1
|
Basak S, Dixit AK, Dey RK, Puia L, Bora M, Kumar Y R S, Babu G. An endocrinological perspective on polycystic ovarian syndrome. Mol Cell Endocrinol 2025; 602:112524. [PMID: 40147712 DOI: 10.1016/j.mce.2025.112524] [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: 11/17/2024] [Revised: 03/04/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Polycystic ovarian syndrome (PCOS) is a complex endocrinological disorder that involves dysfunctions across multiple endocrine axes, including the hypothalamic-pituitary-gonadal (HPG), hypothalamic-pituitary-adrenal (HPA) and hypothalamic-pituitary-thyroid (HPT) axes. Our review focuses on understanding the pathophysiology of PCOS through an endocrinological perspective, emphasizing the complex interactions between multiple endocrine axes. We have discussed the roles of the HPG, HPA, and HPT axes in PCOS. Dysregulation of the HPG axis, particularly the altered gonadotropin-releasing hormone pulse frequency resulting in elevated ratio of luteinizing hormone to follicle stimulating hormone, is central to the hyperandrogenism and anovulation, observed in PCOS. We have further highlighted the contributions of the HPA and HPT axes, where elevated adrenal androgen levels and hypothyroidism intensifies the phenotypes of PCOS. Additionally, insulin resistance and hyperinsulinemia, commonly associated with PCOS, aggravates hormonal disturbances and heighten the risk of metabolic complications such as type 2 diabetes and cardiovascular diseases. Elevated levels of anti-Müllerian hormone have also been emphasized as a key factor in inhibiting follicular growth, leading to impaired ovarian function and hyperandrogenism. This review further supports that PCOS is a multifactorial condition involving complex feedback mechanisms between the endocrine, metabolic, and reproductive systems. Furthermore, there remains a huge scope for deciphering the precise molecular interactions between the HPG, HPA, and HPT axes in PCOS, which could pave the way for targeted therapies for better management of both the endocrine and metabolic aspects of this disorder. This review will benefit researchers to get an endocrine perspective on PCOS.
Collapse
Affiliation(s)
- Smarto Basak
- Central Ayurveda Research Institute, Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Government of India, Kolkata, West Bengal, India
| | - Amit Kumar Dixit
- Central Ayurveda Research Institute, Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Government of India, Kolkata, West Bengal, India.
| | - Ranjit Kumar Dey
- Central Ayurveda Research Institute, Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Government of India, Kolkata, West Bengal, India
| | - Lalrin Puia
- Central Ayurveda Research Institute, Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Government of India, Kolkata, West Bengal, India
| | - Manajit Bora
- Central Ayurveda Research Institute, Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Government of India, Guwahati, Assam, India
| | - Sanjay Kumar Y R
- Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Government of India, Janakpuri, New Delhi, India
| | - Gajji Babu
- Central Ayurveda Research Institute, Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Government of India, Kolkata, West Bengal, India
| |
Collapse
|
2
|
Takizawa M, Miyazaki S, Tsuchida H, Nagae M, Seki S, Hirabayashi M, Osakada F, Inoue N, Tsukamura H, Uenoyama Y. Involvement of nuclear receptor corepressor 2 (NCOR2) in estrogen-induced repression of arcuate Kiss1 expression in female rats. J Reprod Dev 2025; 71:71-84. [PMID: 39864859 PMCID: PMC11999826 DOI: 10.1262/jrd.2024-100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/24/2024] [Indexed: 01/28/2025] Open
Abstract
Hypothalamic arcuate (ARC) kisspeptin neurons are considered the gonadotropin-releasing hormone pulse generator in rats. In virgin rats, the expression of the ARC kisspeptin gene (Kiss1) is repressed by proestrous levels of estradiol-17β (high E2) but not by diestrous levels of E2 (low E2). In lactating rats, ARC Kiss1 expression is repressed by low E2 during late lactation. This study aimed to investigate whether nuclear receptor corepressor 2 (NCOR2, encoded by Ncor2), an estrogen receptor α corepressor, is involved in the estrogen-induced repression of ARC Kiss1 expression in rats. Double in situ hybridization for Kiss1 and Ncor2 revealed that approximately 80% of ARC Kiss1-expressing cells co-expressed Ncor2 in ovariectomized (OVX) + low E2 virgin rats, while approximately 90% of ARC Kiss1-expressing cells co-expressed Ncor2 in OVX + low E2 lactating rats. To further examine the role of Ncor2, we studied the effects of Kiss1-dependent Ncor2 knockdown on ARC Kiss1 expression and luteinizing hormone (LH) pulses. An adeno-associated virus vector carrying Cre-activated short hairpin RNA (shRNA) for Ncor2 was administered to the ARC in two Kiss1-Cre rat models: OVX + high E2 Kiss1-Cre virgin rats and OVX + low E2 Kiss1-Cre lactating rats. Ncor2-shRNA treatment significantly increased the number of ARC Kiss1-expressing cells and the intensity of Kiss1 signals in OVX + high E2 virgin rats but failed to fully restore low E2-induced Kiss1 repression in lactating rats. The Ncor2-shRNA treatment failed to affect LH pulses in both models. These findings suggest that NCOR2 in ARC kisspeptin neurons mediates high E2-induced repression of ARC Kiss1 expression in virgin rats.
Collapse
Affiliation(s)
- Marina Takizawa
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Sae Miyazaki
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Hitomi Tsuchida
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Mayuko Nagae
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Shunsuke Seki
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Masumi Hirabayashi
- Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki 444‑8787, Japan
| | - Fumitaka Osakada
- Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Naoko Inoue
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Hiroko Tsukamura
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Yoshihisa Uenoyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
3
|
Peng D, Lu C, Spadacini V, Mitchell K, Tan Y, Zhang D, Levavi-Sivan B, Hu W, Trudeau VL. Hormonal dynamics reveal a stimulatory role for secretoneurin in zebrafish ovulation. PNAS NEXUS 2025; 4:pgaf097. [PMID: 40191135 PMCID: PMC11969067 DOI: 10.1093/pnasnexus/pgaf097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 03/07/2025] [Indexed: 04/09/2025]
Abstract
Surge release of luteinizing hormone (Lh) from the pituitary is essential for fertility as it triggers ovulation. While secretoneurin (SN) is a phylogenetically conserved secretogranin-2-derived peptide that stimulates Lh, its role in ovulation has not been established. To directly compare periovulatory changes in the classical hormones to the emerging reproductive neuropeptides SNa and SNb, simultaneous mass spectrometry measurement of 9 peptides and 5 steroids was conducted in female zebrafish. Regression analysis indicated that levels of SNa1-34 in the brain peaked when type 3 gonadotropin-releasing hormone (Gnrh3) increased (R 2 = 0.71) at the time of the Lh surge, 3.5 h before ovulation. Levels of the naturally occurring derivative SNa1-14 were highest at ovulation, while SNb1-31 was invariable. The bioactivities of SNa1-34 and SNa1-14 were investigated. After injection of SNa1-34 in females that had been isolated from males, 61% (11/18) ovulated within 6 h, which was like the effects of the Lh analog human chorionic gonadotropin (72%; 13/18 females). SNa1-34 injection induces ovulation by increasing time-dependent expression of gnrh3 in the brain, a likely direct stimulation of chorionic gonadotropin alpha (cga) and luteinizing hormone b (lhb) subunit in pituitary, and via the subsequent time-dependent increase in nuclear progesterone receptor (npr) in ovaries. In contrast, SNa1-14 exhibited far fewer effects on gene expression and did not induce ovulation. Our results support the proposal that SN is a reproductive hormone.
Collapse
Affiliation(s)
- Di Peng
- Department of Biology, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| | - Chunyu Lu
- Department of Biology, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| | - Victoria Spadacini
- Department of Biology, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| | - Kimberly Mitchell
- Department of Biology, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| | - Yongjun Tan
- Department of Biology, Bioinformatics and Computational Biology Program, College of Arts and Sciences, Saint Louis University, Saint Louis, MO 63103-2010, USA
| | - Dapeng Zhang
- Department of Biology, Bioinformatics and Computational Biology Program, College of Arts and Sciences, Saint Louis University, Saint Louis, MO 63103-2010, USA
| | - Berta Levavi-Sivan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Wei Hu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China
| | - Vance L Trudeau
- Department of Biology, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| |
Collapse
|
4
|
Talbi R, Stincic TL, Ferrari K, Hae CJ, Walec K, Medve E, Gerutshang A, León S, McCarthy EA, Rønnekleiv OK, Kelly MJ, Navarro VM. POMC neurons control fertility through differential signaling of MC4R in Kisspeptin neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.18.580873. [PMID: 38915534 PMCID: PMC11195098 DOI: 10.1101/2024.02.18.580873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Inactivating mutations in the melanocortin 4 receptor (MC4R) gene cause monogenic obesity. Interestingly, female patients also display various degrees of reproductive disorders, in line with the subfertile phenotype of MC4RKO female mice. However, the cellular mechanisms by which MC4R regulates reproduction are unknown. Kiss1 neurons directly stimulate gonadotropin-releasing hormone (GnRH) release through two distinct populations; the Kiss1ARH neurons, controlling GnRH pulses, and the sexually dimorphic Kiss1AVPV/PeN neurons controlling the preovulatory LH surge. Here, we show that Mc4r expressed in Kiss1 neurons regulates fertility in females. In vivo, deletion of Mc4r from Kiss1 neurons in female mice replicates the reproductive impairments of MC4RKO mice without inducing obesity. Conversely, reinsertion of Mc4r in Kiss1 neurons of MC4R null mice restores estrous cyclicity and LH pulsatility without reducing their obese phenotype. In vitro, we dissect the specific action of MC4R on Kiss1ARH vs Kiss1AVPV/PeN neurons and show that MC4R activation excites Kiss1ARH neurons through direct synaptic actions. In contrast, Kiss1AVPV/PeN neurons are normally inhibited by MC4R activation except under elevated estradiol levels, thus facilitating the activation of Kiss1AVPV/PeN neurons to induce the LH surge driving ovulation in females. Our findings demonstrate that POMCARH neurons acting through MC4R, directly regulate reproductive function in females by stimulating the "pulse generator" activity of Kiss1ARH neurons and restricting the activation of Kiss1AVPV/PeN neurons to the time of the estradiol-dependent LH surge, and thus unveil a novel pathway of the metabolic regulation of fertility by the melanocortin system.
Collapse
Affiliation(s)
- Rajae Talbi
- Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Todd L. Stincic
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Kaitlin Ferrari
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Choi Ji Hae
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Karol Walec
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Elizabeth Medve
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Achi Gerutshang
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Silvia León
- Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Elizabeth A. McCarthy
- Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Oline K. Rønnekleiv
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Martin J. Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Víctor M. Navarro
- Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Program in Neuroscience, Boston, MA, USA
| |
Collapse
|
5
|
Carrasco RA, Jang J, Jung J, McCosh RB, Kreisman MJ, Breen KM. Prostaglandin synthesis mediates the suppression of arcuate Kiss1 neuron activation and pulsatile luteinizing hormone secretion during immune/inflammatory stress in female mice. J Neuroendocrinol 2025:e70004. [PMID: 40058772 DOI: 10.1111/jne.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 03/14/2025]
Abstract
Stress induces a series of compensatory mechanisms with the objective of restoration or adaptation of physiological function. A common casualty of the response to stress is impaired reproduction via the inhibition of pulsatile luteinizing hormone (LH) secretion; however, how stressors convey LH inhibition remains unclear and may be dependent on stress type. Immune/inflammatory stress, modeled with peripheral lipopolysaccharide (LPS) exposure, induces a systemic inflammatory response which may contrast with the neural mechanisms employed by psychosocial stressors. We examined the suppressive effect of LPS versus psychosocial stress, modeled with restraint, on pulsatile LH secretion and investigated the neural mechanisms underlying LPS-induced LH suppression in ovariectomized (OVX) female mice. We observed that both LPS and restraint significantly suppressed mean LH concentrations; however, the dynamics of pulse suppression displayed stress-type dependency. LPS induced a reduction in both LH pulse frequency and amplitude, whereas restraint suppressed LH pulse frequency without compromising pulse amplitude. Next, we investigated the mediatory role of immune/inflammatory signaling for LPS to impair LH secretion and upstream arcuate Kiss1 cell function. Peripheral administration of flurbiprofen, a prostaglandin synthesis inhibitor, blocked the suppressive effect of LPS on LH pulse frequency and amplitude. Interestingly, flurbiprofen only partially prevented the suppressive effect of LPS on arcuate Kiss1 cell activity, as measured by c-Fos expression. These data demonstrate that immune/inflammatory stress inhibits the activity of the LH pulse generator, in part, via a prostaglandin-dependent pathway and supports the role of differential neural mechanisms mediating LH pulse suppression during stress.
Collapse
Affiliation(s)
- Rodrigo A Carrasco
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, California, USA
| | - Jessica Jang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, California, USA
| | - Jacklyn Jung
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, California, USA
| | - Richard B McCosh
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, California, USA
| | - Michael J Kreisman
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, California, USA
| | - Kellie M Breen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
6
|
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:e70011. [PMID: 40033683 DOI: 10.1111/jne.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
7
|
Griesgraber MJ, Coolen LM, Onslow KM, Corey JR, Rice RE, Aerts EG, Bowdridge EC, Hardy SL, Lehman MN, Goodman RL, Hileman SM. Critical role of arcuate nucleus kisspeptin and Kiss1R in regulation of the ovine luteinizing hormone surge. J Neuroendocrinol 2025:e70010. [PMID: 40033679 DOI: 10.1111/jne.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/28/2025] [Accepted: 02/13/2025] [Indexed: 03/05/2025]
Abstract
Hypothalamic kisspeptin (Kiss), neurokinin B (NKB), and dynorphin-containing (KNDy) neurons in the arcuate nucleus (ARC) have consistently been shown to be the central generator of gonadotropin-releasing hormone (GnRH) and corresponding luteinizing hormone (LH) pulses in mammals and possibly contribute to surge secretion as well. Additionally, recent evidence from experiments in sheep suggests that ARC Kiss1R-containing neurons play an important role in regulating the timing and amplitude of LH pulses. In this study, we examined the functional role of ARC KNDy and Kiss1R-containing neurons in ovine LH surge secretion via injection of saporin-ligand conjugates (SAP) to ablate these neural populations. NKB-SAP injections significantly reduced the percentage of ARC Kiss1 (~65% decrease) cells compared to control animals, and a surge-like increase of LH was prevented in ewes with the greatest degree of Kiss1 cell ablation. Kiss-SAP injections had no effect on Kiss1 cell percentage or ARC Kiss1R cell number compared to controls, the latter perhaps due to Kiss1R suppression in control animals from elevated estradiol concentrations during the LH surge. However, Kiss-SAP injections consistently and robustly decreased LH surge amplitude, with 80% of Kiss-SAP-treated ewes failing to generate a surge. While the exact identity of these ARC Kiss1R neurons has yet to be fully elucidated, they likely act downstream or in concert with KNDy neurons and possibly integrate other surge-centric signaling pathways to generate the ovine LH surge. These results support the conclusion that KNDy neurons contribute significantly to the ovine LH surge, while ARC Kiss1R neurons appear to be necessary for a functional surge to occur in sheep.
Collapse
Affiliation(s)
- Max J Griesgraber
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
| | - Lique M Coolen
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Kayla M Onslow
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Jacob R Corey
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Rachel E Rice
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Eliana G Aerts
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
| | - Elizabeth C Bowdridge
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
| | - Steven L Hardy
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
| | - Michael N Lehman
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Robert L Goodman
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
- Department of Neuroscience, West Virginia University, Morgantown, West Virginia, USA
| | - Stanley M Hileman
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
- Department of Neuroscience, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
8
|
Kacimi L, Prevot V. GnRH and Cognition. Endocrinology 2025; 166:bqaf033. [PMID: 39996304 DOI: 10.1210/endocr/bqaf033] [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: 12/18/2024] [Revised: 02/06/2025] [Accepted: 02/23/2025] [Indexed: 02/26/2025]
Abstract
GnRH is traditionally recognized as the central regulator of reproduction through its pulsatile secretion, which governs the hypothalamic-pituitary-gonadal axis. However, recent evidence has highlighted its broader role in brain development and function, including in cognitive and higher intellectual processes. GnRH production follows distinct phases, from its early activation during minipuberty-the first postnatal activation of GnRH neurons during the infantile period-, its reactivation and stabilization starting at puberty, and its eventual decline with age and the loss of gonadal steroid feedback. This evolution depends on the establishment, maturation and activation of GnRH neurons, a complex process regulated by the cellular and molecular environment of these neurons, including multiple neuronal and glial types as well as a minipubertal "switch" in gene expression, the perturbation of which may have long-term or delayed consequences for both reproductive and cognitive function. The cognitive role of GnRH may be related to its recently revealed involvement in maintaining myelination and synaptic plasticity, whereas disruptions in its finely tuned rhythmic secretion, either age-related or pathological, are associated with cognitive decline and neurodegenerative disorders. Restoring physiological GnRH levels and pulsatility can reverse age-related cognitive decline and improve sensory functions even in adulthood, suggesting a mobilization of the "cognitive reserve" in both animal models and human patients. This review highlights recent advances in our understanding of the GnRH system and the therapeutic potential of pulsatile GnRH therapy to mitigate age-related cognitive decline and neurodegenerative processes.
Collapse
Affiliation(s)
- Loïc Kacimi
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, FHU 1000 days for health, EGID, DistALZ, UMR_S112, Lille 59000, France
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, FHU 1000 days for health, EGID, DistALZ, UMR_S112, Lille 59000, France
| |
Collapse
|
9
|
Carrasco RA, Breen KM. Brainstem Noradrenergic Neuronal Populations: Dual Effects on Regulating GnRH and LH Secretion. Endocrinology 2025; 166:bqaf021. [PMID: 39891672 PMCID: PMC11815497 DOI: 10.1210/endocr/bqaf021] [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: 10/31/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/03/2025]
Abstract
Noradrenergic neurons are a brain network that integrate viscero-sensorial signals to modulate neural and neuroendocrine function. Although it has been known for decades that noradrenergic neural circuits influence neuroendocrine and reproductive function, the cellular and molecular players involved remain largely unknown. The objective of this review is to summarize past and current knowledge regarding the influence of brainstem noradrenergic systems on GnRH and gonadotrophin secretion. The main noradrenergic cell groups A1, A2, and A6, known as the ventrolateral medulla, nucleus of the solitary tract, and locus coeruleus, respectively, are involved in the control of reproductive neuroendocrine secretion. Current evidence suggests that brainstem noradrenergic circuits promote the generation and maintenance of the LH surge in both spontaneous (rats, sheep) and induced (rabbit, ferret) ovulators. In contrast, recent studies have established that LH pulsatile secretion is suppressed by specific activation of brainstem noradrenergic cell groups. The duality of the GnRH/LH response to noradrenaline reflects the inherent complexity of hindbrain noradrenaline neurons, which are responsive to stressors and gonadal steroids (ie, estradiol) and coexpress a variety of neurotransmitters and neuropeptides. Therefore, elucidating the organization and functionality of brainstem noradrenergic systems will provide targets for controlling reproduction and understanding the interconnection with stress.
Collapse
Affiliation(s)
- Rodrigo A Carrasco
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093-0674, USA
| | - Kellie M Breen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093-0674, USA
| |
Collapse
|
10
|
Ignatiuk V, Sharova V, Zakharova L. Prenatal Inflammation Reprograms Hypothalamic-Pituitary-Gonadal Axis Development in Female Rats. Inflammation 2025:10.1007/s10753-025-02243-2. [PMID: 39909991 DOI: 10.1007/s10753-025-02243-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 02/07/2025]
Abstract
The hypothalamic-pituitary-gonadal (HPG) axis development during critical periods of ontogenesis can be disrupted by stress factors, including in particular maternal immune activation by infectious agents. Bacterial lipopolysaccharide (LPS, E.coli) exposure induces inflammation accompanied by proinflammatory cytokine release. The resulting elevated cytokine levels may lead to a disruption of epigenetic mechanisms regulating HPG axis development and to a reduced fertility in the offspring. This study focused on the long-term effects of prenatal LPS exposure on HPG axis development in female rats and the modulation of such effects by anti-inflammatory drugs: polyclonal IgG and monoclonal anti-IL6-receptor antibodies. LPS exposure on embryonic day 12 led to a decrease in the number of synaptic inputs on gonadotropin-releasing-hormone-producing neurons in the hypothalamus, high levels of follicular atresia, and suppressed steroidogenesis in the ovaries of adult female offspring. IgG treatment or IL6 receptor blockade by monoclonal antibodies 40 minutes after LPS exposure prevented these long-term negative effects of LPS. The data obtained suggest that IL6 is involved in the regulation of HPG axis development.
Collapse
Affiliation(s)
- Vasilina Ignatiuk
- Koltsov Institute of Developmental Biology, The Russian Academy of Sciences, Vavilov Street, 26, 119334, Moscow, Russia
| | - Viktoriya Sharova
- Koltsov Institute of Developmental Biology, The Russian Academy of Sciences, Vavilov Street, 26, 119334, Moscow, Russia.
| | - Liudmila Zakharova
- Koltsov Institute of Developmental Biology, The Russian Academy of Sciences, Vavilov Street, 26, 119334, Moscow, Russia
| |
Collapse
|
11
|
Kraynak M, Willging MM, Uhlrich DJ, Shapiro RA, Flowers MT, Manning KA, John SD, Williams SM, Henjum LJ, Marrah RC, Yohnk HR, Berg CB, Brunner K, Colman RJ, Alexander AL, Abbott DH, Levine JE. Hypothalamic Estrogen Receptor α Is Essential for Female Marmoset Sexual Behavior Without Protecting From Obesity. J Endocr Soc 2025; 9:bvaf012. [PMID: 39911518 PMCID: PMC11795203 DOI: 10.1210/jendso/bvaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Indexed: 02/07/2025] Open
Abstract
Context Estrogen receptor α (ERα) in the ventromedial (VMN) and arcuate (ARC) nuclei of female rodent mediobasal hypothalami (MBHs) provides a crucial molecular gateway facilitating estradiol (E2) regulation of sexual behavior, reproductive neuroendocrinology, and metabolic function. In female nonhuman primates (NHPs) and women, however, its hypothalamic counterpart remains unknown. Objective We hypothesized that knockdown (KD) of ERα expression in the hypothalamic VMN and ARC of female marmosets would diminish sexual receptivity, while simultaneously disrupting gonadotropic and metabolic homeostasis. Methods We ovariectomized (OVX) adult female marmosets of comparable age and weight, immediately replaced E2 at midcycle levels, and approximately 1 month later assigned monkeys to diet-induced obesity (DIO) within group (1) control, receiving scrambled short hairpin RNA (shRNA), or (2) ERαKD, receiving selective ERα gene silencing shRNA. Magnetic resonance imaging-guided neural surgery enabled hypothalamic infusion of viral vector shRNA and subsequent brain immunohistochemistry enabled observer-validated, NIS-elements computer software quantification of ERα knockdown. Results ERα expression was significantly diminished in the VMN and ARC, but not the preoptic area (POA), of ERαKD females coincident with elimination of timely female sexual responses, more than 80% loss of female receptivity, modestly elevated gonadotropin levels, hyperglycemia, and diminished calorie consumption. Density and intensity of ERα-expressing cells in the VMN correlated positively with female sexual receptivity and calorie consumption, negatively with timeliness of female sexual responses, and in the ARC, correlated negatively with calorie consumption. Conclusion ERα activation in the female NHP MBH is critically important for female sexual behavior and modestly contributes to gonadotropic and metabolic control.
Collapse
Affiliation(s)
- Marissa Kraynak
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI 53715, USA
- Endocrinology-Reproductive Physiology Training Program, University of Wisconsin–Madison, Madison, WI 53715, USA
| | - Molly M Willging
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI 53715, USA
- Endocrinology-Reproductive Physiology Training Program, University of Wisconsin–Madison, Madison, WI 53715, USA
| | - Daniel J Uhlrich
- Department of Neuroscience, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Robert A Shapiro
- Department of Neuroscience, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Matthew T Flowers
- Department of Medicine, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Karen A Manning
- Department of Neuroscience, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Sara D John
- Department of Radiology, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Samantha M Williams
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI 53715, USA
| | - Lukas J Henjum
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI 53715, USA
| | - Rebecca C Marrah
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI 53715, USA
| | - Hannah R Yohnk
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI 53715, USA
| | - Carter B Berg
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI 53715, USA
| | - Kevin Brunner
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI 53715, USA
| | - Ricki J Colman
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI 53715, USA
- Endocrinology-Reproductive Physiology Training Program, University of Wisconsin–Madison, Madison, WI 53715, USA
- Department of Cell and Regenerative Biology, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Andrew L Alexander
- Department of Cell and Regenerative Biology, University of Wisconsin–Madison, Madison, WI 53705, USA
- Department of Medical Physics, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - David H Abbott
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI 53715, USA
- Endocrinology-Reproductive Physiology Training Program, University of Wisconsin–Madison, Madison, WI 53715, USA
- Department of Obstetrics and Gynecology, University of Wisconsin–Madison, Madison, WI 53715, USA
| | - Jon E Levine
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI 53715, USA
- Endocrinology-Reproductive Physiology Training Program, University of Wisconsin–Madison, Madison, WI 53715, USA
- Department of Neuroscience, University of Wisconsin–Madison, Madison, WI 53705, USA
| |
Collapse
|
12
|
Stojilkovic SS, Sokanovic SJ, Constantin S. What is known and unknown about the role of neuroendocrine genes Ptprn and Ptprn2. Front Endocrinol (Lausanne) 2025; 16:1531723. [PMID: 39926347 PMCID: PMC11802530 DOI: 10.3389/fendo.2025.1531723] [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: 11/20/2024] [Accepted: 01/08/2025] [Indexed: 02/11/2025] Open
Abstract
The protein tyrosine phosphatase receptors N and N2 are encoded by the Ptprn and Ptprn2 genes expressed in neuroendocrine cells of the hypothalamus, pituitary gland, and diffuse neuroendocrine system, including the pancreas, lung, and intestine. Unlike other members of the protein tyrosine phosphatase receptor family, PTPRN and PTPRN2 lack protein tyrosine phosphatase activity due to mutation of two residues in their intracellular catalytic domains. However, during evolution these proteins acquired new cellular roles beyond tyrosine dephosphorylation in the centralized and diffuse neuroendocrine systems. Here we discuss the current understanding and lack of information about the actions of these proteins, focusing on neuroendocrine cells of the hypothalamus, pituitary, and pancreas.
Collapse
Affiliation(s)
- Stanko S. Stojilkovic
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| | | | | |
Collapse
|
13
|
Dai R, Sun Y. Altered GnRH neuron-glia networks close to interface of polycystic ovary syndrome: Molecular mechanism and clinical perspectives. Life Sci 2025; 361:123318. [PMID: 39719166 DOI: 10.1016/j.lfs.2024.123318] [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: 08/27/2024] [Revised: 11/24/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024]
Abstract
Polycystic ovary syndrome (PCOS) has been noticed as a neuroendocrine syndrome manifested by reproductive hormone dysregulation involving increased luteinizing hormone (LH) pulse frequency and an increased LH to follicle-stimulating hormone ratio, yet theory is just beginning to be established. Neuroglia located in the arcuate nucleus and median eminence (ARC-ME) that are close to gonadotropin-releasing hormone (GnRH) axon terminals, comprise the blood-brain barrier and fenestrated vessels implying their putative roles in the modulation of the abnormal GnRH pulse in PCOS. This review outlines the disturbances of neuron-glia networks that underlie hypothetically the deregulation of GnRH-LH release and impaired sex hormone negative feedback in PCOS. We then discuss chronic and low-grade inflammatory status together with gut dysbiosis and how the detriments may intrude the hypothalamus by virtue of violating interfaces between the brain and periphery, which might contribute to the etiology of the impaired neural circuits in the ARC-ME to induce PCOS.
Collapse
Affiliation(s)
- Ruoxi Dai
- Hospital & Institute of Obstetrics and Gynecology, Fudan University, Shanghai 200081, China
| | - Yan Sun
- Hospital & Institute of Obstetrics and Gynecology, Fudan University, Shanghai 200081, China; The Academy of Integrative Medicine, Fudan University, Shanghai 200081, China; Shanghai Key Laboratory of Female Reproductive Endocrine-related Disease, Shanghai 200081, China.
| |
Collapse
|
14
|
Yeo SH, Han SY, Herbison AE. Shifting GnRH Neuron Ensembles Underlie Successive Preovulatory Luteinizing Hormone Surges. J Neurosci 2025; 45:e1383242024. [PMID: 39505408 PMCID: PMC11735651 DOI: 10.1523/jneurosci.1383-24.2024] [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: 07/19/2024] [Revised: 08/30/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024] Open
Abstract
The gonadotropin-releasing hormone (GnRH) neurons operate as a neuronal ensemble exhibiting coordinated activity once every reproductive cycle to generate the preovulatory GnRH surge. Using GCaMP fiber photometry at the GnRH neuron distal dendrons to measure the output of this widely scattered population in female mice, we find that the onset, amplitude, and profile of GnRH neuron surge activity exhibits substantial variability from cycle to cycle both between and within individual mice. This was also evident when measuring successive proestrous luteinizing hormone surges. Studies combining short (c-Fos and c-Jun) and long (genetic robust activity marking) term indices of immediate early gene activation revealed that, while ∼50% of GnRH neurons were activated at the time of each surge, only half of these neurons had been active during the previous proestrous surge. These observations reveal marked inter- and intra-individual variability in the GnRH surge mechanism. Remarkably, different subpopulations of overlapping GnRH neurons are recruited to the ensemble each estrous cycle to generate the GnRH surge. While engendering variability in the surge mechanism itself, this likely provides substantial robustness to a key event underlying mammalian reproduction.
Collapse
Affiliation(s)
- Shel-Hwa Yeo
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | - Su Young Han
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | - Allan E Herbison
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| |
Collapse
|
15
|
Han SY, Yeo SH, Kim JC, Zhou Z, Herbison AE. Multi-dimensional oscillatory activity of mouse GnRH neurons in vivo. eLife 2025; 13:RP100856. [PMID: 39773874 PMCID: PMC11709428 DOI: 10.7554/elife.100856] [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] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
The gonadotropin-releasing hormone (GnRH) neurons represent the key output cells of the neural network controlling mammalian fertility. We used GCaMP fiber photometry to record the population activity of the GnRH neuron distal projections in the ventral arcuate nucleus where they merge before entering the median eminence to release GnRH into the portal vasculature. Recordings in freely behaving intact male and female mice revealed abrupt ~8 min duration increases in activity that correlated perfectly with the appearance of a subsequent pulse of luteinizing hormone (LH). The GnRH neuron dendrons also exhibited a low level of unchanging clustered, rapidly fluctuating baseline activity in males and throughout the estrous cycle in females. In female mice, a gradual increase in basal activity that exhibited ~80 min oscillations began in the afternoon of proestrus and lasted for 12 hr. This was associated with the onset of the LH surge that ended several hours before the fall in the GCaMP signal. Abrupt 8 min duration episodes of GCaMP activity continued to occur on top of the rising surge baseline before ceasing in estrus. These observations provide the first description of GnRH neuron activity in freely behaving animals. They demonstrate that three distinct patterns of oscillatory activity occur in GnRH neurons. These are comprised of low-level rapid baseline activity, abrupt 8 min duration oscillations that drive pulsatile gonadotropin secretion, and, in females, a gradual and very prolonged oscillating increase in activity responsible for the preovulatory LH surge.
Collapse
Affiliation(s)
- Su Young Han
- Department of Physiology, Development and Neuroscience, Downing site, University of CambridgeCambridgeUnited Kingdom
| | - Shel-Hwa Yeo
- Department of Physiology, Development and Neuroscience, Downing site, University of CambridgeCambridgeUnited Kingdom
| | - Jae-Chang Kim
- Zurich Center for Neuroeconomics, Department of Economics, University of ZurichZurichSwitzerland
| | - Ziyue Zhou
- Department of Physiology, Development and Neuroscience, Downing site, University of CambridgeCambridgeUnited Kingdom
| | - Allan E Herbison
- Department of Physiology, Development and Neuroscience, Downing site, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
16
|
Zhou Z, Han SY, Pardo-Navarro M, Wall EG, Desai R, Vas S, Handelsman DJ, Herbison AE. GnRH pulse generator activity in mouse models of polycystic ovary syndrome. eLife 2025; 13:RP97179. [PMID: 39761106 DOI: 10.7554/elife.97179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
One in ten women in their reproductive age suffer from polycystic ovary syndrome (PCOS) that, alongside subfertility and hyperandrogenism, typically presents with increased luteinizing hormone (LH) pulsatility. As such, it is suspected that the arcuate kisspeptin (ARNKISS) neurons that represent the GnRH pulse generator are dysfunctional in PCOS. We used here in vivo GCaMP fiber photometry and other approaches to examine the behavior of the GnRH pulse generator in two mouse models of PCOS. We began with the peripubertal androgen (PPA) mouse model of PCOS but found that it had a reduction in the frequency of ARNKISS neuron synchronization events (SEs) that drive LH pulses. Examining the prenatal androgen (PNA) model of PCOS, we observed highly variable patterns of pulse generator activity with no significant differences detected in ARNKISS neuron SEs, pulsatile LH secretion, or serum testosterone, estradiol, and progesterone concentrations. However, a machine learning approach identified that the ARNKISS neurons of acyclic PNA mice continued to exhibit cyclical patterns of activity similar to that of normal mice. The frequency of ARNKISS neuron SEs was significantly increased in algorithm-identified 'diestrous stage' PNA mice compared to controls. In addition, ARNKISS neurons exhibited reduced feedback suppression to progesterone in PNA mice and their gonadotrophs were also less sensitive to GnRH. These observations demonstrate the importance of understanding GnRH pulse generator activity in mouse models of PCOS. The existence of cyclical GnRH pulse generator activity in the acyclic PNA mouse indicates the presence of a complex phenotype with deficits at multiple levels of the hypothalamo-pituitary-gonadal axis.
Collapse
Affiliation(s)
- Ziyue Zhou
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Su Young Han
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Maria Pardo-Navarro
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Ellen G Wall
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Reena Desai
- ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Szilvia Vas
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | | | - Allan E Herbison
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
17
|
Pertynska-Marczewska M, Pertynski T. Non-hormonal pharmacological interventions for managing vasomotor symptoms-how can we help: 2024 landscape. Eur J Obstet Gynecol Reprod Biol 2024; 302:141-148. [PMID: 39270577 DOI: 10.1016/j.ejogrb.2024.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Vasomotor symptoms (VMS) affect 70% of menopausal women and are considered as hallmark symptoms of the menopausal transition experienced by over three quarters of women and severely by 25% of women. Estrogen withdrawal alone is not fully responsible for the onset of the menopausal vasomotor symptoms and the mechanism of altered thermoregulation appears to be centrally mediated with alterations in hypothalamic neurotransmitters playing a key part. The loss of thermoregulatory control coexists with the altered Kisspeptin- Neurokinin B-Dynorphin-expressing (KNDy) neurons of the arcuate nucleus signaling triggered by menopause. OBJECTIVE Aim of the review was to explore evidence-based non-hormonal pharmacological interventions for treating vasomotor symptoms. METHODS Comprehensive overview of relevant literature. CONCLUSIONS In the population where, hormonal options are contraindicated or not preferred by the patient, it is essential to explore evidence-based non-hormonal pharmacological interventions for treating vasomotor symptoms. The 2024 landscape of available treatments has expanded yet again, arming the providers with an even wider range of possibilities to help their patients. Fezolinetant, is the first NK3R antagonist developed for the purpose of treating hot flashes in menopausal women. NK3R antagonists provide a safe and effective treatment option for managing menopausal women with VMS.
Collapse
|
18
|
Coutinho EA, Esparza LA, Steffen PH, Liaw R, Bolleddu S, Kauffman AS. Selective depletion of kisspeptin neurons in the hypothalamic arcuate nucleus in early juvenile life reduces pubertal LH secretion and delays puberty onset in mice. FASEB J 2024; 38:e70078. [PMID: 39377760 PMCID: PMC11804785 DOI: 10.1096/fj.202401696r] [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: 07/24/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024]
Abstract
Puberty is the critical developmental transition to reproductive capability driven by the activation of gonadotropin-releasing hormone (GnRH) neurons. The complex neural mechanisms underlying pubertal activation of GnRH secretion still remain unknown, yet likely include kisspeptin neurons. However, kisspeptin neurons reside in several hypothalamic areas and the specific kisspeptin population timing pubertal onset remains undetermined. To investigate this, we strategically capitalized on the differential ontological expression of the Kiss1 gene in different hypothalamic nuclei to selectively ablate just arcuate kisspeptin neurons (aka KNDy neurons) during the early juvenile period, well before puberty, while sparing RP3V kisspeptin neurons. Both male and female transgenic mice with a majority of their KNDy neurons ablated (KNDyABL) by diphtheria toxin treatment in juvenile life demonstrated significantly delayed puberty onset and lower peripubertal LH secretion than controls. In adulthood, KNDyABL mice demonstrated normal in vivo LH pulse frequency with lower basal and peak LH levels, suggesting that only a small subset of KNDy neurons is sufficient for normal GnRH pulse timing but more KNDy cells are needed to secrete normal LH concentrations. Unlike prior KNDy ablation studies in rats, there was no alteration in the occurrence or magnitude of estradiol-induced LH surges in KNDyABL female mice, indicating that a complete KNDy neuronal population is not essential for normal LH surge generation. This study teases apart the contributions of different kisspeptin neural populations to the control of puberty onset, demonstrating that a majority of KNDy neurons in the arcuate nucleus are necessary for the proper timing of puberty in both sexes.
Collapse
Affiliation(s)
- Eulalia A Coutinho
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - Lourdes A Esparza
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - Paige H Steffen
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - Reanna Liaw
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - Shreyana Bolleddu
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - Alexander S Kauffman
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
19
|
Uenoyama Y, Nonogaki M, Tsuchida H, Takizawa M, Matsuzaki S, Inoue N, Tsukamura H. Central δ/κ opioid receptor signaling pathways mediate chronic and/or acute suckling-induced LH suppression in rats during late lactation. J Reprod Dev 2024; 70:327-337. [PMID: 39155080 PMCID: PMC11461525 DOI: 10.1262/jrd.2024-045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
In mammals, secretion of tonic (pulsatile) gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) is often suppressed during lactation. Suppression of GnRH/LH pulses in lactating dams is assumed to be caused by suckling stimuli and a chronic negative energy balance due to milk production. The present study aimed to investigate whether the central enkephalin-δ opioid receptor (DOR) signaling mediated the suppression of LH secretion by acute suckling stimuli and/or chronic negative energy balance due to milk production in rats during late lactation when dams were under a heavy energy demand. On postpartum day 16, the number of Penk (enkephalin mRNA)-expressing cells in the arcuate nucleus was significantly higher in lactating rats than in non-lactating control rats. Pulsatile LH secretion was suppressed in rats with chronic suckling or acute 1-h suckling stimuli 6 h after pup removal on day 16 of lactation. Central DOR antagonism significantly increased the mean LH concentrations and the baseline of LH pulses in rats with chronic suckling but not with acute suckling stimuli on day 16 of lactation. Besides, central κ opioid receptor (KOR) antagonism increased the amplitude of LH pulses in rats with the acute suckling stimuli on day 16 of lactation. These results suggest that central DOR signaling mediates the suppression of LH secretion caused by a negative energy balance in rats receiving chronic suckling during late lactation. On the other hand, central KOR signaling likely mediates acute suckling stimuli-induced suppression of LH secretion in rats during late lactation.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Miku Nonogaki
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Hitomi Tsuchida
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Marina Takizawa
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Sena Matsuzaki
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
20
|
Moenter SM, Starrett JR. Estradiol action in the female hypothalamo-pituitary-gonadal axis. J Neuroendocrinol 2024; 36:e13390. [PMID: 38606585 PMCID: PMC11444910 DOI: 10.1111/jne.13390] [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: 01/15/2024] [Revised: 03/06/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024]
Abstract
It has now been about a century since a flurry of discoveries identified first the pituitary, then more specifically the anterior pituitary and soon thereafter the central nervous system as components regulating gonadal and downstream reproductive functions. This was an era of ablation/replacement designs using at first rudimentary and then increasingly pure preparations of gonadal and pituitary "activities" or transplanting actual glands, whole or homogenized, among subjects. There was, of course, controversy as is typical of lively and productive scientific debates to this day. The goals of this commentary are to briefly review the history of this work and how the terms referring to interactions among the components of the hypothalamo (as the central neural component was soon associated with)-pituitary-gonadal (HPG) axis evolved, and then to question if the current terms used have kept up with our understanding of the system. The focus in this review will be the actions of estradiol primarily upon the hypothalamus. Important actions of progesterone on the hypothalamus as well as both steroids on the pituitary response to hypothalamic factors are both acknowledged and largely ignored in this document, as are any sex differences as we focus on females.
Collapse
Affiliation(s)
- Suzanne M. Moenter
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
- Departments of Internal Medicine, Obstetrics & Gynecology, the Reproductive Sciences Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - J. Rudolph Starrett
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
21
|
Faure MC, Corona R, Roomans C, Lenfant F, Foidart JM, Cornil CA. Role of Membrane Estrogen Receptor Alpha on the Positive Feedback of Estrogens on Kisspeptin and GnRH Neurons. eNeuro 2024; 11:ENEURO.0271-23.2024. [PMID: 39375032 PMCID: PMC11520851 DOI: 10.1523/eneuro.0271-23.2024] [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: 08/02/2023] [Revised: 06/14/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024] Open
Abstract
Estrogens act through nuclear and membrane-initiated signaling. Estrogen receptor alpha (ERα) is critical for reproduction, but the relative contribution of its nuclear and membrane signaling to the central regulation of reproduction is unclear. To address this question, two complementary approaches were used: estetrol (E4) a natural estrogen acting as an agonist of nuclear ERs, but as an antagonist of their membrane fraction, and the C451A-ERα mouse lacking mERα. E4 dose- dependently blocks ovulation in female rats, but the central mechanism underlying this effect is unknown. To determine whether E4 acts centrally to control ovulation, its effect was tested on the positive feedback of estradiol (E2) on neural circuits underlying luteinizing hormone (LH) secretion. In ovariectomized females chronically exposed to a low dose of E2, estradiol benzoate (EB) alone or combined with progesterone (P) induced an increase in the number of kisspeptin (Kp) and gonadotropin-releasing hormone (GnRH) neurons coexpressing Fos, a marker of neuronal activation. E4 blocked these effects of EB, but not when combined to P. These results indicate that E4 blocked the central induction of the positive feedback in the absence of P, suggesting an antagonistic effect of E4 on mERα in the brain as shown in peripheral tissues. In parallel, as opposed to wild-type females, C451A-ERα females did not show the activation of Kp and GnRH neurons in response to EB unless they are treated with P. Together these effects support a role for membrane-initiated estrogen signaling in the activation of the circuit mediating the LH surge.
Collapse
Affiliation(s)
- Mélanie C. Faure
- Laboratory of Neuroendocrinology, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Rebeca Corona
- Laboratory of Neuroendocrinology, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Céline Roomans
- Laboratory of Neuroendocrinology, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Françoise Lenfant
- Institute of Metabolic and Cardiovascular Diseases (I2MC) Equipe 4, Inserm U1297-UPS, CHU, Toulouse 31432, France
| | - Jean-Michel Foidart
- Department of Obstetrics and Gynecology, University of Liège, Liège, Belgium
- Estetra SRL, Légiapark, Boulevard Patience et Beaujonc 3, 4000 Liège, Belgium
| | - Charlotte A. Cornil
- Laboratory of Neuroendocrinology, GIGA Neurosciences, University of Liège, Liège, Belgium
| |
Collapse
|
22
|
Coman S, Berean DI, Cimpean R, Ciupe S, Coman I, Bogdan LM. Clinical Modalities for Enhancing Reproductive Efficiency in Buffaloes: A Review and Practical Aspects for Veterinary Practitioners. Animals (Basel) 2024; 14:2642. [PMID: 39335232 PMCID: PMC11428391 DOI: 10.3390/ani14182642] [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: 07/22/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
This review aimed to bring a comprehensive analysis of key clinical strategies for enhancing reproductive efficiency in buffaloes, a species that exhibit low reproductive performance under conventional reproductive management compared to that exhibited by cattle. It considers key ART techniques including estrus synchronization for artificial insemination, and ovulation induction, highlighting their role in improving fertility and overall herd productivity. However, it also addresses common postpartum inflammatory and functional reproductive disorders, discussing their diagnosis and treatment protocols, stressing their impact on the overall reproductive outcome in buffalo farming.
Collapse
Affiliation(s)
- Stefan Coman
- Department of Reproduction, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Calea Manastur 3-5, 400372 Cluj-Napoca, Romania; (S.C.); (S.C.); (L.M.B.)
| | - Daniel Ionut Berean
- Department of Reproduction, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Calea Manastur 3-5, 400372 Cluj-Napoca, Romania; (S.C.); (S.C.); (L.M.B.)
| | - Raluca Cimpean
- Department of Animal Breeding and Food Safety, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Calea Manastur 3-5, 400372 Cluj-Napoca, Romania;
| | - Simona Ciupe
- Department of Reproduction, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Calea Manastur 3-5, 400372 Cluj-Napoca, Romania; (S.C.); (S.C.); (L.M.B.)
| | | | - Liviu Marian Bogdan
- Department of Reproduction, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Calea Manastur 3-5, 400372 Cluj-Napoca, Romania; (S.C.); (S.C.); (L.M.B.)
| |
Collapse
|
23
|
Coutinho EA, Esparza LA, Rodriguez J, Yang J, Schafer D, Kauffman AS. Targeted inhibition of kisspeptin neurons reverses hyperandrogenemia and abnormal hyperactive LH secretion in a preclinical mouse model of polycystic ovary syndrome. Hum Reprod 2024; 39:2089-2103. [PMID: 38978296 PMCID: PMC11373419 DOI: 10.1093/humrep/deae153] [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: 11/16/2023] [Revised: 06/11/2024] [Indexed: 07/10/2024] Open
Abstract
STUDY QUESTION Do hyperactive kisspeptin neurons contribute to abnormally high LH secretion and downstream hyperandrogenemia in polycystic ovary syndrome (PCOS)-like conditions and can inhibition of kisspeptin neurons rescue such endocrine impairments? SUMMARY ANSWER Targeted inhibition of endogenous kisspeptin neuron activity in a mouse model of PCOS reduced the abnormally hyperactive LH pulse secretion and hyperandrogenemia to healthy control levels. WHAT IS KNOWN ALREADY PCOS is a reproductive disorder characterized by hyperandrogenemia, anovulation, and/or polycystic ovaries, along with a hallmark feature of abnormal LH hyper-pulsatility, but the mechanisms underlying the endocrine impairments remain unclear. A chronic letrozole (LET; aromatase inhibitor) mouse model recapitulates PCOS phenotypes, including polycystic ovaries, anovulation, high testosterone, and hyperactive LH pulses. LET PCOS-like females also have increased hypothalamic kisspeptin neuronal activation which may drive their hyperactive LH secretion and hyperandrogenemia, but this has not been tested. STUDY DESIGN, SIZE, DURATION Transgenic KissCRE+/hM4Di female mice or littermates Cre- controls were treated with placebo, or chronic LET (50 µg/day) to induce a PCOS-like phenotype, followed by acute (once) or chronic (2 weeks) clozapine-N-oxide (CNO) exposure to chemogenetically inhibit kisspeptin cells (n = 6 to 10 mice/group). PARTICIPANTS/MATERIALS, SETTING, METHODS Key endocrine measures, including in vivo LH pulse secretion patterns and circulating testosterone levels, were assessed before and after selective kisspeptin neuron inhibition and compared between PCOS groups and healthy controls. Alterations in body weights were measured and pituitary and ovarian gene expression was determined by qRT-PCR. MAIN RESULTS AND THE ROLE OF CHANCE Acute targeted inhibition of kisspeptin neurons in PCOS mice successfully lowered the abnormally hyperactive LH pulse secretion (P < 0.05). Likewise, chronic selective suppression of kisspeptin neuron activity reversed the previously high LH and testosterone levels (P < 0.05) down to healthy control levels and rescued reproductive gene expression (P < 0. 05). LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Ovarian morphology was not assessed in this study. Additionally, mouse models can offer mechanistic insights into neuroendocrine processes in PCOS-like conditions but may not perfectly mirror PCOS in women. WIDER IMPLICATIONS OF THE FINDINGS These data support the hypothesis that overactive kisspeptin neurons can drive neuroendocrine PCOS-like impairments, and this may occur in PCOS women. Our findings complement recent clinical investigations using NKB receptor antagonists to lower LH in PCOS women and suggest that pharmacological dose-dependent modulation of kisspeptin neuron activity may be a valuable future therapeutic target to clinically treat hyperandrogenism and lower elevated LH in PCOS women. STUDY FUNDING/COMPETING INTEREST(S) This research was supported by NIH grants R01 HD111650, R01 HD090161, R01 HD100580, P50 HD012303, R01 AG078185, and NIH R24 HD102061, and a pilot project award from the British Society for Neuroendocrinology. There are no competing interests.
Collapse
Affiliation(s)
- Eulalia A Coutinho
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Lourdes A Esparza
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Julian Rodriguez
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jason Yang
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Danielle Schafer
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Alexander S Kauffman
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
24
|
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
|
25
|
Villa PA, Ruggiero-Ruff RE, Jamieson BB, Campbell RE, Coss D. Obesity Alters POMC and Kisspeptin Neuron Cross Talk Leading to Reduced Luteinizing Hormone in Male Mice. J Neurosci 2024; 44:e0222242024. [PMID: 38744532 PMCID: PMC11236585 DOI: 10.1523/jneurosci.0222-24.2024] [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: 02/01/2024] [Revised: 04/19/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Obesity is associated with hypogonadism in males, characterized by low testosterone and sperm number. Previous studies determined that these stem from dysregulation of hypothalamic circuitry that regulates reproduction, by unknown mechanisms. Herein, we used mice fed chronic high-fat diet, which mimics human obesity, to determine mechanisms of impairment at the level of the hypothalamus, in particular gonadotropin-releasing hormone (GnRH) neurons that regulate luteinizing hormone (LH), which then regulates testosterone. Consistent with obese humans, we demonstrated lower LH, and lower pulse frequency of LH secretion, but unchanged pituitary responsiveness to GnRH. LH pulse frequency is regulated by pulsatile GnRH secretion, which is controlled by kisspeptin. Peripheral and central kisspeptin injections, and DREADD-mediated activation of kisspeptin neurons, demonstrated that kisspeptin neurons were suppressed in obese mice. Thus, we investigated regulators of kisspeptin secretion. We determined that the LH response to NMDA was lower in obese mice, corresponding to fewer glutamate receptors in kisspeptin neurons, which may be critical for kisspeptin synchronization. Given that kisspeptin neurons also interact with anorexigenic POMC neurons, which are affected by obesity, we examined their cross talk, and determined that the LH response to either DREADD-mediated activation of POMC neurons or central injection of αMSH, a product of POMC, is abolished in obese mice. This was accompanied by diminished levels of αMSH receptor, MC4R, in kisspeptin neurons. Together, our studies determined that obesity leads to the downregulation of receptors that regulate kisspeptin neurons, which is associated with lower LH pulse frequency, leading to lower LH and hypogonadism.
Collapse
Affiliation(s)
- Pedro A Villa
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California 92521
| | - Rebecca E Ruggiero-Ruff
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California 92521
| | - Bradley B Jamieson
- Centre for Neuroendocrinology, and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California 92521
| |
Collapse
|
26
|
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
|
27
|
Nakamura S, Sasaki T, Uenoyama Y, Inoue N, Nakanishi M, Yamada K, Morishima A, Suzumura R, Kitagawa Y, Morita Y, Ohkura S, Tsukamura H. Raphe glucose-sensing serotonergic neurons stimulate KNDy neurons to enhance LH pulses via 5HT2CR: rat and goat studies. Sci Rep 2024; 14:10190. [PMID: 38702366 PMCID: PMC11068885 DOI: 10.1038/s41598-024-58470-4] [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: 11/02/2023] [Accepted: 03/29/2024] [Indexed: 05/06/2024] Open
Abstract
Dysfunction of central serotonergic neurons is known to cause depressive disorders in humans, who often show reproductive and/or glucose metabolism disorders. This study examined whether dorsal raphe (DR) serotonergic neurons sense high glucose availability to upregulate reproductive function via activating hypothalamic arcuate (ARC) kisspeptin neurons (= KNDy neurons), a dominant stimulator of gonadotropin-releasing hormone (GnRH)/gonadotropin pulses, using female rats and goats. RNA-seq and histological analysis revealed that stimulatory serotonin-2C receptor (5HT2CR) was mainly expressed in the KNDy neurons in female rats. The serotonergic reuptake inhibitor administration into the mediobasal hypothalamus (MBH), including the ARC, significantly blocked glucoprivic suppression of luteinizing hormone (LH) pulses and hyperglycemia induced by intravenous 2-deoxy-D-glucose (2DG) administration in female rats. A local infusion of glucose into the DR significantly increased in vivo serotonin release in the MBH and partly restored LH pulses and hyperglycemia in the 2DG-treated female rats. Furthermore, central administration of serotonin or a 5HT2CR agonist immediately evoked GnRH pulse generator activity, and central 5HT2CR antagonism blocked the serotonin-induced facilitation of GnRH pulse generator activity in ovariectomized goats. These results suggest that DR serotonergic neurons sense high glucose availability to reduce gluconeogenesis and upregulate reproductive function by activating GnRH/LH pulse generator activity in mammals.
Collapse
Affiliation(s)
- Sho Nakamura
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, 470-0151, Japan
| | - Takuya Sasaki
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, 470-0151, Japan
| | - Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Marina Nakanishi
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Koki Yamada
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Ai Morishima
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, 470-0151, Japan
| | - Reika Suzumura
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, 470-0151, Japan
| | - Yuri Kitagawa
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, 470-0151, Japan
| | - Yasuhiro Morita
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, 470-0151, Japan
| | - Satoshi Ohkura
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, 470-0151, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan.
| |
Collapse
|
28
|
Cox JF, Carrasco A, Navarrete F, Bocic A, Saravia F, Dorado J. A Subovulatory Dose of Human Chorionic Gonadotropin (hCG) May Sustain Terminal Follicle Development and Reproductive Efficiency during Anestrus in Sheep. Animals (Basel) 2024; 14:1096. [PMID: 38612335 PMCID: PMC11011159 DOI: 10.3390/ani14071096] [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: 01/25/2024] [Revised: 03/02/2024] [Accepted: 03/15/2024] [Indexed: 04/14/2024] Open
Abstract
The study tested the hypothesis that a single administration of hCG supports the LH-dependent phase of terminal follicular development in synchronized sheep during anestrus, using eCG as a functional reference. Using a clinical approach, four experiments were designed to achieve the following: (1) Identify the inhibitory influence of anestrus on reproduction efficiency; (2) Assess the potential of hCG to keep functional blood concentrations after a single dose; (3) Characterize the effect of different doses of hCG on reproductive functional markers; (4) To compare the ability of hCG to that of eCG to support follicular development and fertility based on the same markers. The results showed that anestrus seems to affect follicular and luteal function under LH dependency as FSH-dependent markers are not compromised; hCG maintains higher blood concentrations than controls for at least 48 h; hCG improves follicular development and ovulatory rates compared to controls and at standards comparable to a breeding season; and ewes treated with hCG exhibit similar performance to those treated with eCG. Our results conclude that hCG can be used to support follicular function during anestrus in sheep, aiming to perfect its regulation in assisted reproduction.
Collapse
Affiliation(s)
- José Francisco Cox
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Vicente Méndez 595, Chillán 3780000, Chile (A.B.); (F.S.)
| | - Albert Carrasco
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Vicente Méndez 595, Chillán 3780000, Chile (A.B.); (F.S.)
| | - Felipe Navarrete
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Vicente Méndez 595, Chillán 3780000, Chile (A.B.); (F.S.)
| | - Antonio Bocic
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Vicente Méndez 595, Chillán 3780000, Chile (A.B.); (F.S.)
| | - Fernando Saravia
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Vicente Méndez 595, Chillán 3780000, Chile (A.B.); (F.S.)
| | - Jesús Dorado
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, University of Cordoba, Campus Rabanales, 14014 Córdoba, Spain
| |
Collapse
|
29
|
Cotellessa L, Giacobini P. Role of Anti-Müllerian Hormone in the Central Regulation of Fertility. Semin Reprod Med 2024; 42:34-40. [PMID: 38608673 DOI: 10.1055/s-0044-1786050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
In recent years, the expanding roles of anti-Müllerian hormone (AMH) in various aspects of reproductive health have attracted significant attention. Initially recognized for its classical role in male sexual differentiation, AMH is produced postnatally by the Sertoli cells in the male testes and by the granulosa cells in the female ovaries. Traditionally, it was believed to primarily influence gonadal development and function. However, research over the last decade has unveiled novel actions of AMH beyond the gonads, specifically all along the hypothalamic-pituitary-gonadal axis. This review will focus on the emerging roles of AMH within the hypothalamus and discusses its potential implications in reproductive physiology. Additionally, recent preclinical and clinical studies have suggested that elevated levels of AMH may disrupt the hypothalamic network regulating reproduction, which could contribute to the central pathophysiology of polycystic ovary syndrome. These findings underscore the intricate interplay between AMH and the neuroendocrine system, offering new avenues for understanding the mechanisms underlying fertility and reproductive disorders.
Collapse
Affiliation(s)
- Ludovica Cotellessa
- Inserm, CHU Lille, Unit 1172, Lille Neuroscience & Cognition (LilNCog), University of Lille, Lille, France
| | - Paolo Giacobini
- Inserm, CHU Lille, Unit 1172, Lille Neuroscience & Cognition (LilNCog), University of Lille, Lille, France
| |
Collapse
|
30
|
Vas S, Wall E, Zhou Z, Kalmar L, Han SY, Herbison AE. Long-term Recordings of Arcuate Nucleus Kisspeptin Neurons Across the Mouse Estrous Cycle. Endocrinology 2024; 165:bqae009. [PMID: 38279940 PMCID: PMC10873703 DOI: 10.1210/endocr/bqae009] [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: 11/20/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 01/29/2024]
Abstract
The arcuate nucleus kisspeptin (ARNKISS) neurons represent the GnRH pulse generator that likely drives pulsatile gonadotropin secretion in all mammals. Using an improved GCaMP fiber photometry system enabling long-term continuous recordings, we aimed to establish a definitive profile of ARNKISS neuronal activity across the murine estrous cycle. As noted previously, a substantial reduction in the frequency of ARNKISS neuron synchronization events (SEs) occurs on late proestrus and extends into estrus. The SE amplitude remains constant throughout the cycle. During metestrus, we unexpectedly detected many multipeak SEs where many SEs occurred rapidly, within 160 seconds of each other. By applying a machine learning-based, k-means clustering analysis, we were further able to detect substantial within-stage variability in the patterns of pulse generator activity. Estrous cycle-dependent changes in SE activity occurred around the time of lights on and off. We also find that a mild stressor such as vaginal lavage reduces ARNKISS neuron SE frequency for up to 3 hours. These observations provide a comprehensive account of ARNKISS neuron activity across the estrous cycle, highlight a new pattern of multipeak SE activity, and introduce a new k-means clustering approach for analyzing ARNKISS neuron population behavior.
Collapse
Affiliation(s)
- Szilvia Vas
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Ellen Wall
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Ziyue Zhou
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Lajos Kalmar
- Department of Toxicology, University of Cambridge, Cambridge CB2 1QR, UK
| | - Su Young Han
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Allan E Herbison
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| |
Collapse
|
31
|
Kunimura Y, Iwata K, Ishii H, Ozawa H. Chronic estradiol exposure suppresses luteinizing hormone surge without affecting kisspeptin neurons and estrogen receptor alpha in anteroventral periventricular nucleus†. Biol Reprod 2024; 110:90-101. [PMID: 37774351 DOI: 10.1093/biolre/ioad129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/01/2023] Open
Abstract
Mammalian ovulation is induced by a luteinizing hormone surge, which is triggered by elevated plasma estrogen levels; however, chronic exposure to high levels of estradiol is known to inhibit luteinizing hormone secretion. In the present study, we hypothesized that the inhibition of the luteinizing hormone surge by chronic estradiol exposure is due to the downregulation of the estrogen receptor alpha in kisspeptin neurons at hypothalamic anteroventral periventricular nucleus, which is known as the gonadotropin-releasing hormone/luteinizing hormone surge generator. Animals exposed to estradiol for 2 days showed an luteinizing hormone surge, whereas those exposed for 14 days showed a significant suppression of luteinizing hormone. Chronic estradiol exposure did not affect the number of kisspeptin neurons and the percentage of kisspeptin neurons with estrogen receptor alpha or c-Fos in anteroventral periventricular nucleus, but it did affect the number of kisspeptin neurons in arcuate nucleus. Furthermore, chronic estradiol exposure did not affect gonadotropin-releasing hormone neurons. In the pituitary, 14-day estradiol exposure significantly reduced the expression of Lhb mRNA and LHβ-immunoreactive areas. Gonadotropin-releasing hormone-induced luteinizing hormone release was also reduced significantly by 14-day estradiol exposure. We revealed that the suppression of an luteinizing hormone surge by chronic estradiol exposure was induced in association with the significant reduction in kisspeptin neurons in arcuate nucleus, luteinizing hormone expression in the pituitary, and pituitary responsiveness to gonadotropin-releasing hormone, and this was not caused by changes in the estrogen receptor alpha-expressing kisspeptin neurons in anteroventral periventricular nucleus and gonadotropin-releasing hormone neurons, which are responsible for estradiol positive feedback.
Collapse
Affiliation(s)
- Yuyu Kunimura
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Kinuyo Iwata
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Hirotaka Ishii
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Hitoshi Ozawa
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
- Department of Physical Therapy, Faculty of Health Science, Bukkyo University, Kyoto, Japan
| |
Collapse
|
32
|
Yu J, Li XF, Tsaneva-Atanasova K, Zavala E, O’Byrne KT. Chemogenetic activation of PVN CRH neurons disrupts the estrous cycle and LH dynamics in female mice. Front Endocrinol (Lausanne) 2024; 14:1322662. [PMID: 38264285 PMCID: PMC10803550 DOI: 10.3389/fendo.2023.1322662] [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: 10/16/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024] Open
Abstract
Introduction The impact of stress on reproductive function is significant. Hypothalamic paraventricular nucleus (PVN) corticotrophin-releasing hormone (CRH) plays a major role in regulating the stress response. Understanding how the hypothalamic-pituitary-adrenal (HPA) axis and the hypothalamic-pituitary-gonadal (HPG) axis interact is crucial for comprehending how stress can lead to reproductive dysfunction. However, whether stress influences reproductive function via modulating PVN CRH or HPA sequelae is not fully elucidated. Methods In this study, we investigated the impact of chemogenetic activation of PVN CRH neurons on reproductive function. We chronically and selectively stimulated PVN CRH neurons in female CRH-Cre mice using excitatory designer receptor exclusively activated by designer drugs (DREADDs) viral constructs, which were bilaterally injected into the PVN. The agonist compound-21 (C21) was delivered through the drinking water. We determined the effects of DREADDs activation of PVN CRH neurons on the estrous cycles, LH pulse frequency in diestrus and metestrus and LH surge in proestrus mice. The effect of long-term C21 administration on basal corticosterone secretion and the response to acute restraint stress during metestrus was also examined. Additionally, computer simulations of a mathematical model were used to determine the effects of DREADDs activation of PVN CRH neurons, simulating chronic stress, on the physiological parameters examined experimentally. Results As a result, and consistent with our mathematical model predictions, the length of the estrous cycle was extended, with an increase in the time spent in estrus and metestrus, and a decrease in proestrus and diestrus. Additionally, the frequency of LH pulses during metestrus was decreased, but unaffected during diestrus. The occurrence of the preovulatory LH surge during proestrus was disrupted. The basal level of corticosterone during metestrus was not affected, but the response to acute restraint stress was diminished after long-term C21 application. Discussion These data suggest that PVN CRH neurons play a functional role in disrupting ovarian cyclicity and the preovulatory LH surge, and that the activity of the GnRH pulse generator remains relatively robust during diestrus but not during metestrus under chronic stress exposure in accordance with our mathematical model predictions.
Collapse
Affiliation(s)
- Junru Yu
- Department of Women and Children’s Health, School of Life Course and Population Sciences, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
| | - Xiao-Feng Li
- Department of Women and Children’s Health, School of Life Course and Population Sciences, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics and Statistics, Faculty of Environment, Science and Economy, University of Exeter, Exeter, United Kingdom
- Living Systems Institute, University of Exeter, Exeter, United Kingdom
| | - Eder Zavala
- Centre for Systems Modelling and Quantitative Biomedicine, University of Birmingham, Edgbaston, United Kingdom
| | - Kevin T. O’Byrne
- Department of Women and Children’s Health, School of Life Course and Population Sciences, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
33
|
Clarkson J, Yip SH, Porteous R, Kauff A, Heather AK, Herbison AE. CRISPR-Cas9 knockdown of ESR1 in preoptic GABA-kisspeptin neurons suppresses the preovulatory surge and estrous cycles in female mice. eLife 2023; 12:RP90959. [PMID: 38126277 PMCID: PMC10735218 DOI: 10.7554/elife.90959] [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: 12/23/2023] Open
Abstract
Evidence suggests that estradiol-sensing preoptic area GABA neurons are involved in the preovulatory surge mechanism necessary for ovulation. In vivo CRISPR-Cas9 editing was used to achieve a 60-70% knockdown in estrogen receptor alpha (ESR1) expression by GABA neurons located within the regions of the rostral periventricular area of the third ventricle (RP3V) and medial preoptic nuclei (MPN) in adult female mice. Mice exhibited variable reproductive phenotypes with the only significant finding being mice with bilateral ESR1 deletion in RP3V GABA neurons having reduced cFos expression in gonadotropin-releasing hormone (GnRH) neurons at the time of the surge. One sub-population of RP3V GABA neurons expresses kisspeptin. Re-grouping ESR1-edited mice on the basis of their RP3V kisspeptin expression revealed a highly consistent phenotype; mice with a near-complete loss of kisspeptin immunoreactivity displayed constant estrus and failed to exhibit surge activation but retained pulsatile luteinizing hormone (LH) secretion. These observations demonstrate that ESR1-expressing GABA-kisspeptin neurons in the RP3V are essential for the murine preovulatory LH surge mechanism.
Collapse
Affiliation(s)
- Jenny Clarkson
- Centre for NeuroendocrinologyDunedinNew Zealand
- Department of Physiology, University of Otago School of Biomedical SciencesDunedinNew Zealand
| | - Siew Hoong Yip
- Centre for NeuroendocrinologyDunedinNew Zealand
- Department of Physiology, University of Otago School of Biomedical SciencesDunedinNew Zealand
| | - Robert Porteous
- Centre for NeuroendocrinologyDunedinNew Zealand
- Department of Physiology, University of Otago School of Biomedical SciencesDunedinNew Zealand
| | - Alexia Kauff
- Department of Physiology, University of Otago School of Biomedical SciencesDunedinNew Zealand
| | - Alison K Heather
- Department of Physiology, University of Otago School of Biomedical SciencesDunedinNew Zealand
| | - Allan E Herbison
- Centre for NeuroendocrinologyDunedinNew Zealand
- Department of Physiology, University of Otago School of Biomedical SciencesDunedinNew Zealand
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
34
|
Panda SP, Kesharwani A, Singh GD, Prasanth D, Vatchavai BR, Kumari PVK, Panda SK, Mallick SP. Impose of KNDy/GnRH neural circuit in PCOS, ageing, cancer and Alzheimer's disease: StAR actions in prevention of neuroendocrine dysfunction. Ageing Res Rev 2023; 92:102086. [PMID: 37821047 DOI: 10.1016/j.arr.2023.102086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/06/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
The Kisspeptin1 (KISS1)/neurokinin B (NKB)/Dynorphin (Dyn) [KNDy] neurons in the hypothalamus regulate the reproduction stage in human beings and rodents. KNDy neurons co-expressed all KISS1, NKB, and Dyn peptides, and hence commonly regarded as KISS1 neurons. KNDy neurons contribute to the "GnRH pulse generator" and are implicated in the regulation of pulsatile GnRH release. The estradiol (E2)-estrogen receptor (ER) interactions over GnRH neurons in the hypothalamus cause nitric oxide (NO) discharge, in addition to presynaptic GABA and glutamate discharge from respective neurons. The released GABA and glutamate facilitate the activity of GnRH neurons via GABAA-R and AMPA/kainate-R. The KISS1 stimulates MAPK/ERK1/2 signaling and cause the release of Ca2+ from intracellular store, which contribute to neuroendocrine function, increase apoptosis and decrease cell proliferation and metastasis. The ageing in women deteriorates KISS1/KISS1R interaction in the hypothalamus which causes lower levels of GnRH. Because examining the human brain is so challenging, decades of clinical research have failed to find the causes of KNDy/GnRH dysfunction. The KISS1/KISS1R interactions in the brain have a neuroprotective effect against Alzheimer's disease (AD). These findings modulate the pathophysiological role of the KNDy/GnRH neural network in polycystic ovarian syndrome (PCOS) associated with ageing and, its protective role in cancer and AD. This review concludes with protecting effect of the steroid-derived acute regulatory enzyme (StAR) against neurotoxicity in the hippocampus, and hypothalamus, and these measures are fundamental for delaying ageing with PCOS. StAR could serve as novel diagnostic marker and therapeutic target for the most prevalent hormone-sensitive breast cancers (BCs).
Collapse
Affiliation(s)
- Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| | - Adarsh Kesharwani
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | | | - Dsnbk Prasanth
- KVSR Siddhartha College of Pharmaceutical Sciences, Vijayawada, Andhrapradesh, India
| | - Bhaskara Raju Vatchavai
- Sri Vasavi Institute of Pharmaceutical Sciences, Pedatadepalli, Tadepalligudem, Andhrapradesh, India
| | - P V Kamala Kumari
- Vignan Institute of Pharmaceutical Technology, Duvvada, Visakhapatnam, Andhrapradesh, India
| | | | | |
Collapse
|
35
|
Nagae M, Yamada K, Enomoto Y, Kometani M, Tsuchida H, Panthee A, Nonogaki M, Matsunaga N, Takizawa M, Matsuzaki S, Hirabayashi M, Inoue N, Tsukamura H, Uenoyama Y. Conditional Oprk1-dependent Kiss1 deletion in kisspeptin neurons caused estrogen-dependent LH pulse disruption and LH surge attenuation in female rats. Sci Rep 2023; 13:20495. [PMID: 37993510 PMCID: PMC10665460 DOI: 10.1038/s41598-023-47222-5] [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: 07/05/2023] [Accepted: 11/10/2023] [Indexed: 11/24/2023] Open
Abstract
The gonadotropin-releasing hormone (GnRH) pulse and surge are considered to be generated by arcuate kisspeptin/neurokinin B/dynorphin A (KNDy) neurons and anteroventral periventricular nucleus (AVPV) kisspeptin neurons, respectively, in female rodents. The majority of KNDy and AVPV kisspeptin neurons express κ-opioid receptors (KORs, encoded by Oprk1) in female rodents. Thus, this study aimed to investigate the effect of a conditional Oprk1-dependent Kiss1 deletion in kisspeptin neurons on the luteinizing hormone (LH) pulse/surge and fertility using Kiss1-floxed/Oprk1-Cre rats, in which Kiss1 was deleted in cells expressing or once expressed the Oprk1/Cre. The Kiss1-floxed/Oprk1-Cre female rats, with Kiss1 deleted in a majority of KNDy neurons, showed normal puberty while having a one-day longer estrous cycle and fewer pups than Kiss1-floxed controls. Notably, ovariectomized (OVX) Kiss1-floxed/Oprk1-Cre rats showed profound disruption of LH pulses in the presence of a diestrous level of estrogen but showed apparent LH pulses without estrogen treatment. Furthermore, Kiss1-floxed/Oprk1-Cre rats, with Kiss1 deleted in approximately half of AVPV kisspeptin neurons, showed a lower peak of the estrogen-induced LH surge than controls. These results suggest that arcuate and AVPV kisspeptin neurons expressing or having expressed Oprk1 have a role in maintaining normal GnRH pulse and surge generation, the normal length of the estrous cycle, and the normal offspring number in female rats.
Collapse
Affiliation(s)
- Mayuko Nagae
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
- Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi, 444-8787, Japan
| | - Koki Yamada
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Yuki Enomoto
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Mari Kometani
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Hitomi Tsuchida
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Arvinda Panthee
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Miku Nonogaki
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Nao Matsunaga
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Marina Takizawa
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Sena Matsuzaki
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Masumi Hirabayashi
- Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi, 444-8787, Japan
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan.
| | - Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan.
| |
Collapse
|
36
|
Morris PG, Herbison AE. Mechanism of Arcuate Kisspeptin Neuron Synchronization in Acute Brain Slices From Female Mice. Endocrinology 2023; 164:bqad167. [PMID: 37936337 PMCID: PMC10652333 DOI: 10.1210/endocr/bqad167] [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: 08/01/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023]
Abstract
The mechanism by which arcuate kisspeptin (ARNKISS) neurons co-expressing glutamate, neurokinin B, and dynorphin intermittently synchronize their activity to drive pulsatile hormone secretion remains unclear in females. In order to study spontaneous synchronization within the ARNKISS neuron network, acute brain slices were prepared from adult female Kiss1-GCaMP6 mice. Analysis of both spontaneous synchronizations and those driven by high frequency stimulation of individual ARNKISS neurons revealed that the network exhibits semi-random emergent excitation dependent upon glutamate signaling through AMPA receptors. No role for NMDA receptors was identified. In contrast to male mice, ongoing tachykinin receptor tone within the slice operated to promote spontaneous synchronizations in females. As previously observed in males, we found that ongoing dynorphin transmission in the slice did not contribute to synchronization events. These observations indicate that a very similar AMPA receptor-dependent mechanism underlies ARNKISS neuron synchronizations in the female mouse supporting the "glutamate two-transition" model for kisspeptin neuron synchronization. However, a potentially important sex difference appears to exist with a more prominent facilitatory role for tachykinin transmission in the female.
Collapse
Affiliation(s)
- Paul G Morris
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Allan E Herbison
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| |
Collapse
|
37
|
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
|
38
|
Buo C, Bearss RJ, Novak AG, Anello AE, Dakin JJ, Piet R. Serotonin stimulates female preoptic area kisspeptin neurons via activation of type 2 serotonin receptors in mice. Front Endocrinol (Lausanne) 2023; 14:1212854. [PMID: 37900129 PMCID: PMC10602649 DOI: 10.3389/fendo.2023.1212854] [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: 04/27/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023] Open
Abstract
Background The neuroendocrine control of ovulation is orchestrated by neuronal circuits that ultimately drive the release of gonadotropin-releasing hormone (GnRH) from the hypothalamus to trigger the preovulatory surge in luteinizing hormone (LH) secretion. While estrogen feedback signals are determinant in triggering activation of GnRH neurons, through stimulation of afferent kisspeptin neurons in the rostral periventricular area of the third ventricle (RP3VKISS1 neurons), many neuropeptidergic and classical neurotransmitter systems have been shown to regulate the LH surge. Among these, several lines of evidence indicate that the monoamine neurotransmitter serotonin (5-HT) has an excitatory, permissive, influence over the generation of the surge, via activation of type 2 5-HT (5-HT2) receptors. The mechanisms through which this occurs, however, are not well understood. We hypothesized that 5-HT exerts its influence on the surge by stimulating RP3VKISS1 neurons in a 5-HT2 receptor-dependent manner. Methods We tested this using kisspeptin neuron-specific calcium imaging and electrophysiology in brain slices obtained from male and female mice. Results We show that exogenous 5-HT reversibly increases the activity of the majority of RP3VKISS1 neurons. This effect is more prominent in females than in males, is likely mediated directly at RP3VKISS1 neurons and requires activation of 5-HT2 receptors. The functional impact of 5-HT on RP3VKISS1 neurons, however, does not significantly vary during the estrous cycle. Conclusion Taken together, these data suggest that 5-HT2 receptor-mediated stimulation of RP3VKISS1 neuron activity might be involved in mediating the influence of 5-HT on the preovulatory LH surge.
Collapse
Affiliation(s)
- Carrie Buo
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Robin J. Bearss
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Alyssa G. Novak
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Anna E. Anello
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Jordan J. Dakin
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Richard Piet
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| |
Collapse
|
39
|
Goodman RL, Moore AM, Onslow K, Hileman SM, Hardy SL, Bowdridge EC, Walters BA, Agus S, Griesgraber MJ, Aerts EG, Lehman MN, Coolen LM. Lesions of KNDy and Kiss1R Neurons in the Arcuate Nucleus Produce Different Effects on LH Pulse Patterns in Female Sheep. Endocrinology 2023; 164:bqad148. [PMID: 37776515 PMCID: PMC10587900 DOI: 10.1210/endocr/bqad148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/02/2023]
Abstract
The current model for the synchronization of GnRH neural activity driving GnRH and LH pulses proposes that a set of arcuate (ARC) neurons that contain kisspeptin, neurokinin B, and dynorphin (KNDy neurons) is the GnRH pulse generator. This study tested the functional role of ovine KNDy neurons in pulse generation and explored the roles of nearby Kiss1 receptor (Kiss1R)-containing cells using lesions produced with saporin (SAP) conjugates. Injection of NK3-SAP ablated over 90% of the KNDy cells, while Kiss-SAP (saporin conjugated to kisspeptin-54) lesioned about two-thirds of the Kiss1R population without affecting KNDy or GnRH cell number. Both lesions produced a dramatic decrease in LH pulse amplitude but had different effects on LH pulse patterns. NK3-SAP increased interpulse interval, but Kiss-SAP did not. In contrast, Kiss-SAP disrupted the regular hourly occurrence of LH pulses, but NK3-SAP did not. Because Kiss1R is not expressed in KNDy cells, HiPlex RNAScope was used to assess the colocalization of 8 neurotransmitters and 3 receptors in ARC Kiss1R-containing cells. Kiss1R cells primarily contained transcript markers for GABA (68%), glutamate (28%), ESR1 (estrogen receptor-α) mRNA, and OPRK1 (kappa opioid receptor) mRNA. These data support the conclusion that KNDy neurons are essential for GnRH pulses in ewes, whereas ARC Kiss1R cells are not but do maintain the amplitude and regularity of GnRH pulses. We thus propose that in sheep, ARC Kiss1R neurons form part of a positive feedback circuit that reinforces the activity of the KNDy neural network, with GABA or glutamate likely being involved.
Collapse
Affiliation(s)
- Robert L Goodman
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, USA
| | - Aleisha M Moore
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Kayla Onslow
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Stanley M Hileman
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, USA
| | - Steve L Hardy
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, USA
| | - Elizabeth C Bowdridge
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, USA
| | - Burgundy A Walters
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Sami Agus
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Max J Griesgraber
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, USA
| | - Eliana G Aerts
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, USA
| | - Michael N Lehman
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Lique M Coolen
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
40
|
Prévot V, Duittoz A. A role for GnRH in olfaction and cognition: Implications for veterinary medicine. Reprod Domest Anim 2023; 58 Suppl 2:109-124. [PMID: 37329313 DOI: 10.1111/rda.14411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023]
Abstract
Pulsatile secretion of gonadotropin-releasing hormone (GnRH) is essential for the activation and maintenance of the function of the hypothalamic-pituitary-gonadal (HPG) axis, which controls the onset of puberty and fertility. Two provocative recent studies suggest that, in addition to control reproduction, the neurons in the brain that produce GnRH are also involved in the control postnatal brain maturation, odour discrimination and adult cognition. Long-acting GnRH antagonists and agonists are commonly used to control fertility and behaviour in veterinary medicine, primarily in males. This review puts into perspective the potential risks of these androgen deprivation therapies and immunization on olfactory and cognitive performances and well-aging in domestic animals, including pets. We will also discuss the results reporting beneficial effects of pharmacological interventions restoring physiological GnRH levels on olfactory and cognitive alterations in preclinical models of Alzheimer's disease, which shares many pathophysiological and behavioural hallmarks with canine cognitive dysfunction. These novel findings raise the intriguing possibility that pulsatile GnRH therapy holds therapeutic potential for the management of this behavioural syndrome affecting older dogs.
Collapse
Affiliation(s)
- Vincent Prévot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, Lille, France
| | - Anne Duittoz
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRAE Val de Loire, IFCE, Université de Tours, Nouzilly, France
| |
Collapse
|
41
|
Uenoyama Y, Tsukamura H. KNDy neurones and GnRH/LH pulse generation: Current understanding and future aspects. J Neuroendocrinol 2023; 35:e13285. [PMID: 37232103 DOI: 10.1111/jne.13285] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/31/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023]
Abstract
Uncovering the central mechanism underlying mammalian reproduction is warranted to develop new therapeutic approaches for reproductive disorders in humans and domestic animals. The present study focused on the role of arcuate kisspeptin neurones (also known as KNDy neurones) as an intrinsic gonadotropin-releasing hormone (GnRH) pulse generator, which plays a fundamental role in mammalian reproduction via the stimulation of pituitary gonadotropin synthesis and release and thereby in gametogenesis and steroidogenesis in the gonads of mammals. We also discuss the mechanism that inhibits pulsatile GnRH/gonadotropin release under a negative energy balance, considering that reproductive disorders often occur during malnutrition in humans and livestock.
Collapse
Affiliation(s)
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Nagoya University, Nagoya, Japan
| |
Collapse
|
42
|
Uenoyama Y, Inoue N, Tsukamura H. Kisspeptin and lactational anestrus: Current understanding and future prospects. Peptides 2023; 166:171026. [PMID: 37230188 DOI: 10.1016/j.peptides.2023.171026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 05/27/2023]
Abstract
Lactational anestrus, characterized by the suppression of pulsatile gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) release, would be a strategic adaptation to ensure survival by avoiding pregnancy during lactation in mammals. In the present article, we first provide a current understanding of the central regulation of reproduction in mammals, i.e., a fundamental role of arcuate kisspeptin neurons in mammalian reproduction by driving GnRH/LH pulses. Second, we discuss the central mechanism inhibiting arcuate Kiss1 (encoding kisspeptin) expression and GnRH/LH pulses during lactation with a focus on suckling stimulus, negative energy balance due to milk production, and the role of circulating estrogen in rats. We also discuss upper regulators that control arcuate kisspeptin neurons in rats during the early and late lactation periods based on the findings obtained by a lactating rat model. Finally, we discuss potential reproductive technology for the improvement of reproductive performance in milking cows.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan.
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
43
|
Silva MSB, Navarro VM, Chachlaki K. Editorial: The neuroendocrine female brain: from normal reproductive function to disease. Front Neurosci 2023; 17:1243349. [PMID: 37521681 PMCID: PMC10374340 DOI: 10.3389/fnins.2023.1243349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 08/01/2023] Open
Affiliation(s)
- Mauro S. B. Silva
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Victor M. Navarro
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Konstantina Chachlaki
- Université de Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille, France
- FHU 1000 Days for Health, School of Medicine, Lille, France
- University Research Institute of Child Health and Precision Medicine, National and Kapodistrian University of Athens, “Aghia Sophia” Children's Hospital, Athens, Greece
| |
Collapse
|
44
|
Vaudry H, Schoofs L, Civelli O, Kojima M. Editorial: Neuropeptide GPCRs in neuroendocrinology, Volume II. Front Endocrinol (Lausanne) 2023; 14:1219530. [PMID: 37415662 PMCID: PMC10321770 DOI: 10.3389/fendo.2023.1219530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/25/2023] [Indexed: 07/08/2023] Open
Affiliation(s)
- Hubert Vaudry
- Institute of Biomedical Research and Innovation, University of Rouen Normandy, Mont-Saint-Aignan, France
| | | | - Olivier Civelli
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
| | - Masayasu Kojima
- Institute of Life Science, Kurume University, Fukuoka, Japan
| |
Collapse
|
45
|
Abstract
OBJECTIVE To update the evidence-based Nonhormonal Management of Menopause-Associated Vasomotor Symptoms: 2015 Position Statement of The North American Menopause Society. METHODS An advisory panel of clinicians and research experts in women's health were selected to review and evaluate the literature published since the Nonhormonal Management of Menopause-Associated Vasomotor Symptoms: 2015 Position Statement of The North American Menopause Society. Topics were divided into five sections for ease of review: lifestyle; mind-body techniques; prescription therapies; dietary supplements; and acupuncture, other treatments, and technologies. The panel assessed the most current and available literature to determine whether to recommend or not recommend use based on these levels of evidence: Level I, good and consistent scientific evidence; Level II, limited or inconsistent scientific evidence, and Level III, consensus and expert opinion. RESULTS Evidence-based review of the literature resulted in several nonhormone options for the treatment of vasomotor symptoms. Recommended: Cognitive-behavioral therapy, clinical hypnosis, selective serotonin reuptake inhibitors/serotonin-norepinephrine reuptake inhibitors, gabapentin, fezolinetant (Level I); oxybutynin (Levels I-II); weight loss, stellate ganglion block (Levels II-III). Not recommended: Paced respiration (Level I); supplements/herbal remedies (Levels I-II); cooling techniques, avoiding triggers, exercise, yoga, mindfulness-based intervention, relaxation, suvorexant, soy foods and soy extracts, soy metabolite equol, cannabinoids, acupuncture, calibration of neural oscillations (Level II); chiropractic interventions, clonidine; (Levels I-III); dietary modification and pregabalin (Level III). CONCLUSION Hormone therapy remains the most effective treatment for vasomotor symptoms and should be considered in menopausal women within 10 years of their final menstrual periods. For women who are not good candidates for hormone therapy because of contraindications (eg, estrogen-dependent cancers or cardiovascular disease) or personal preference, it is important for healthcare professionals to be well informed about nonhormone treatment options for reducing vasomotor symptoms that are supported by the evidence.
Collapse
|
46
|
Sokanovic SJ, Constantin S, Lamarca Dams A, Mochimaru Y, Smiljanic K, Bjelobaba I, Prévide RM, Stojilkovic SS. Common and female-specific roles of protein tyrosine phosphatase receptors N and N2 in mice reproduction. Sci Rep 2023; 13:355. [PMID: 36611058 PMCID: PMC9825377 DOI: 10.1038/s41598-023-27497-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
Simultaneous knockout of the neuroendocrine marker genes Ptprn and Ptprn2, which encode the protein tyrosine phosphatase receptors N and N2, causes infertility in female mice while males are fertile. To elucidate the mechanism of the sex-specific roles of Ptprn and Ptprn2 in mouse reproduction, we analyzed the effects of their double knockout (DKO) on the hypothalamic-pituitary-gonadal axis. In DKO females, delayed puberty and lack of ovulation were observed, complemented by changes in ovarian gene expression and steroidogenesis. In contrast, testicular gene expression, steroidogenesis, and reproductive organs development were not significantly affected in DKO males. However, in both sexes, pituitary luteinizing hormone (LH) beta gene expression and LH levels were reduced, as well as follicle-stimulating hormone beta gene and gonadotropin-releasing hormone (GnRH) gene, while the calcium-mobilizing and LH secretory actions of GnRH were preserved. Hypothalamic Gnrh1 and Kiss1 gene expression was also reduced in DKO females and males. In parallel, a significant decrease in the density of immunoreactive GnRH and kisspeptin fibers was detected in the hypothalamic arcuate nucleus of DKO females and males. The female-specific kisspeptin immunoreactivity in the rostral periventricular region of the third ventricle was also reduced in DKO females, but not in DKO males. These data indicate a critical role of Ptprn and Ptprn2 in kisspeptin-GnRH neuronal function and sexual dimorphism in the threshold levels of GnRH required to preserve reproductive functions.
Collapse
Affiliation(s)
- Srdjan J Sokanovic
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Stephanie Constantin
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Aloa Lamarca Dams
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yuta Mochimaru
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kosara Smiljanic
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ivana Bjelobaba
- Department for Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000, Belgrade, Serbia
| | - Rafael M Prévide
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Stanko S Stojilkovic
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
47
|
Suzuki E, Miyado M, Kuroki Y, Fukami M. Genetic variants of G-protein coupled receptors associated with pubertal disorders. Reprod Med Biol 2023; 22:e12515. [PMID: 37122876 PMCID: PMC10134480 DOI: 10.1002/rmb2.12515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/02/2023] [Accepted: 04/11/2023] [Indexed: 05/02/2023] Open
Abstract
Background The human hypothalamic-pituitary-gonadal (HPG) axis is the regulatory center for pubertal development. This axis involves six G-protein coupled receptors (GPCRs) encoded by KISS1R, TACR3, PROKR2, GNRHR, LHCGR, and FSHR. Methods Previous studies have identified several rare variants of the six GPCR genes in patients with pubertal disorders. In vitro assays and animal studies have provided information on the function of wild-type and variant GPCRs. Main Findings Of the six GPCRs, those encoded by KISS1R and TACR3 are likely to reside at the top of the HPG axis. Several loss-of-function variants in the six genes were shown to cause late/absent puberty. In particular, variants in KISS1R, TACR3, PROKR2, and GNRHR lead to hypogonadotropic hypogonadism in autosomal dominant, recessive, and oligogenic manners. Furthermore, a few gain-of-function variants of KISS1R, PROKR2, and LHCGR have been implicated in precocious puberty. The human HPG axis may contain additional GPCRs. Conclusion The six GPCRs in the HPG axis govern pubertal development through fine-tuning of hormone secretion. Rare sequence variants in these genes jointly account for a certain percentage of genetic causes of pubertal disorders. Still, much remains to be clarified about the molecular network involving the six GPCRs.
Collapse
Affiliation(s)
- Erina Suzuki
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Mami Miyado
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
- Department of Food and NutritionBeppu UniversityOitaJapan
| | - Yoko Kuroki
- Department of Genome Medicine, National Center for Child Health and DevelopmentTokyoJapan
- Division of Collaborative Research, National Center for Child Health and DevelopmentTokyoJapan
- Division of Diversity ResearchNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Maki Fukami
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
- Division of Diversity ResearchNational Research Institute for Child Health and DevelopmentTokyoJapan
| |
Collapse
|
48
|
Franssen D, Johansson HKL, Lopez-Rodriguez D, Lavergne A, Terwagne Q, Boberg J, Christiansen S, Svingen T, Parent AS. Perinatal exposure to the fungicide ketoconazole alters hypothalamic control of puberty in female rats. Front Endocrinol (Lausanne) 2023; 14:1140886. [PMID: 37077353 PMCID: PMC10108553 DOI: 10.3389/fendo.2023.1140886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/03/2023] [Indexed: 04/05/2023] Open
Abstract
INTRODUCTION Estrogenic endocrine disrupting chemicals (EDCs) such as diethylstilbestrol (DES) are known to alter the timing of puberty onset and reproductive function in females. Accumulating evidence suggests that steroid synthesis inhibitors such as ketoconazole (KTZ) or phthalates may also affect female reproductive health, however their mode of action is poorly understood. Because hypothalamic activity is very sensitive to sex steroids, we aimed at determining whether and how EDCs with different mode of action can alter the hypothalamic transcriptome and GnRH release in female rats. DESIGN Female rats were exposed to KTZ or DES during perinatal (DES 3-6-12μg/kg.d; KTZ 3-6-12mg/kg.d), pubertal or adult periods (DES 3-12-48μg/kg.d; KTZ 3-12-48mg/kg.d). RESULTS Ex vivo study of GnRH pulsatility revealed that perinatal exposure to the highest doses of KTZ and DES delayed maturation of GnRH secretion before puberty, whereas pubertal or adult exposure had no effect on GnRH pulsatility. Hypothalamic transcriptome, studied by RNAsequencing in the preoptic area and in the mediobasal hypothalamus, was found to be very sensitive to perinatal exposure to all doses of KTZ before puberty with effects persisting until adulthood. Bioinformatic analysis with Ingenuity Pathway Analysis predicted "Creb signaling in Neurons" and "IGF-1 signaling" among the most downregulated pathways by all doses of KTZ and DES before puberty, and "PPARg" as a common upstream regulator driving gene expression changes. Deeper screening ofRNAseq datasets indicated that a high number of genes regulating the activity of the extrinsic GnRH pulse generator were consistently affected by all the doses of DES and KTZ before puberty. Several, including MKRN3, DNMT3 or Cbx7, showed similar alterations in expression at adulthood. CONCLUSION nRH secretion and the hypothalamic transcriptome are highly sensitive to perinatal exposure to both DES and KTZ. The identified pathways should be exploredfurther to identify biomarkers for future testing strategies for EDC identification and when enhancing the current standard information requirements in regulation.
Collapse
Affiliation(s)
- Delphine Franssen
- Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Liège, Belgium
- *Correspondence: Delphine Franssen,
| | | | | | - Arnaud Lavergne
- GIGA-Bioinformatics, GIGA Institute, Université de Liège, Liège, Belgium
| | - Quentin Terwagne
- Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Julie Boberg
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Sofie Christiansen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Terje Svingen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Anne-Simone Parent
- Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Liège, Belgium
- Department of Pediatrics, University Hospital Liege, Liege, Belgium
| |
Collapse
|
49
|
Street ME, Ponzi D, Renati R, Petraroli M, D’Alvano T, Lattanzi C, Ferrari V, Rollo D, Stagi S. Precocious puberty under stressful conditions: new understanding and insights from the lessons learnt from international adoptions and the COVID-19 pandemic. Front Endocrinol (Lausanne) 2023; 14:1149417. [PMID: 37201098 PMCID: PMC10187034 DOI: 10.3389/fendo.2023.1149417] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 04/06/2023] [Indexed: 05/20/2023] Open
Abstract
Neuro-biological variations in the timing of sexual maturation within a species are part of an evolved strategy that depend on internal and external environmental conditions. An increased incidence of central precocious puberty (CPP) has been described in both adopted and "covid-19 pandemic" children. Until recently, it was hypothesised that the triggers for CPP in internationally adopted children were likely to be better nutrition, greater environmental stability, and improved psychological wellbeing. However, following data collected during and after the coronavirus (COVID-19) global pandemic, other possibilities must be considered. In a society with high levels of child wellbeing, the threat to life presented by an unknown and potentially serious disease and the stressful environment created by lockdowns and other public health measures could trigger earlier pubertal maturation as an evolutionary response to favour early reproduction. The main driver for increased rates of precocious and rapidly progressive puberty during the pandemic could have been the environment of "fear and stress" in schools and households. In many children, CPP may have been triggered by the psychological effects of living without normal social contact, using PPE, being near adults concerned about financial and other issues and the fear of getting ill. The features and time of progression of CPP in children during the pandemic are similar to those observed in adopted children. This review considers the mechanisms regulating puberty with a focus on neurobiological and evolutionary mechanisms, and analyses precocious puberty both during the pandemic and in internationally adopted children searching for common yet unconsidered factors in an attempt to identify the factors which may have acted as triggers. In particular, we focus on stress as a potential factor in the early activation of the hypothalamic-pituitary-gonadal axis and its correlation with rapid sexual maturation.
Collapse
Affiliation(s)
- Maria Elisabeth Street
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Unit of Paediatrics, P. Barilla Children’s Hospital, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
- *Correspondence: Maria Elisabeth Street,
| | - Davide Ponzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Roberta Renati
- Department of Pedagogy, Psychology and Philosophy, University of Cagliari, Cagliari, Italy
| | - Maddalena Petraroli
- Unit of Paediatrics, P. Barilla Children’s Hospital, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Tiziana D’Alvano
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Unit of Paediatrics, P. Barilla Children’s Hospital, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Claudia Lattanzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Unit of Paediatrics, P. Barilla Children’s Hospital, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Vittorio Ferrari
- Health Sciences Department, University of Florence, Florence, Italy
| | - Dolores Rollo
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Stefano Stagi
- Health Sciences Department, University of Florence, Florence, Italy
- Azienda Ospedaliero Univesitaria Meyer IRCCS, Florence, Italy
| |
Collapse
|
50
|
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: 2] [Impact Index Per Article: 0.7] [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
|