. Neuroendocrine control of gonadotropin-releasing hormone: Pulsatile and surge modes of secretion. J Neuroendocrinol 2022; 34:e13094. [PMID: 35107859 PMCID: PMC9948945 DOI: 10.1111/jne.13094] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 11/28/2022]
Abstract
The concept that different systems control episodic and surge secretion of gonadotropin-releasing hormone (GnRH) was well established by the time that GnRH was identified and formed the framework for studies of the physiological roles of GnRH, and later kisspeptin. Here, we focus on recent studies identifying the neural mechanisms underlying these two modes of secretion, with an emphasis on their core components. There is now compelling data that kisspeptin neurons in the arcuate nucleus that also contain neurokinin B (NKB) and dynorphin (i.e., KNDy cells) and their projections to GnRH dendrons constitute the GnRH pulse generator in mice and rats. There is also strong evidence for a similar role for KNDy neurons in sheep and goats, and weaker data in monkeys and humans. However, whether KNDy neurons act on GnRH dendrons and/or GnRH soma and dendrites that are found in the mediobasal hypothalamus (MBH) of these species remains unclear. The core components of the GnRH/luteinising hormone surge consist of an endocrine signal that initiates the process and a neural trigger that drives GnRH secretion during the surge. In all spontaneous ovulators, the core endocrine signal is a rise in estradiol secretion from the maturing follicle(s), with the site of estrogen positive feedback being the rostral periventricular kisspeptin neurons in rodents and neurons in the MBH of sheep and primates. There is considerable species variations in the neural trigger, with three major classes. First, in reflex ovulators, this trigger is initiated by coitus and carried to the hypothalamus by neural or vascular pathways. Second, in rodents, there is a time of day signal that originates in the suprachiasmatic nucleus and activates rostral periventricular kisspeptin neurons and GnRH soma and dendrites. Finally, in sheep nitric oxide-producing neurons in the ventromedial nucleus, KNDy neurons and rostral kisspeptin neurons all appear to participate in driving GnRH release during the surge.
Collapse
Affiliation(s)
- Robert L. Goodman
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Allan E. Herbison
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Michael N. Lehman
- Brain Health Research Institute, Kent State University, Kent, OH, USA
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Victor M. Navarro
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School and Department of Medicine, Boston, MA, USA
| |
Collapse
|
45
|
Dardente H, Simonneaux V. GnRH and the photoperiodic control of seasonal reproduction: Delegating the task to kisspeptin and RFRP-3. J Neuroendocrinol 2022; 34:e13124. [PMID: 35384117 DOI: 10.1111/jne.13124] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/22/2022] [Accepted: 03/03/2022] [Indexed: 10/18/2022]
Abstract
Synchronization of mammalian breeding activity to the annual change of photoperiod and environmental conditions is of the utmost importance for individual survival and species perpetuation. Subsequent to the early 1960s, when the central role of melatonin in this adaptive process was demonstrated, our comprehension of the mechanisms through which light regulates gonadal activity has increased considerably. The current model for the photoperiodic neuroendocrine system points to pivotal roles for the melatonin-sensitive pars tuberalis (PT) and its seasonally-regulated production of thyroid-stimulating hormone (TSH), as well as for TSH-sensitive hypothalamic tanycytes, radial glia-like cells located in the basal part of the third ventricle. Tanycytes respond to TSH through increased expression of thyroid hormone (TH) deiodinase 2 (Dio2), which leads to heightened production of intrahypothalamic triiodothyronine (T3) during longer days of spring and summer. There is strong evidence that this local, long-day driven, increase in T3 links melatonin input at the PT to gonadotropin-releasing hormone (GnRH) output, to align breeding with the seasons. The mechanism(s) through which T3 impinges upon GnRH remain(s) unclear. However, two distinct neuronal populations of the medio-basal hypothalamus, which express the (Arg)(Phe)-amide peptides kisspeptin and RFamide-related peptide-3, appear to be well-positioned to relay this seasonal T3 message towards GnRH neurons. Here, we summarize our current understanding of the cellular, molecular and neuroendocrine players, which keep track of photoperiod and ultimately govern GnRH output and seasonal breeding.
Collapse
Affiliation(s)
- Hugues Dardente
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Valérie Simonneaux
- Institute for Cellular and Integrative Neuroscience, University of Strasbourg, Strasbourg, France
| |
Collapse
|
46
|
Lavalle SN, Chou T, Hernandez J, Naing NCP, He MY, Tonsfeldt KJ, Mellon PL. Deletion of the homeodomain gene Six3 from kisspeptin neurons causes subfertility in female mice. Mol Cell Endocrinol 2022; 546:111577. [PMID: 35121076 PMCID: PMC8934285 DOI: 10.1016/j.mce.2022.111577] [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: 10/27/2021] [Revised: 01/13/2022] [Accepted: 01/30/2022] [Indexed: 01/27/2023]
Abstract
The homeodomain transcription factor SIX3 is a known regulator of eye, nose, and forebrain development, and has recently been implicated in female reproduction. Germline heterozygosity of SIX3 is sufficient to cause subfertility, but the cell populations that mediate this role are unknown. The neuropeptide kisspeptin is a critical component of the reproductive axis and plays roles in sexual maturation, ovulation, and the maintenance of gonadotropin secretion. We used Cre-Lox technology to remove Six3 specifically from kisspeptin neurons in mice to test the hypothesis that SIX3 in kisspeptin neurons is required for reproduction. We found that loss of Six3 in kisspeptin neurons causes subfertility and estrous cycle irregularities in females, but no effect in males. Overall, we find that SIX3 expression in kisspeptin neurons is an important contributor to female fertility.
Collapse
Affiliation(s)
- Shanna N Lavalle
- Department of Obstetrics, Gynecology, And Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Teresa Chou
- Department of Obstetrics, Gynecology, And Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Jacqueline Hernandez
- Department of Obstetrics, Gynecology, And Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Nay Chi P Naing
- Department of Obstetrics, Gynecology, And Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Michelle Y He
- Department of Obstetrics, Gynecology, And Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Karen J Tonsfeldt
- Department of Obstetrics, Gynecology, And Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Pamela L Mellon
- Department of Obstetrics, Gynecology, And Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
47
|
Coyle CS, Prescott M, Handelsman DJ, Walters KA, Campbell RE. Chronic androgen excess in female mice does not impact luteinizing hormone pulse frequency or putative GABAergic inputs to GnRH neurons. J Neuroendocrinol 2022; 34:e13110. [PMID: 35267218 PMCID: PMC9286661 DOI: 10.1111/jne.13110] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/17/2022] [Accepted: 02/17/2022] [Indexed: 11/28/2022]
Abstract
Polycystic ovary syndrome (PCOS) is associated with androgen excess and, frequently, hyperactive pulsatile luteinizing hormone (LH) secretion. Although the origins of PCOS are unclear, evidence from pre-clinical models implicates androgen signalling in the brain in the development of PCOS pathophysiology. Chronic exposure of female mice to dihydrotestosterone (DHT) from 3 weeks of age drives both reproductive and metabolic impairments that are ameliorated by selective androgen receptor (AR) loss from the brain. This suggests centrally driven mechanisms in hyperandrogen-mediated PCOS-like pathophysiology that remain to be defined. Acute prenatal DHT exposure can also model the hyperandrogenism of PCOS, and this is accompanied by increased LH pulse frequency and increased GABAergic innervation of gonadotrophin-releasing hormone (GnRH) neurons. We aimed to determine the impact of chronic exposure of female mice to DHT, which models the hyperandrogenism of PCOS, on pulsatile LH secretion and putative GABAergic input to GnRH neurons. To do this, GnRH-green fluorescent protein (GFP) female mice received either DHT or blank capsules for 90 days from postnatal day 21 (n = 6 or 7 per group). Serial tail-tip blood sampling was used to measure LH dynamics and perfusion-fixed brains were collected and immunolabelled for vesicular GABA transporter (VGAT) to assess putative GABAergic terminals associated with GFP-labelled GnRH neurons. As expected, chronic DHT resulted in acyclicity and significantly increased body weight. However, no differences in LH pulse frequency or the density of VGAT appositions to GnRH neurons were identified between ovary-intact DHT-treated females and controls. Chronic DHT exposure significantly increased the number of AR expressing cells in the hypothalamus, whereas oestrogen receptor α-expressing neuron number was unchanged. Therefore, although chronic DHT exposure from 3 weeks of age increases AR expressing neurons in the brain, the GnRH neuronal network changes and hyperactive LH secretion associated with prenatal androgen excess are not evident. These findings suggest that unique central mechanisms are involved in the reproductive impairments driven by exposure to androgen excess at different developmental stages.
Collapse
Affiliation(s)
- Chris S. Coyle
- Centre for Neuroendocrinology and Department of PhysiologySchool of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| | - Melanie Prescott
- Centre for Neuroendocrinology and Department of PhysiologySchool of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| | - David J Handelsman
- Andrology LaboratoryANZAC Research InstituteConcord HospitalUniversity of SydneySydneyNSWAustralia
| | - Kirsty A. Walters
- Fertility and Research CentreSchool of Women’s and Children’s HealthUniversity of New South WalesSydneyNSWAustralia
| | - Rebecca E. Campbell
- Centre for Neuroendocrinology and Department of PhysiologySchool of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| |
Collapse
|
48
|
Grassi D, Marraudino M, Garcia-Segura LM, Panzica GC. The hypothalamic paraventricular nucleus as a central hub for the estrogenic modulation of neuroendocrine function and behavior. Front Neuroendocrinol 2022; 65:100974. [PMID: 34995643 DOI: 10.1016/j.yfrne.2021.100974] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/17/2022]
Abstract
Estradiol and hypothalamic paraventricular nucleus (PVN) help coordinate reproduction with body physiology, growth and metabolism. PVN integrates hormonal and neural signals originating in the periphery, generating an output mediated both by its long-distance neuronal projections, and by a variety of neurohormones produced by its magnocellular and parvocellular neurosecretory cells. Here we review the cyto-and chemo-architecture, the connectivity and function of PVN and the sex-specific regulation exerted by estradiol on PVN neurons and on the expression of neurotransmitters, neuromodulators, neuropeptides and neurohormones in PVN. Classical and non-classical estrogen receptors (ERs) are expressed in neuronal afferents to PVN and in specific PVN interneurons, projecting neurons, neurosecretory neurons and glial cells that are involved in the input-output integration and coordination of neurohormonal signals. Indeed, PVN ERs are known to modulate body homeostatic processes such as autonomic functions, stress response, reproduction, and metabolic control. Finally, the functional implications of the estrogenic modulation of the PVN for body homeostasis are discussed.
Collapse
Affiliation(s)
- D Grassi
- Department of Anatomy, Histology and Neuroscience, Universidad Autonoma de Madrid, Madrid, Spain
| | - M Marraudino
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy
| | - L M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - G C Panzica
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy; Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy.
| |
Collapse
|
49
|
Lee EB, Dilower I, Marsh CA, Wolfe MW, Masumi S, Upadhyaya S, Rumi MAK. Sexual Dimorphism in Kisspeptin Signaling. Cells 2022; 11:1146. [PMID: 35406710 PMCID: PMC8997554 DOI: 10.3390/cells11071146] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 02/05/2023] Open
Abstract
Kisspeptin (KP) and kisspeptin receptor (KPR) are essential for the onset of puberty, development of gonads, and maintenance of gonadal function in both males and females. Hypothalamic KPs and KPR display a high degree of sexual dimorphism in expression and function. KPs act on KPR in gonadotropin releasing hormone (GnRH) neurons and induce distinct patterns of GnRH secretion in males and females. GnRH acts on the anterior pituitary to secrete gonadotropins, which are required for steroidogenesis and gametogenesis in testes and ovaries. Gonadal steroid hormones in turn regulate the KP neurons. Gonadal hormones inhibit the KP neurons within the arcuate nucleus and generate pulsatile GnRH mediated gonadotropin (GPN) secretion in both sexes. However, the numbers of KP neurons in the anteroventral periventricular nucleus and preoptic area are greater in females, which release a large amount of KPs in response to a high estrogen level and induce the preovulatory GPN surge. In addition to the hypothalamus, KPs and KPR are also expressed in various extrahypothalamic tissues including the liver, pancreas, fat, and gonads. There is a remarkable difference in circulating KP levels between males and females. An increased level of KPs in females can be linked to increased numbers of KP neurons in female hypothalamus and more KP production in the ovaries and adipose tissues. Although the sexually dimorphic features are well characterized for hypothalamic KPs, very little is known about the extrahypothalamic KPs. This review article summarizes current knowledge regarding the sexual dimorphism in hypothalamic as well as extrahypothalamic KP and KPR system in primates and rodents.
Collapse
Affiliation(s)
- Eun Bee Lee
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Iman Dilower
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Courtney A. Marsh
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Michael W. Wolfe
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Saeed Masumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| | - Sameer Upadhyaya
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| | - Mohammad A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| |
Collapse
|
50
|
Fukui K, Sato K, Murakawa S, Minami M, Amano T. Estrogen signaling modulates behavioral selection toward pups and amygdalohippocampal area in the rhomboid nucleus of the bed nucleus of the stria terminalis circuit. Neuropharmacology 2022; 204:108879. [PMID: 34785164 DOI: 10.1016/j.neuropharm.2021.108879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 09/15/2021] [Accepted: 11/08/2021] [Indexed: 01/08/2023]
Abstract
Gonadal steroid hormone influences behavioral choice of adult animals toward pups, parental or aggressive. We previously reported that long-term administration of 17β-estradiol (E2) to male mice during sexual maturation induces aggressive behavior toward conspecific pups, which is called "infanticide," and significantly enhanced excitatory synaptic transmission in the rhomboid nucleus of bed nucleus of the stria terminalis (BSTrh), which is an important brain region for infanticide. However, it is unclear how estrogen receptor-dependent signaling after sexual maturity regulates neural circuits including the BSTrh. Here we revealed that E2 administration to gonadectomized mice in adulthood elicited infanticidal behavior and enhanced excitatory synaptic transmission in the BSTrh by increasing the probability of glutamate release from the presynaptic terminalis. Next, we performed whole-brain mapping of E2-sensitive brain regions projecting to the BSTrh and found that amygdalohippocampal area (AHi) neurons that project to the BSTrh densely express estrogen receptor 1 (Esr1). Moreover, E2 treatment enhanced synaptic connectivity in the AHi-BSTrh pathway. Together, these results suggest that reinforcement of excitatory inputs from AHi neurons into the BSTrh by estrogen receptor-dependent signaling may contribute to the expression of infanticide.
Collapse
Affiliation(s)
- Kiyoshiro Fukui
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan
| | - Keiichiro Sato
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan
| | - Shunsaku Murakawa
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan
| | - Masabumi Minami
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan
| | - Taiju Amano
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan.
| |
Collapse
|