1
|
Griesgraber MJ, Coolen LM, Onslow KM, Corey JR, Rice RE, Aerts EG, Bowdridge EC, Hardy SL, Lehman MN, Goodman RL, Hileman SM. Critical role of arcuate nucleus kisspeptin and Kiss1R in regulation of the ovine luteinizing hormone surge. J Neuroendocrinol 2025; 37:e70010. [PMID: 40033679 PMCID: PMC12045732 DOI: 10.1111/jne.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/28/2025] [Accepted: 02/13/2025] [Indexed: 03/05/2025]
Abstract
Hypothalamic kisspeptin (Kiss), neurokinin B (NKB), and dynorphin-containing (KNDy) neurons in the arcuate nucleus (ARC) have consistently been shown to be the central generator of gonadotropin-releasing hormone (GnRH) and corresponding luteinizing hormone (LH) pulses in mammals and possibly contribute to surge secretion as well. Additionally, recent evidence from experiments in sheep suggests that ARC Kiss1R-containing neurons play an important role in regulating the timing and amplitude of LH pulses. In this study, we examined the functional role of ARC KNDy and Kiss1R-containing neurons in ovine LH surge secretion via injection of saporin-ligand conjugates (SAP) to ablate these neural populations. NKB-SAP injections significantly reduced the percentage of ARC Kiss1 (~65% decrease) cells compared to control animals, and a surge-like increase of LH was prevented in ewes with the greatest degree of Kiss1 cell ablation. Kiss-SAP injections had no effect on Kiss1 cell percentage or ARC Kiss1R cell number compared to controls, the latter perhaps due to Kiss1R suppression in control animals from elevated estradiol concentrations during the LH surge. However, Kiss-SAP injections consistently and robustly decreased LH surge amplitude, with 80% of Kiss-SAP-treated ewes failing to generate a surge. While the exact identity of these ARC Kiss1R neurons has yet to be fully elucidated, they likely act downstream or in concert with KNDy neurons and possibly integrate other surge-centric signaling pathways to generate the ovine LH surge. These results support the conclusion that KNDy neurons contribute significantly to the ovine LH surge, while ARC Kiss1R neurons appear to be necessary for a functional surge to occur in sheep.
Collapse
Affiliation(s)
- Max J Griesgraber
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
| | - Lique M Coolen
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Kayla M Onslow
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Jacob R Corey
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Rachel E Rice
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Eliana G Aerts
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
| | - Elizabeth C Bowdridge
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
| | - Steven L Hardy
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
| | - Michael N Lehman
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Robert L Goodman
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
- Department of Neuroscience, West Virginia University, Morgantown, West Virginia, USA
| | - Stanley M Hileman
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, West Virginia, USA
- Department of Neuroscience, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
2
|
Joy KP, Chaube R. Kisspeptin control of hypothalamus-pituitary-ovarian functions. VITAMINS AND HORMONES 2024; 127:153-206. [PMID: 39864941 DOI: 10.1016/bs.vh.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The discovery of Kisspeptin (Kiss) has opened a new direction in research on neuroendocrine control of reproduction in vertebrates. Belonging to the RF amide family of peptides, Kiss and its cognate receptor Gpr54 (Kissr) have a long and complex evolutionary history. Multiple forms of Kiss and Kissr are identified in non-mammalian vertebrates, with the exception of birds, and monotreme mammals. However, only a single form of the ligand (KISS1/Kiss1) and receptor (KISS1R/Kiss1r) is retained in higher mammals. Kiss1 is distributed in the hypothalamus-pituitary-gonadal (HPG) axis and its primary function is to stimulate gonadotropin-releasing hormone (GnRH) secretion. Kiss1 neurons are distributed in the rostral periventricular area of the third ventricle (RP3V) and arcuate/infundibular nucleus (ARN/IFN). The ARN/IFN is considered the GnRH pulse generator controlled by steroid negative feedback, and the RP3V neurons is concerned with GnRH surge induced by steroid positive feedback in females. The Kiss1-Kiss1r signaling is important in all aspects of reproduction: puberty onset, maintenance of adult gonadal functions and reproductive aging, and hence assumes therapeutic potentials in the treatment of reproductive dysfunctions and induction of artificial reproduction. This chapter reviews involvement of Kiss1 in the control of the HPG axis functions in female mammals.
Collapse
Affiliation(s)
- K P Joy
- Retired Professor, Department of Zoology, Banaras Hindu University, Varanasi, Uttar pradesh, India.
| | - R Chaube
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar pradesh, India
| |
Collapse
|
3
|
Panting EN, Weight JH, Sartori JA, Coall DA, Smith JT. The role of placental kisspeptin in trophoblast invasion and migration: an assessment in Kiss1r knockout mice, BeWo cell lines and human term placenta. Reprod Fertil Dev 2024; 36:RD23230. [PMID: 38976640 DOI: 10.1071/rd23230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 06/17/2024] [Indexed: 07/10/2024] Open
Abstract
Context There is mounting evidence implicating kisspeptin signalling in placental development and function. Aims This study aimed to elucidate kisspeptin's role in trophoblast invasion and migration using three experimental models. Methods First, we examined the mouse fetus and placenta in a kisspeptin receptor (Kiss1r) knockout (KO) model. Fetal/placental weights and gene expression (quantitative polymerase chain reaction) were assessed. Second, we determined kisspeptin effects on a human trophoblast (BeWo) cell line in vitro . Third, we examined KISS1 and KISS1R gene expression in human placenta from term and pre-term pregnancies. Key results No difference was found in fetal or placental weight between Kiss1r KO and wildtype mice. However, expression of the trophoblast invasion marker, Mmp2 mRNA, was greater in the placental labyrinth zone of Kiss1r KO mice. BeWo cell models of villus cytotrophoblast and syncytiotrophoblast cells exhibited kisspeptin protein expression, with greater expression in syncytiotrophoblast, consistent with KISS1 mRNA. Kisspeptin treatment inhibited the migratory potential of cytotrophoblast-like cells. Finally, while no difference was seen in KISS1 and KISS1R mRNA between term and pre-term placentas, we saw a difference in the relative expression of each gene pre-term. We also observed a positive correlation between KISS1 expression and maternal body mass index. Conclusions Our results indicate that kisspeptin may inhibit trophoblast invasion. Implications Further investigation is required to clarify specific regulatory mechanisms.
Collapse
Affiliation(s)
- E N Panting
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - J H Weight
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - J A Sartori
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - D A Coall
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - J T Smith
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
4
|
Dorfman VB. Distribution of the kisspeptin system and its relation with gonadotropin-releasing hormone in the hypothalamus. VITAMINS AND HORMONES 2024; 127:51-78. [PMID: 39864946 DOI: 10.1016/bs.vh.2024.06.004] [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
Kisspeptin (KISS1), originally catalogued as metastin because of its capacity as a metastasis suppressor in human melanoma and breast cancer, is now recognized as the major puberty gatekeeper and gonadotropin-releasing hormone (GnRH) neuroendocrine system modulator. It is a member of the family of RFamide-related peptides that also includes the neuropeptide FF group, the gonadotropin-inhibitory hormone, the prolactin-releasing peptide, and the 26RFa peptides. The KISS1 precursor peptide is processed into a family of peptides known as kisspeptins. Its expression has been described in the hypothalamus as well as in the whole reproductive axis and several extra reproductive tissues of mammals as well as fish and amphibians, but not in birds. KISS1 plays an essential role as a regulator of the reproductive axis by inducing the synthesis and release of GnRH, acting through specific receptors. The study of the kisspeptin system and its relation with reproduction in wild and non-classical laboratory species is extremely useful to understand and become aware of the role of KISS1 in the wide variety of possible different reproductive strategies. In this chapter, KISS1 involvement in non-classical laboratory rodents, fishes, and birds is discussed.
Collapse
Affiliation(s)
- Verónica Berta Dorfman
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
5
|
McGrath BM, Norman ST, Gaspardis CA, Rose JL, Scott CJ. Characterizing the relationship between gonadotropin releasing hormone (GnRH), kisspeptin, and RFamide related peptide 3 (RFRP-3) neurons in the equine hypothalamus across the estrous cycle and in the anovulatory seasons. Theriogenology 2024; 219:157-166. [PMID: 38432143 DOI: 10.1016/j.theriogenology.2024.02.027] [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: 10/17/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
To understand better the role that kisspeptin plays in regulating seasonal and estrous cycle changes in the mare, this study investigated the number, location and interactions between GnRH, kisspeptin and RFRP-3 neurons in the equine hypothalamus. Hypothalami were collected from mares during the non-breeding season, vernal transition and various stages of the breeding season. Fluorescent immunohistochemistry was used to label the neuropeptides of interest. GnRH cells were observed primarily in the arcuate nucleus (ARC), while very few labeled cells were identified in the pre-optic area (POA). Kisspeptin cells were identified primarily in the ARC, with a small number of cells observed dorsal to the ARC, surrounding the third ventricle (3V). The mean number of kisspeptin cells varied between animals and typically showed no pattern associated with season or stage of estrous cycle, but a seasonal difference was identified in the ARC population. Small numbers of RFRP-3 cells were observed in the ARC, ventromedial hypothalamus (VMH) and dorsomedial hypothalamus (DMH). The mean number of RFRP-3 cells appeared higher in pre-ovulatory animals compared to all other stages. The percentage of GnRH cell bodies with kisspeptin appositions did not change with season or stage of estrous cycle. The percentage of kisspeptin cells receiving inputs from RFRP-3 fibers did not vary with season or stage of estrous cycle. These interactions suggest the possibility of the presence of an ultra-short loop feedback system between these three peptides. The changes in RFRP-3 neurons suggest the possibility of a role in the regulation of reproduction in the horse, but it is unlikely to be as a gonadotropin inhibitory factor.
Collapse
Affiliation(s)
- B M McGrath
- School of Dentistry & Medical Sciences, Locked bag 588, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia.
| | - S T Norman
- School of Animal and Veterinary Sciences, Locked bag 588, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia.
| | - C A Gaspardis
- School of Animal and Veterinary Sciences, Locked bag 588, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia.
| | - J L Rose
- School of Dentistry & Medical Sciences, Locked bag 588, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia.
| | - C J Scott
- School of Dentistry & Medical Sciences, Locked bag 588, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia.
| |
Collapse
|
6
|
Voigt C, Gahr M, Bennett NC. Differential regulation of Kiss1 gene expression by oestradiol in the hypothalamus of the female Damaraland mole-rat, an induced ovulator. Gen Comp Endocrinol 2023; 341:114334. [PMID: 37302764 DOI: 10.1016/j.ygcen.2023.114334] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/25/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Kisspeptin, a product of the Kiss1 gene is considered a potent stimulator of gonadotropin release, by interacting with its receptor, the G protein-coupled receptor 54. Kiss1 neurons are known to mediate the positive and negative feedback effects of oestradiol on GnRH neurons that control the pulsatile and surge secretion of GnRH. While in spontaneously ovulating mammals the GnRH/LH surge is initiated by a rise in ovarian oestradiol secreted from maturing follicles, in induced ovulators, the primary trigger is the mating stimulus. Damaraland mole rats (Fukomys damarensis) are cooperatively breeding, subterranean rodents that exhibit induced ovulation. We have previously described in this species the distribution and differential expression pattern of Kiss1-expressing neurons in the hypothalamus of males and females. Here we examine whether oestradiol (E2) regulates the hypothalamic Kiss1 expression in a similar way as described for spontaneously ovulating rodent species. By means of in situ hybridisation, we measured Kiss1 mRNA among groups of ovary-intact, ovariectomized (OVX) and OVX females treated with E2 (OVX + E2). In the arcuate nucleus (ARC), Kiss1 expression increased after ovariectomy and decreased with E2 treatment. In the preoptic region, Kiss1 expression after gonadectomy was similar to the level of wild-caught gonad-intact controls, but was dramatically upregulated with E2 treatment. The data suggest that, similar to other species, Kiss1 neurons in the ARC, which are inhibited by E2, play a role in the negative feedback control on GnRH release. The exact role of the Kiss1 neuron population in the preoptic region, which is stimulated by E2, remains to be determined.
Collapse
Affiliation(s)
- Cornelia Voigt
- Department of Zoology and Entomology, University of Pretoria, 0028 Pretoria, South Africa.
| | - Manfred Gahr
- Department of Behavioural Neurobiology, Max Planck Institute for Biological Intelligence, D-82319 Seewiesen, Germany.
| | - Nigel C Bennett
- Department of Zoology and Entomology, University of Pretoria, 0028 Pretoria, South Africa.
| |
Collapse
|
7
|
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
|
8
|
Magata F, Tsukamura H, Matsuda F. The impact of inflammatory stress on hypothalamic kisspeptin neurons: Mechanisms underlying inflammation-associated infertility in humans and domestic animals. Peptides 2023; 162:170958. [PMID: 36682622 DOI: 10.1016/j.peptides.2023.170958] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/27/2022] [Accepted: 01/17/2023] [Indexed: 01/21/2023]
Abstract
Inflammatory diseases attenuate reproductive functions in humans and domestic animals. Lipopolysaccharide (LPS), an endotoxin released by bacteria, is known to disrupt female reproductive functions in various inflammatory diseases. LPS administration has been used to elucidate the impact of pathophysiological activation of the immune system on reproduction. Hypothalamic kisspeptin neurons are the master regulators of mammalian reproduction, mediating direct stimulation of hypothalamic gonadotropin-releasing hormone (GnRH) release and consequent release of gonadotropins, such as luteinizing hormone (LH) and follicle-stimulating hormone from the pituitary. The discovery of kisspeptin neurons in the mammalian hypothalamus has drastically advanced our understanding of how inflammatory stress causes reproductive dysfunction in both humans and domestic animals. Inflammation-induced ovarian dysfunction could be caused, at least partly, by aberrant GnRH and LH secretion, which is regulated by kisspeptin signaling. In this review, we focus on the effects of LPS on hypothalamic kisspeptin neurons to outline the impact of inflammatory stress on neuroendocrine regulation of mammalian reproductive systems. First, we summarize the attenuation of female reproduction by LPS during inflammation and the effects of LPS on ovarian and pituitary function. Second, we outline the inhibitory effects of LPS on pulsatile- and surge-mode GnRH/LH release. Third, we discuss the LPS-responsive hypothalamic-pituitary-adrenal axis and hypothalamic neural systems in terms of the cytokine-mediated pathway and the possible direct action of LPS via its hypothalamic receptors. This article describes the impact of LPS on hypothalamic kisspeptin neurons and the possible mechanisms underlying LPS-mediated disruption of LH pulses/surge via kisspeptin neurons.
Collapse
Affiliation(s)
- Fumie Magata
- Department of Veterinary Medical Sciences, the University of Tokyo, Tokyo 113-8657, Japan.
| | - Hiroko Tsukamura
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi 464-8601, Japan.
| | - Fuko Matsuda
- Department of Veterinary Medical Sciences, the University of Tokyo, Tokyo 113-8657, Japan.
| |
Collapse
|
9
|
Inoue N, Hazim S, Tsuchida H, Dohi Y, Ishigaki R, Takahashi A, Otsuka Y, Yamada K, Uenoyama Y, Tsukamura H. Hindbrain Adenosine 5-Triphosphate (ATP)-Purinergic Signaling Triggers LH Surge and Ovulation via Activation of AVPV Kisspeptin Neurons in Rats. J Neurosci 2023; 43:2140-2152. [PMID: 36813577 PMCID: PMC10039743 DOI: 10.1523/jneurosci.1496-22.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 12/15/2022] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
Ovulation disorders are a serious problem for humans and livestock. In female rodents, kisspeptin neurons in the anteroventral periventricular nucleus (AVPV) are responsible for generating a luteinizing hormone (LH) surge and consequent ovulation. Here, we report that adenosine 5-triphosphate (ATP), a purinergic receptor ligand, is a possible neurotransmitter that stimulates AVPV kisspeptin neurons to induce an LH surge and consequent ovulation in rodents. Administration of an ATP receptor antagonist (PPADS) into the AVPV blocked the LH surge in ovariectomized (OVX) rats treated with a proestrous level of estrogen (OVX + high E2) and significantly reduced the ovulation rate in proestrous ovary-intact rats. AVPV ATP administration induced a surge-like LH increase in OVX + high E2 rats in the morning. Importantly, AVPV ATP administration could not induce the LH increase in Kiss1 KO rats. Furthermore, ATP significantly increased intracellular Ca2+ levels in immortalized kisspeptin neuronal cell line, and coadministration of PPADS blocked the ATP-induced Ca2+ increase. Histologic analysis revealed that the proestrous level of estrogen significantly increased the number of P2X2 receptor (an ATP receptor)-immunopositive AVPV kisspeptin neurons visualized by tdTomato in Kiss1-tdTomato rats. The proestrous level of estrogen significantly increased varicosity-like vesicular nucleotide transporter (a purinergic marker)-immunopositive fibers projecting to the vicinity of AVPV kisspeptin neurons. Furthermore, we found that some hindbrain vesicular nucleotide transporter-positive neurons projected to the AVPV and expressed estrogen receptor α, and the neurons were activated by the high E2 treatment. These results suggest that hindbrain ATP-purinergic signaling triggers ovulation via activation of AVPV kisspeptin neurons.SIGNIFICANCE STATEMENT Ovulation disorders, which cause infertility and low pregnancy rates, are a serious problem for humans and livestock. The present study provides evidence that adenosine 5-triphosphate, acting as a neurotransmitter in the brain, stimulates kisspeptin neurons in the anteroventral periventricular nucleus, known as the gonadotropin-releasing hormone surge generator, via purinergic receptors to induce the gonadotropin-releasing hormone/luteinizing hormone surge and ovulation in rats. In addition, histologic analyses indicate that adenosine 5-triphosphate is likely to be originated from the purinergic neurons in the A1 and A2 of the hindbrain. These findings may contribute to new therapeutic controls for hypothalamic ovulation disorders in humans and livestock.
Collapse
Affiliation(s)
- Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Safiullah Hazim
- 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
| | - Yuri Dohi
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Ren Ishigaki
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Ai Takahashi
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Yuki Otsuka
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Koki Yamada
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Yoshihisa Uenoyama
- 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
|
10
|
Masumi S, Lee EB, Dilower I, Upadhyaya S, Chakravarthi VP, Fields PE, Rumi MAK. The role of Kisspeptin signaling in Oocyte maturation. Front Endocrinol (Lausanne) 2022; 13:917464. [PMID: 36072937 PMCID: PMC9441556 DOI: 10.3389/fendo.2022.917464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022] Open
Abstract
Kisspeptins (KPs) secreted from the hypothalamic KP neurons act on KP receptors (KPRs) in gonadotropin (GPN) releasing hormone (GnRH) neurons to produce GnRH. GnRH acts on pituitary gonadotrophs to induce secretion of GPNs, namely follicle stimulating hormone (FSH) and luteinizing hormone (LH), which are essential for ovarian follicle development, oocyte maturation and ovulation. Thus, hypothalamic KPs regulate oocyte maturation indirectly through GPNs. KPs and KPRs are also expressed in the ovarian follicles across species. Recent studies demonstrated that intraovarian KPs also act directly on the KPRs expressed in oocytes to promote oocyte maturation and ovulation. In this review article, we have summarized published reports on the role of hypothalamic and ovarian KP-signaling in oocyte maturation. Gonadal steroid hormones regulate KP secretion from hypothalamic KP neurons, which in turn induces GPN secretion from the hypothalamic-pituitary (HP) axis. On the other hand, GPNs secreted from the HP axis act on the granulosa cells (GCs) and upregulate the expression of ovarian KPs. While KPs are expressed predominantly in the GCs, the KPRs are in the oocytes. Expression of KPs in the ovaries increases with the progression of the estrous cycle and peaks during the preovulatory GPN surge. Intrafollicular KP levels in the ovaries rise with the advancement of developmental stages. Moreover, loss of KPRs in oocytes in mice leads to failure of oocyte maturation and ovulation similar to that of premature ovarian insufficiency (POI). These findings suggest that GC-derived KPs may act on the KPRs in oocytes during their preovulatory maturation. In addition to the intraovarian role of KP-signaling in oocyte maturation, in vivo, a direct role of KP has been identified during in vitro maturation of sheep, porcine, and rat oocytes. KP-stimulation of rat oocytes, in vitro, resulted in Ca2+ release and activation of the mitogen-activated protein kinase, extracellular signal-regulated kinase 1 and 2. In vitro treatment of rat or porcine oocytes with KPs upregulated messenger RNA levels of the factors that favor oocyte maturation. In clinical trials, human KP-54 has also been administered successfully to patients undergoing assisted reproductive technologies (ARTs) for increasing oocyte maturation. Exogenous KPs can induce GPN secretion from hypothalamus; however, the possibility of direct KP action on the oocytes cannot be excluded. Understanding the direct in vivo and in vitro roles of KP-signaling in oocyte maturation will help in developing novel KP-based ARTs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - M. A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
11
|
Uenoyama Y, Tsuchida H, Nagae M, Inoue N, Tsukamura H. Opioidergic pathways and kisspeptin in the regulation of female reproduction in mammals. Front Neurosci 2022; 16:958377. [PMID: 36033602 PMCID: PMC9404872 DOI: 10.3389/fnins.2022.958377] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Endogenous opioid peptides have attracted attention as critical neuropeptides in the central mechanism regulating female reproduction ever since the discovery that arcuate dynorphin neurons that coexpress kisspeptin and neurokinin B (NKB), which are also known as kisspeptin/neurokinin B/dynorphin (KNDy) neurons, play a role as a master regulator of pulsatile gonadotropin-releasing hormone (GnRH) release in mammals. In this study, we first focus on the role of dynorphin released by KNDy neurons in the GnRH pulse generation. Second, we provide a historical overview of studies on endogenous opioid peptides. Third, we discuss how endogenous opioid peptides modulate tonic GnRH/gonadotropin release in female mammals as a mediator of inhibitory internal and external cues, such as ovarian steroids, nutritional status, or stress, on reproduction. Then, we discuss the role of endogenous opioid peptides in GnRH surge generation in female mammals.
Collapse
|
12
|
Kauffman AS. Neuroendocrine mechanisms underlying estrogen positive feedback and the LH surge. Front Neurosci 2022; 16:953252. [PMID: 35968365 PMCID: PMC9364933 DOI: 10.3389/fnins.2022.953252] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/08/2022] [Indexed: 01/26/2023] Open
Abstract
A fundamental principle in reproductive neuroendocrinology is sex steroid feedback: steroid hormones secreted by the gonads circulate back to the brain to regulate the neural circuits governing the reproductive neuroendocrine axis. These regulatory feedback loops ultimately act to modulate gonadotropin-releasing hormone (GnRH) secretion, thereby affecting gonadotropin secretion from the anterior pituitary. In females, rising estradiol (E2) during the middle of the menstrual (or estrous) cycle paradoxically "switch" from being inhibitory on GnRH secretion ("negative feedback") to stimulating GnRH release ("positive feedback"), resulting in a surge in GnRH secretion and a downstream LH surge that triggers ovulation. While upstream neural afferents of GnRH neurons, including kisspeptin neurons in the rostral hypothalamus, are proposed as critical loci of E2 feedback action, the underlying mechanisms governing the shift between E2 negative and positive feedback are still poorly understood. Indeed, the precise cell targets, neural signaling factors and receptors, hormonal pathways, and molecular mechanisms by which ovarian-derived E2 indirectly stimulates GnRH surge secretion remain incompletely known. In many species, there is also a circadian component to the LH surge, restricting its occurrence to specific times of day, but how the circadian clock interacts with endocrine signals to ultimately time LH surge generation also remains a major gap in knowledge. Here, we focus on classic and recent data from rodent models and discuss the consensus knowledge of the neural players, including kisspeptin, the suprachiasmatic nucleus, and glia, as well as endocrine players, including estradiol and progesterone, in the complex regulation and generation of E2-induced LH surges in females.
Collapse
|
13
|
Semaan SJ, Kauffman AS. Developmental sex differences in the peri-pubertal pattern of hypothalamic reproductive gene expression, including Kiss1 and Tac2, may contribute to sex differences in puberty onset. Mol Cell Endocrinol 2022; 551:111654. [PMID: 35469849 PMCID: PMC9889105 DOI: 10.1016/j.mce.2022.111654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 02/03/2023]
Abstract
The mechanisms regulating puberty still remain elusive, as do the underlying causes for sex differences in puberty onset (girls before boys) and pubertal disorders. Neuroendocrine puberty onset is signified by increased pulsatile GnRH secretion, yet how and when various upstream reproductive neural circuits change developmentally to govern this process is poorly understood. We previously reported day-by-day peri-pubertal increases (Kiss1, Tac2) or decreases (Rfrp) in hypothalamic gene expression of female mice, with several brain mRNA changes preceding external pubertal markers. However, similar pubertal measures in males were not previously reported. Here, to identify possible neural sex differences underlying sex differences in puberty onset, we analyzed peri-pubertal males and directly compared them with female littermates. Kiss1 expression in male mice increased over the peri-pubertal period in both the AVPV and ARC nuclei but with lower levels than in females at several ages. Likewise, Tac2 expression in the male ARC increased between juvenile and older peri-pubertal stages but with levels lower than females at most ages. By contrast, both DMN Rfrp expressionand Rfrp neuronal activation strongly decreased in males between juvenile and peri-pubertal stages, but with similar levels as females. Neither ARC KNDy neuronal activation nor Kiss1r expression in GnRH neurons differed between males and females or changed with age. These findings delineate several peri-pubertal changes in neural populations in developing males, with notable sex differences in kisspeptin and NKB neuron developmental patterns. Whether these peri-pubertal hypothalamic sex differences underlie sex differences in puberty onset deserves future investigation.
Collapse
Affiliation(s)
- Sheila J Semaan
- 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
|
14
|
Hu KL, Chen Z, Li X, Cai E, Yang H, Chen Y, Wang C, Ju L, Deng W, Mu L. Advances in clinical applications of kisspeptin-GnRH pathway in female reproduction. Reprod Biol Endocrinol 2022; 20:81. [PMID: 35606759 PMCID: PMC9125910 DOI: 10.1186/s12958-022-00953-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/30/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Kisspeptin is the leading upstream regulator of pulsatile and surge Gonadotrophin-Releasing Hormone secretion (GnRH) in the hypothalamus, which acts as the key governor of the hypothalamic-pituitary-ovary axis. MAIN TEXT Exogenous kisspeptin or its receptor agonist can stimulate GnRH release and subsequent physiological gonadotropin secretion in humans. Based on the role of kisspeptin in the hypothalamus, a broad application of kisspeptin and its receptor agonist has been recently uncovered in humans, including central control of ovulation, oocyte maturation (particularly in women at a high risk of ovarian hyperstimulation syndrome), test for GnRH neuronal function, and gatekeepers of puberty onset. In addition, the kisspeptin analogs, such as TAK-448, showed promising agonistic activity in healthy women as well as in women with hypothalamic amenorrhoea or polycystic ovary syndrome. CONCLUSION More clinical trials should focus on the therapeutic effect of kisspeptin, its receptor agonist and antagonist in women with reproductive disorders, such as hypothalamic amenorrhoea, polycystic ovary syndrome, and endometriosis.
Collapse
Affiliation(s)
- Kai-Lun Hu
- Center for Reproductive Medicine, Peking University Third Hospital, No.49 Huayuan North Road, Haidian District, Beijing, People's Republic of China, 100191
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Zimiao Chen
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Xiaoxue Li
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Enci Cai
- Department of Nutrition and Food Science, College of Food, Agricultural and Natural Resource Sciences, University of Minnesota, Twin Cities, Minneapolis, MN, 55455, USA
| | - Haiyan Yang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Yi Chen
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Congying Wang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Liping Ju
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Wenhai Deng
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China, 325006.
| | - Liangshan Mu
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200.
| |
Collapse
|
15
|
Campbell RE, Coolen LM, Hoffman GE, Hrabovszky E. Highlights of neuroanatomical discoveries of the mammalian gonadotropin-releasing hormone system. J Neuroendocrinol 2022; 34:e13115. [PMID: 35502534 PMCID: PMC9232911 DOI: 10.1111/jne.13115] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/09/2022] [Accepted: 03/01/2022] [Indexed: 11/17/2022]
Abstract
The anatomy and morphology of gonadotropin-releasing hormone (GnRH) neurons makes them both a joy and a challenge to investigate. They are a highly unique population of neurons given their developmental migration into the brain from the olfactory placode, their relatively small number, their largely scattered distribution within the rostral forebrain, and, in some species, their highly varied individual anatomical characteristics. These unique features have posed technological hurdles to overcome and promoted fertile ground for the establishment and use of creative approaches. Historical and more contemporary discoveries defining GnRH neuron anatomy remain critical in shaping and challenging our views of GnRH neuron function in the regulation of reproductive function. We begin this review with a historical overview of anatomical discoveries and developing methodologies that have shaped our understanding of the reproductive axis. We then highlight significant discoveries across specific groups of mammalian species to address some of the important comparative aspects of GnRH neuroanatomy. Lastly, we touch on unresolved questions and opportunities for future neuroanatomical research on this fascinating and important population of neurons.
Collapse
Affiliation(s)
- Rebecca E. Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| | - Lique M. Coolen
- Department of Biological SciencesKent State UniversityKentOhioUSA
| | | | - Erik Hrabovszky
- Laboratory of Reproductive NeurobiologyInstitute of Experimental MedicineBudapestHungary
| |
Collapse
|
16
|
Goodman RL, Herbison AE, Lehman MN, Navarro VM. Neuroendocrine control of gonadotropin-releasing hormone: Pulsatile and surge modes of secretion. J Neuroendocrinol 2022; 34:e13094. [PMID: 35107859 PMCID: PMC9948945 DOI: 10.1111/jne.13094] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 11/28/2022]
Abstract
The concept that different systems control episodic and surge secretion of gonadotropin-releasing hormone (GnRH) was well established by the time that GnRH was identified and formed the framework for studies of the physiological roles of GnRH, and later kisspeptin. Here, we focus on recent studies identifying the neural mechanisms underlying these two modes of secretion, with an emphasis on their core components. There is now compelling data that kisspeptin neurons in the arcuate nucleus that also contain neurokinin B (NKB) and dynorphin (i.e., KNDy cells) and their projections to GnRH dendrons constitute the GnRH pulse generator in mice and rats. There is also strong evidence for a similar role for KNDy neurons in sheep and goats, and weaker data in monkeys and humans. However, whether KNDy neurons act on GnRH dendrons and/or GnRH soma and dendrites that are found in the mediobasal hypothalamus (MBH) of these species remains unclear. The core components of the GnRH/luteinising hormone surge consist of an endocrine signal that initiates the process and a neural trigger that drives GnRH secretion during the surge. In all spontaneous ovulators, the core endocrine signal is a rise in estradiol secretion from the maturing follicle(s), with the site of estrogen positive feedback being the rostral periventricular kisspeptin neurons in rodents and neurons in the MBH of sheep and primates. There is considerable species variations in the neural trigger, with three major classes. First, in reflex ovulators, this trigger is initiated by coitus and carried to the hypothalamus by neural or vascular pathways. Second, in rodents, there is a time of day signal that originates in the suprachiasmatic nucleus and activates rostral periventricular kisspeptin neurons and GnRH soma and dendrites. Finally, in sheep nitric oxide-producing neurons in the ventromedial nucleus, KNDy neurons and rostral kisspeptin neurons all appear to participate in driving GnRH release during the surge.
Collapse
Affiliation(s)
- Robert L. Goodman
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Allan E. Herbison
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Michael N. Lehman
- Brain Health Research Institute, Kent State University, Kent, OH, USA
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Victor M. Navarro
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School and Department of Medicine, Boston, MA, USA
| |
Collapse
|
17
|
Dardente H, Simonneaux V. GnRH and the photoperiodic control of seasonal reproduction: Delegating the task to kisspeptin and RFRP-3. J Neuroendocrinol 2022; 34:e13124. [PMID: 35384117 DOI: 10.1111/jne.13124] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/22/2022] [Accepted: 03/03/2022] [Indexed: 10/18/2022]
Abstract
Synchronization of mammalian breeding activity to the annual change of photoperiod and environmental conditions is of the utmost importance for individual survival and species perpetuation. Subsequent to the early 1960s, when the central role of melatonin in this adaptive process was demonstrated, our comprehension of the mechanisms through which light regulates gonadal activity has increased considerably. The current model for the photoperiodic neuroendocrine system points to pivotal roles for the melatonin-sensitive pars tuberalis (PT) and its seasonally-regulated production of thyroid-stimulating hormone (TSH), as well as for TSH-sensitive hypothalamic tanycytes, radial glia-like cells located in the basal part of the third ventricle. Tanycytes respond to TSH through increased expression of thyroid hormone (TH) deiodinase 2 (Dio2), which leads to heightened production of intrahypothalamic triiodothyronine (T3) during longer days of spring and summer. There is strong evidence that this local, long-day driven, increase in T3 links melatonin input at the PT to gonadotropin-releasing hormone (GnRH) output, to align breeding with the seasons. The mechanism(s) through which T3 impinges upon GnRH remain(s) unclear. However, two distinct neuronal populations of the medio-basal hypothalamus, which express the (Arg)(Phe)-amide peptides kisspeptin and RFamide-related peptide-3, appear to be well-positioned to relay this seasonal T3 message towards GnRH neurons. Here, we summarize our current understanding of the cellular, molecular and neuroendocrine players, which keep track of photoperiod and ultimately govern GnRH output and seasonal breeding.
Collapse
Affiliation(s)
- Hugues Dardente
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Valérie Simonneaux
- Institute for Cellular and Integrative Neuroscience, University of Strasbourg, Strasbourg, France
| |
Collapse
|
18
|
PASSARELLI A, LETTIERI A, DEMIRCI TN, MAGNI P. Gonadotropin-releasing hormone-secreting neuron development and function: an update. Minerva Endocrinol (Torino) 2022; 47:58-69. [DOI: 10.23736/s2724-6507.22.03683-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
19
|
Abstract
Hypothalamic kisspeptin (Kiss1) neurons provide indispensable excitatory transmission to gonadotropin-releasing hormone (GnRH) neurons for the coordinated release of gonadotropins, estrous cyclicity, and ovulation. But maintaining reproductive functions is metabolically demanding so there must be a coordination with multiple homeostatic functions, and it is apparent that Kiss1 neurons play that role. There are 2 distinct populations of hypothalamic Kiss1 neurons, namely arcuate nucleus (Kiss1ARH) neurons and anteroventral periventricular and periventricular nucleus (Kiss1AVPV/PeN) neurons in rodents, both of which excite GnRH neurons via kisspeptin release but are differentially regulated by ovarian steroids. Estradiol (E2) increases the expression of kisspeptin in Kiss1AVPV/PeN neurons but decreases its expression in Kiss1ARH neurons. Also, Kiss1ARH neurons coexpress glutamate and Kiss1AVPV/PeN neurons coexpress gamma aminobutyric acid (GABA), both of which are upregulated by E2 in females. Also, Kiss1ARH neurons express critical metabolic hormone receptors, and these neurons are excited by insulin and leptin during the fed state. Moreover, Kiss1ARH neurons project to and excite the anorexigenic proopiomelanocortin neurons but inhibit the orexigenic neuropeptide Y/Agouti-related peptide neurons, highlighting their role in regulating feeding behavior. Kiss1ARH and Kiss1AVPV/PeN neurons also project to the preautonomic paraventricular nucleus (satiety) neurons and the dorsomedial nucleus (energy expenditure) neurons to differentially regulate their function via glutamate and GABA release, respectively. Therefore, this review will address not only how Kiss1 neurons govern GnRH release, but how they control other homeostatic functions through their peptidergic, glutamatergic and GABAergic synaptic connections, providing further evidence that Kiss1 neurons are the key neurons coordinating energy states with reproduction.
Collapse
Affiliation(s)
- Oline K Rønnekleiv
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Jian Qiu
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97239, USA
| | - Martin J Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| |
Collapse
|
20
|
Jamieson BB, Moore AM, Lohr DB, Thomas SX, Coolen LM, Lehman MN, Campbell RE, Piet R. Prenatal androgen treatment impairs the suprachiasmatic nucleus arginine-vasopressin to kisspeptin neuron circuit in female mice. Front Endocrinol (Lausanne) 2022; 13:951344. [PMID: 35992143 PMCID: PMC9388912 DOI: 10.3389/fendo.2022.951344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/06/2022] [Indexed: 01/13/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is associated with elevated androgen and luteinizing hormone (LH) secretion and with oligo/anovulation. Evidence indicates that elevated androgens impair sex steroid hormone feedback regulation of pulsatile LH secretion. Hyperandrogenemia in PCOS may also disrupt the preovulatory LH surge. The mechanisms through which this might occur, however, are not fully understood. Kisspeptin (KISS1) neurons of the rostral periventricular area of the third ventricle (RP3V) convey hormonal cues to gonadotropin-releasing hormone (GnRH) neurons. In rodents, the preovulatory surge is triggered by these hormonal cues and coincident timing signals from the central circadian clock in the suprachiasmatic nucleus (SCN). Timing signals are relayed to GnRH neurons, in part, via projections from SCN arginine-vasopressin (AVP) neurons to RP3VKISS1 neurons. Because rodent SCN cells express androgen receptors (AR), we hypothesized that these circuits are impaired by elevated androgens in a mouse model of PCOS. In prenatally androgen-treated (PNA) female mice, SCN Ar expression was significantly increased compared to that found in prenatally vehicle-treated mice. A similar trend was seen in the number of Avp-positive SCN cells expressing Ar. In the RP3V, the number of kisspeptin neurons was preserved. Anterograde tract-tracing, however, revealed reduced SCNAVP neuron projections to the RP3V and a significantly lower proportion of RP3VKISS1 neurons with close appositions from SCNAVP fibers. Functional assessments showed, on the other hand, that RP3VKISS1 neuron responses to AVP were maintained in PNA mice. These findings indicate that PNA changes some of the neural circuits that regulate the preovulatory surge. These impairments might contribute to ovulatory dysfunction in PNA mice modeling PCOS.
Collapse
Affiliation(s)
- Bradley B. Jamieson
- Centre for Neuroendocrinology and Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Aleisha M. Moore
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Dayanara B. Lohr
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Simone X. Thomas
- Centre for Neuroendocrinology and Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Lique M. Coolen
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Michael N. Lehman
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Rebecca E. Campbell
- Centre for Neuroendocrinology and Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Richard Piet
- Centre for Neuroendocrinology and Department of Physiology, University of Otago, Dunedin, New Zealand
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
- *Correspondence: Richard Piet,
| |
Collapse
|
21
|
Uenoyama Y, Inoue N, Nakamura S, Tsukamura H. Kisspeptin Neurons and Estrogen-Estrogen Receptor α Signaling: Unraveling the Mystery of Steroid Feedback System Regulating Mammalian Reproduction. Int J Mol Sci 2021; 22:ijms22179229. [PMID: 34502135 PMCID: PMC8430864 DOI: 10.3390/ijms22179229] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/21/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
Estrogen produced by ovarian follicles plays a key role in the central mechanisms controlling reproduction via regulation of gonadotropin-releasing hormone (GnRH) release by its negative and positive feedback actions in female mammals. It has been well accepted that estrogen receptor α (ERα) mediates both estrogen feedback actions, but precise targets had remained as a mystery for decades. Ever since the discovery of kisspeptin neurons as afferent ERα-expressing neurons to govern GnRH neurons, the mechanisms mediating estrogen feedback are gradually being unraveled. The present article overviews the role of kisspeptin neurons in the arcuate nucleus (ARC), which are considered to drive pulsatile GnRH/gonadotropin release and folliculogenesis, in mediating the estrogen negative feedback action, and the role of kisspeptin neurons located in the anteroventral periventricular nucleus-periventricular nucleus (AVPV-PeN), which are thought to drive GnRH/luteinizing hormone (LH) surge and consequent ovulation, in mediating the estrogen positive feedback action. This implication has been confirmed by the studies showing that estrogen-bound ERα down- and up-regulates kisspeptin gene (Kiss1) expression in the ARC and AVPV-PeN kisspeptin neurons, respectively. The article also provides the molecular and epigenetic mechanisms regulating Kiss1 expression in kisspeptin neurons by estrogen. Further, afferent ERα-expressing neurons that may regulate kisspeptin release are discussed.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan; (Y.U.); (N.I.)
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan; (Y.U.); (N.I.)
| | - Sho Nakamura
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari 794-8555, Japan;
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan; (Y.U.); (N.I.)
- Correspondence:
| |
Collapse
|
22
|
Bakker J. Kisspeptin and neurokinin B expression in the human hypothalamus: Relation to reproduction and gender identity. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:297-313. [PMID: 34225936 DOI: 10.1016/b978-0-12-820107-7.00018-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus are at the core of reproductive functioning. GnRH released into the median eminence regulates the secretion of the gonadotropins from the anterior pituitary, which in turn activates gametogenesis and steroid synthesis by the gonads. The GnRH system displays functional sex differences: GnRH is secreted in pulses at a constant frequency in men, whereas in women, pulse frequency varies over the menstrual cycle. In both sexes, GnRH release is regulated by sex steroid hormones, acting at the level of the hypothalamus and the anterior pituitary in a classic feedback loop. Because GnRH neurons do not express sex steroid receptors, hormone effects on GnRH release are presumed to be mediated indirectly through other steroid-sensitive neuronal systems, which then converge onto GnRH cell bodies and/or terminals. Human genetic studies demonstrated that kisspeptin (KP) as well as neurokinin B (NKB) signaling are both potent regulators of GNRH secretion. In humans, postmortem studies using immunohistochemistry have shown that women have higher KP and NKB expression in the infundibular nucleus than men. Sex differences in KP expression are present throughout life, which is from the infant/prepubertal into the elderly period, whereas sex differences in NKB expression do not emerge until adulthood. KP and NKB are often coexpressed together with dynorphin by the same population of neurons, also known as KDNy neurons in other species. Indeed, significant coexpression between KP and NKB but not with Dynorphin has been observed thereby challenging the KDNy concept in humans. Female-typical expression of both KP and NKB were observed in the infundibular nucleus of trans women (male sex assigned at birth and female gender identity). Taken together, sex differences in KP and NKB expression most likely reflect organizational actions of sex steroid hormones on the developing brain but they also remain sensitive to circulating sex steroids in adulthood. The female-dominant sex difference in infundibular KP and NKB expression suggests that this brain region is most likely involved in both the negative and positive feedback actions of estrogens on GnRH secretion. Finally, the sex-reversal observed in KP and NKB expression in trans women might reflect, at least partially, an atypical sexual differentiation of the brain.
Collapse
Affiliation(s)
- Julie Bakker
- GIGA Neurosciences, Liège University, Liège, Belgium.
| |
Collapse
|
23
|
Uenoyama Y, Nagae M, Tsuchida H, Inoue N, Tsukamura H. Role of KNDy Neurons Expressing Kisspeptin, Neurokinin B, and Dynorphin A as a GnRH Pulse Generator Controlling Mammalian Reproduction. Front Endocrinol (Lausanne) 2021; 12:724632. [PMID: 34566891 PMCID: PMC8458932 DOI: 10.3389/fendo.2021.724632] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/23/2021] [Indexed: 01/16/2023] Open
Abstract
Increasing evidence accumulated during the past two decades has demonstrated that the then-novel kisspeptin, which was discovered in 2001, the known neuropeptides neurokinin B and dynorphin A, which were discovered in 1983 and 1979, respectively, and their G-protein-coupled receptors, serve as key molecules that control reproduction in mammals. The present review provides a brief historical background and a summary of our recent understanding of the roles of hypothalamic neurons expressing kisspeptin, neurokinin B, and dynorphin A, referred to as KNDy neurons, in the central mechanism underlying gonadotropin-releasing hormone (GnRH) pulse generation and subsequent tonic gonadotropin release that controls mammalian reproduction.
Collapse
|
24
|
Rumpler É, Skrapits K, Takács S, Göcz B, Trinh SH, Rácz G, Matolcsy A, Kozma Z, Ciofi P, Dhillo WS, Hrabovszky E. Characterization of Kisspeptin Neurons in the Human Rostral Hypothalamus. Neuroendocrinology 2021; 111:249-262. [PMID: 32299085 DOI: 10.1159/000507891] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/14/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Kisspeptin (KP) neurons in the rostral periventricular region of the 3rd ventricle (RP3V) of female rodents mediate positive estrogen feedback to gonadotropin-releasing hormone neurons and, thus, play a fundamental role in the mid-cycle luteinizing hormone (LH) surge. The RP3V is sexually dimorphic, and male rodents with lower KP cell numbers are unable to mount estrogen-induced LH surges. OBJECTIVE To find and characterize the homologous KP neurons in the human brain, we studied formalin-fixed post-mortem hypothalami. METHODS Immunohistochemical techniques were used. RESULTS The distribution of KP neurons in the rostral hypothalamus overlapped with distinct subdivisions of the paraventricular nucleus. The cell numbers decreased after menopause, indicating that estrogens positively regulate KP gene expression in the rostral hypothalamus in humans, similarly to several other species. Young adult women and men had similar cell numbers, as opposed to rodents reported to have more KP neurons in the RP3V of females. Human KP neurons differed from the homologous rodent cells as well, in that they were devoid of enkephalins, galanin and tyrosine hydroxylase. Further, they did not contain known KP neuron markers of the human infundibular nucleus, neurokinin B, substance P and cocaine- and amphetamine-regulated transcript, while they received afferent input from these KP neurons. CONCLUSIONS The identification and positive estrogenic regulation of KP neurons in the human rostral hypothalamus challenge the long-held view that positive estrogen feedback may be restricted to the mediobasal part of the hypothalamus in primates and point to the need of further anatomical, molecular and functional studies of rostral hypothalamic KP neurons.
Collapse
Affiliation(s)
- Éva Rumpler
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Szabolcs Takács
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Göcz
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Sarolta H Trinh
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Gergely Rácz
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - András Matolcsy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Zsolt Kozma
- Department of Forensic Medicine, Faculty of Medicine, University of Pécs, Pécs, Hungary
| | | | - Waljit S Dhillo
- Department of Investigative Medicine, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary,
| |
Collapse
|
25
|
Hrabovszky E, Takács S, Rumpler É, Skrapits K. The human hypothalamic kisspeptin system: Functional neuroanatomy and clinical perspectives. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:275-296. [PMID: 34225935 DOI: 10.1016/b978-0-12-820107-7.00017-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In mammals, kisspeptin neurons are the key components of the hypothalamic neuronal networks that regulate the onset of puberty, account for the pulsatile secretion of gonadotropin-releasing hormone (GnRH) and mediate negative and positive estrogen feedback signals to GnRH neurons. Being directly connected anatomically and functionally to the hypophysiotropic GnRH system, the major kisspeptin cell groups of the preoptic area/rostral hypothalamus and the arcuate (or infundibular) nucleus, respectively, are ideally positioned to serve as key nodes which integrate various types of environmental, endocrine, and metabolic signals that can influence fertility. This chapter provides an overview of the current state of knowledge on the anatomy, functions, and plasticity of brain kisspeptin systems based on the wide literature available from different laboratory and domestic species. Then, the species-specific features of human hypothalamic kisspeptin neurons are described, covering their topography, morphology, unique neuropeptide content, plasticity, and connectivity to hypophysiotropic GnRH neurons. Some newly emerging roles of central kisspeptin signaling in behavior and finally, clinical perspectives, are discussed.
Collapse
Affiliation(s)
- Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary.
| | - Szabolcs Takács
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Éva Rumpler
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
26
|
Patel R, Smith JT. Novel actions of kisspeptin signaling outside of GnRH-mediated fertility: a potential role in energy balance. Domest Anim Endocrinol 2020; 73:106467. [PMID: 32278499 DOI: 10.1016/j.domaniend.2020.106467] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 01/08/2023]
Abstract
Kisspeptin, encoded by Kiss1 gene expressing neurons in the hypothalamus, is a requisite for fertility and now appears critical in the regulation of energy balance. Kisspeptin neurons, particularly those in the arcuate nucleus (ARC), receive information directly and indirectly from a diverse array of brain regions including the bed nucleus of the stria terminalis, amygdala, interpeduncular nucleus, hippocampus, and cortex. On the other hand, kisspeptin neuron projections clearly extend to GnRH neuron cell bodies in rodents, sheep, and primates and beyond to other-non-GnRH-brain areas. Kiss1r, the kisspeptin receptor, is expressed on GnRH neurons and also in additional brain areas and peripheral tissues, indicating a nonreproductive role. Kisspeptin neurons clearly receive signals pertinent to deviations in energy balance but are now recognized as a novel neuroendocrine player in the fine balance of energy intake and expenditure. Mice that have a dysfunctional gene for Kiss1r develop an obese and diabetic phenotype. The mechanism behind this altered metabolic state is still mostly unknown; however, Kiss1r expression in the pancreas and brown adipose tissue is clearly functional and required for normal glucose tolerance and energy expenditure, respectively. Kisspeptin neurons in the ARC also participate in the generation of circadian rhythms, specifically those concerning food intake and metabolism, offering a potential explanation for the obesity in Kiss1r knockout mice. Overall, the discoveries of new mechanistic roles for kisspeptin in both normal and pathophysiologic states of energy balance may lead to further understating of obesity prevalence and novel therapeutic targets and interventions.
Collapse
Affiliation(s)
- R Patel
- School of Human Sciences, M309, The University of Western Australia, 35 Stirling Highway Crawley, Perth, Western Australia, Australia 6009
| | - J T Smith
- School of Human Sciences, M309, The University of Western Australia, 35 Stirling Highway Crawley, Perth, Western Australia, Australia 6009.
| |
Collapse
|
27
|
Młotkowska P, Marciniak E, Roszkowicz-Ostrowska K, Misztal T. Effects of allopregnanolone on central reproductive functions in sheep under natural and stressful conditions. Theriogenology 2020; 158:138-147. [PMID: 32956862 DOI: 10.1016/j.theriogenology.2020.09.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 10/23/2022]
Abstract
Reproductive functions may be affected by internal and external factors that are integrated in the central nervous system (CNS). Stressful stimuli induce the neuroendocrine response of the hypothalamic-pituitary-adrenal axis, as well as the synthesis of the neurosteroid allopregnanolone (AL) in the brain. This study tested the hypothesis that centrally administered AL could affect the expression of certain genes involved in reproductive functions at the hypothalamus and pituitary levels, as well as pulsatile gonadotropin secretion in sheep under both natural and stressful conditions. Luteal-phase sheep (n = 24) were subjected to a three-day (day 12-14 of the estrous cycle) series of control or AL (4 × 15 μg/60 μL/30 min, at 30 min intervals) infusions into the third ventricle. Acute stressful stimuli (isolation from other sheep and partial movement restriction) were used in the third day of infusion. Stressful stimuli reduced kisspeptin-1 mRNA levels in both the mediobasal hypothalamus (MBH) and the preoptic area (POA), while pro-dynorphin (PDYN) mRNA level only in the MBH. AL alone decreased the abundances of these transcripts in both structures. Stress increased the expression of gonadotropin-releasing hormone (GnRH) mRNA in the MBH and POA, luteinizing hormone (LH) β subunit (LHβ) mRNA in the anterior pituitary (AP) and pulsatile LH secretion. In contrast, mRNA level of follicle stimulating hormone (FSH) β subunit (FSHβ) was decreased in the AP, with no effect of stress on pulsatile FSH secretion. In stressed sheep, AL counteracted the increase in GnRH mRNA expression only in the POA, but it decreased the level of this transcript in both hypothalamic tissues when infused alone. AL prevented the stress-induced increase in LHβ mRNA expression in the AP and pulsatile LH secretion, as well as inhibited almost all aspects of FSH secretion when administered alone. The suppressive effect of AL on GnRH receptor mRNA expression was also observed in both MBH and AP. We concluded that acute stress and AL exerted multidirectional effects on hypothalamic centers that regulate reproductive functions and secretory activity of AP gonadotrophs in sheep. However, we indicated the dominant inhibitory effect of AL under natural and stressful conditions.
Collapse
Affiliation(s)
- Patrycja Młotkowska
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3 St, 05-110, Jablonna, Poland.
| | - Elżbieta Marciniak
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3 St, 05-110, Jablonna, Poland.
| | - Katarzyna Roszkowicz-Ostrowska
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3 St, 05-110, Jablonna, Poland.
| | - Tomasz Misztal
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3 St, 05-110, Jablonna, Poland.
| |
Collapse
|
28
|
Horihata K, Inoue N, Uenoyama Y, Maeda KI, Tsukamura H. Retinoblastoma binding protein 7 is involved in Kiss1 mRNA upregulation in rodents. J Reprod Dev 2020; 66:125-133. [PMID: 31956172 PMCID: PMC7175387 DOI: 10.1262/jrd.2019-149] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Kisspeptin, encoded by Kiss1, is essential for reproduction in mammals. Kiss1 expression is regulated by estrogen via histone acetylation in the
Kiss1 promotor region. Thus, elucidation of histone modification factor(s) involved in the regulation of Kiss1 expression is required to gain further
understanding of the mechanisms of its control. The RNA-seq analysis of isolated kisspeptin neurons, obtained from the arcuate nucleus (ARC) of female rats, revealed that
Rbbp7, encoding retinoblastoma binding protein 7 (RBBP7), a member of histone modification and chromatin remodeling complexes, is highly expressed in the ARC kisspeptin
neurons. Thus, the present study aimed to investigate whether RBBP7 is involved in Kiss1 expression. Histological analysis using in situ hybridization (ISH)
revealed that Rbbp7 expression was located in several hypothalamic nuclei, including the ARC and the anteroventral periventricular nucleus (AVPV), where kisspeptin neurons
are located. Double ISH for Rbbp7 and Kiss1 showed that a majority of kisspeptin neurons (more than 85%) expressed Rbbp7 mRNA in both the
ARC and the AVPV of female rats. Further, Rbbp7 mRNA knockdown significantly decreased in vitro expression of Kiss1 in a mouse immortalized
kisspeptin neuronal cell line (mHypoA-55). Estrogen treatment significantly decreased and increased Kiss1 mRNA levels in the ARC and AVPV of ovariectomized female rats,
respectively, but failed to affect Rbbp7 mRNA levels in both the nuclei. Taken together, these findings suggest that RBBP7 is involved in the upregulation of
Kiss1 expression in kisspeptin neurons of rodents in an estrogen-independent manner.
Collapse
Affiliation(s)
- Kei Horihata
- 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
| | - Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Kei-Ichiro Maeda
- Laboratory of Theriogenology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
29
|
Gotlieb N, Baker CN, Moeller J, Kriegsfeld LJ. Time-of-day-dependent sensitivity of the reproductive axis to RFamide-related peptide-3 inhibition in female Syrian hamsters. J Neuroendocrinol 2019; 31:e12798. [PMID: 31550401 PMCID: PMC6991702 DOI: 10.1111/jne.12798] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 08/15/2019] [Accepted: 09/19/2019] [Indexed: 12/15/2022]
Abstract
In spontaneously ovulating rodent species, the timing of the luteinising hormone (LH) surge is controlled by the master circadian pacemaker in the suprachiasmatic nucleus (SCN). The SCN initiates the LH surge via the coordinated control of two opposing neuropeptidergic systems that lie upstream of the gonadotrophin-releasing hormone (GnRH) neuronal system: the stimulatory peptide, kisspeptin, and the inhibitory peptide, RFamide-related peptide-3 (RFRP-3; the mammalian orthologue of avian gonadotrophin-inhibitory hormone [GnIH]). We have previously shown that the GnRH system exhibits time-dependent sensitivity to kisspeptin stimulation, further contributing to the precise timing of the LH surge. To examine whether this time-dependent sensitivity of the GnRH system is unique to kisspeptin or a more common mechanism of regulatory control, we explored daily changes in the response of the GnRH system to RFRP-3 inhibition. Female Syrian hamsters were ovariectomised to eliminate oestradiol (E2 )-negative-feedback and RFRP-3 or saline was centrally administered in the morning or late afternoon. LH concentrations and Lhβ mRNA expression did not differ between morning RFRP-3-and saline-treated groups, although they were markedly suppressed by RFRP-3 administration in the afternoon. However, RFRP-3 inhibition of circulating LH at the time of the surge does not appear to act via the GnRH system because no differences in medial preoptic area Gnrh or RFRP-3 receptor Gpr147 mRNA expression were observed. Rather, RFRP-3 suppressed arcuate nucleus Kiss1 mRNA expression and potentially impacted pituitary gonadotrophs directly. Taken together, these findings reveal time-dependent responsiveness of the reproductive axis to RFRP-3 inhibition, possibly via variation in the sensitivity of arcuate nucleus kisspeptin neurones to this neuropeptide.
Collapse
Affiliation(s)
- Neta Gotlieb
- Department of Psychology, University of California Berkeley, Berkeley, CA, USA
| | - Cydni N. Baker
- Department of Psychology, University of California Berkeley, Berkeley, CA, USA
| | - Jacob Moeller
- Department of Integrative Biology, University of California Berkeley, Berkeley, CA, USA
| | - Lance J. Kriegsfeld
- Department of Psychology, University of California Berkeley, Berkeley, CA, USA
- Department of Integrative Biology, University of California Berkeley, Berkeley, CA, USA
- Graduate Group in Endocrinology, University of California Berkeley, Berkeley, CA, USA
- The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| |
Collapse
|
30
|
Matsuda F, Ohkura S, Magata F, Munetomo A, Chen J, Sato M, Inoue N, Uenoyama Y, Tsukamura H. Role of kisspeptin neurons as a GnRH surge generator: Comparative aspects in rodents and non-rodent mammals. J Obstet Gynaecol Res 2019; 45:2318-2329. [PMID: 31608564 DOI: 10.1111/jog.14124] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 08/30/2019] [Indexed: 02/01/2023]
Abstract
Ovulation is an essential phenomenon for reproduction in mammalian females along with follicular growth. It is well established that gonadal function is controlled by the neuroendocrine system called the hypothalamus-pituitary-gonadal (HPG) axis. Gonadotropin-releasing hormone (GnRH) neurons, localized in the hypothalamus, had been considered to be the head in governing the HPG axis for a long time until the discovery of kisspeptin. In females, induction of ovulation and folliculogenesis has been linked to a surge mode and pulse mode of GnRH releases, respectively. The mechanisms of how the two modes of GnRH are differently regulated had long remained elusive. The discovery of kisspeptin neurons, distributed in two hypothalamic nuclei, such as the arcuate nucleus in the caudal hypothalamus and preoptic area or the anteroventral periventricular nucleus in the rostral hypothalamic regions, and analyses of the detailed functions of kisspeptin neurons have led marked progress on the understanding of different mechanisms regulating GnRH surges (ovulation) and GnRH pulses (folliculogenesis). The present review will focus on the role of kisspeptin neurons as the GnRH surge generator, including the sexual differentiation of the surge generation system and factors that regulate the surge generator. Comparative aspects between mammalian species are especially focused on.
Collapse
Affiliation(s)
- Fuko Matsuda
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Satoshi Ohkura
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Fumie Magata
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Arisa Munetomo
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Jing Chen
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Marimo Sato
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
31
|
Ainani H, El Bousmaki N, Poirel VJ, Achaâban MR, Ouassat M, Piro M, Klosen P, Simonneaux V, El Allali K. The dromedary camel displays annual variation in hypothalamic kisspeptin and Arg-Phe-amide-related peptide-3 according to sex, season, and breeding activity. J Comp Neurol 2019; 528:32-47. [PMID: 31251823 DOI: 10.1002/cne.24736] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 01/25/2023]
Abstract
The dromedary camel (Camelus dromedarius) is a desert mammal whose cycles in reproductive activity ensure that the offspring's birth and weaning coincide with periods of abundant food resources and favorable climate conditions. In this study, we assessed whether kisspeptin (Kp) and arginine-phenylalanine (RF)-amide related peptide-3 (RFRP-3), two hypothalamic peptides known to regulate the mammalian hypothalamo-pituitary gonadal axis, may be involved in the seasonal control of camel's reproduction. Using specific antibodies and riboprobes, we found that Kp neurons are present in the preoptic area (POA), suprachiasmatic (SCN), and arcuate (ARC) nuclei, and that RFRP-3 neurons are present in the paraventricular (PVN), dorsomedial (DMH), and ventromedial (VMH) hypothalamic nuclei. Kp fibers are found in various hypothalamic areas, notably the POA, SCN, PVN, DMH, VMH, supraoptic nucleus, and the ventral and dorsal premammillary nucleus. RFRP-3 fibers are found in the POA, SCN, PVN, DMH, VMH, and ARC. POA and ARC Kp neurons and DMH RFRP-3 neurons display sexual dimorphism with more neurons in female than in male. Both neuronal populations display opposed seasonal variations with more Kp neurons and less RFRP-3 neurons during the breeding (December-January) than the nonbreeding (July-August) season. This study is the first describing Kp and RFRP-3 in the camel's brain with, during the winter period lower RFRP-3 expression and higher Kp expression possibly responsible for the HPG axis activation. Altogether, our data indicate the involvement of both Kp and RFRP-3 in the seasonal control of the dromedary camel's breeding activity.
Collapse
Affiliation(s)
- Hassan Ainani
- Comparative Anatomy Unit, Hassan II Agronomy and Veterinary Institute, Rabat Instituts, Rabat, Morocco.,Institute of Cellular and Integrative Neurosciences, CNRS UPR 3212, University of Strasbourg, Strasbourg, France
| | - Najlae El Bousmaki
- Comparative Anatomy Unit, Hassan II Agronomy and Veterinary Institute, Rabat Instituts, Rabat, Morocco
| | - Vincent-Joseph Poirel
- Institute of Cellular and Integrative Neurosciences, CNRS UPR 3212, University of Strasbourg, Strasbourg, France
| | - Mohamed Rachid Achaâban
- Comparative Anatomy Unit, Hassan II Agronomy and Veterinary Institute, Rabat Instituts, Rabat, Morocco
| | - Mohammed Ouassat
- Comparative Anatomy Unit, Hassan II Agronomy and Veterinary Institute, Rabat Instituts, Rabat, Morocco
| | - Mohammed Piro
- Medicine and Surgical Unit of Domestic animals, Hassan II Agronomy and Veterinary Institute, Rabat Instituts, Rabat, Morocco
| | - Paul Klosen
- Institute of Cellular and Integrative Neurosciences, CNRS UPR 3212, University of Strasbourg, Strasbourg, France
| | - Valérie Simonneaux
- Institute of Cellular and Integrative Neurosciences, CNRS UPR 3212, University of Strasbourg, Strasbourg, France
| | - Khalid El Allali
- Comparative Anatomy Unit, Hassan II Agronomy and Veterinary Institute, Rabat Instituts, Rabat, Morocco
| |
Collapse
|
32
|
Ubuka T, Tsutsui K. Reproductive neuroendocrinology of mammalian gonadotropin-inhibitory hormone. Reprod Med Biol 2019; 18:225-233. [PMID: 31312100 PMCID: PMC6613023 DOI: 10.1002/rmb2.12272] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/25/2019] [Accepted: 04/05/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Gonadotropin-inhibitory hormone (GnIH) was discovered in the Japanese quail brain in 2000 as a hypothalamic neuropeptide that suppresses luteinizing hormone release from cultured quail anterior pituitary. METHODS The authors investigated the existence of mammalian orthologous peptides to GnIH and their physiological functions in the following 19 years of research. MAIN FINDINGS Mammals have orthologous peptide to GnIH, often described RFamide-related peptide, expressed in the hypothalamus and gonads. Mammalian GnIH may also suppress gonadotropin synthesis and release by suppressing gonadotropin-releasing hormone (GnRH) synthesis and release in addition to directly suppressing gonadotropin synthesis and release from the pituitary. Mammalian GnIH may also suppress kisspeptin, a stimulator of GnRH, release. Mammalian GnIH is also expressed in the testis and ovary and suppresses gametogenesis and sex steroid production acting in an autocrine/paracrine manner. Thus, mammalian GnIH may act at all levels of the hypothalamic-pituitary-gonadal axis to suppress reproduction. GnIH may be involved in the regulation of puberty, estrous or menstrual cycle, seasonal reproduction, and stress responses. CONCLUSION Studies suggest that mammalian GnIH is an important neuroendocrine suppressor of reproduction in mammals.
Collapse
Affiliation(s)
- Takayoshi Ubuka
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life ScienceWaseda UniversityShinjukuJapan
| | - Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life ScienceWaseda UniversityShinjukuJapan
| |
Collapse
|
33
|
Angelopoulou E, Quignon C, Kriegsfeld LJ, Simonneaux V. Functional Implications of RFRP-3 in the Central Control of Daily and Seasonal Rhythms in Reproduction. Front Endocrinol (Lausanne) 2019; 10:183. [PMID: 31024442 PMCID: PMC6467943 DOI: 10.3389/fendo.2019.00183] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/05/2019] [Indexed: 12/20/2022] Open
Abstract
Adaptation of reproductive activity to environmental changes is essential for breeding success and offspring survival. In mammals, the reproductive system displays regular cycles of activation and inactivation which are synchronized with seasonal and/or daily rhythms in environmental factors, notably light intensity and duration. Thus, most species adapt their breeding activity along the year to ensure that birth and weaning of the offspring occur at a time when resources are optimal. Additionally, female reproductive activity is highest at the beginning of the active phase during the period of full oocyte maturation, in order to improve breeding success. In reproductive physiology, it is therefore fundamental to delineate how geophysical signals are integrated in the hypothalamo-pituitary-gonadal axis, notably by the neurons expressing gonadotropin releasing hormone (GnRH). Several neurochemicals have been reported to regulate GnRH neuronal activity, but recently two hypothalamic neuropeptides belonging to the superfamily of (Arg)(Phe)-amide peptides, RFRP-3 and kisspeptin, have emerged as critical for the integration of environmental cues within the reproductive axis. The goal of this review is to survey the current understanding of the role played by RFRP-3 in the temporal regulation of reproduction, and consider how its effect might combine with that of kisspeptin to improve the synchronization of reproduction to environmental challenges.
Collapse
Affiliation(s)
- Eleni Angelopoulou
- Institut des Neurosciences Cellulaires et Intégratives (CNRS UPR 3212), Université de Strasbourg, Strasbourg, France
- Netherlands Institute for Neuroscience (NIN), Amsterdam, Netherlands
| | - Clarisse Quignon
- Institut des Neurosciences Cellulaires et Intégratives (CNRS UPR 3212), Université de Strasbourg, Strasbourg, France
| | - Lance J. Kriegsfeld
- Department of Psychology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| | - Valérie Simonneaux
- Institut des Neurosciences Cellulaires et Intégratives (CNRS UPR 3212), Université de Strasbourg, Strasbourg, France
- *Correspondence: Valérie Simonneaux
| |
Collapse
|
34
|
Hu KL, Chang HM, Li R, Yu Y, Qiao J. Regulation of LH secretion by RFRP-3 - From the hypothalamus to the pituitary. Front Neuroendocrinol 2019; 52:12-21. [PMID: 29608929 DOI: 10.1016/j.yfrne.2018.03.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 02/12/2018] [Accepted: 03/29/2018] [Indexed: 12/17/2022]
Abstract
RFamide-related peptides (RFRPs) have long been identified as inhibitors of the hypothalamus-pituitary-gonad axis in mammals. However, less progress has been made in the detailed roles of RFRPs in the control of LH secretion. Recent studies have suggested that RFRP-3 neurons in the hypothalamus can regulate the secretion of LH at different levels, including kisspeptin neurons, GnRH neurons, and the pituitary. Additionally, conflicting results regarding the effects of RFRP-3 on these levels exist. In this review, we collect the latest evidence related to the effects of RFRP-3 neurons in regulating LH secretion by acting on kisspeptin neurons, GnRH neurons, and the pituitary and discuss the potential role of the timely reduction of RFRP-3 signaling in the modulation of the preovulatory LH surge.
Collapse
Affiliation(s)
- Kai-Lun Hu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Hsun-Ming Chang
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Department of Obstetrics and Gynaecology, University of British Columbia, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Rong Li
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Yang Yu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China.
| | - Jie Qiao
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
35
|
Uenoyama Y, Inoue N, Nakamura S, Tsukamura H. Central Mechanism Controlling Pubertal Onset in Mammals: A Triggering Role of Kisspeptin. Front Endocrinol (Lausanne) 2019; 10:312. [PMID: 31164866 PMCID: PMC6536648 DOI: 10.3389/fendo.2019.00312] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 04/30/2019] [Indexed: 01/29/2023] Open
Abstract
Pubertal onset is thought to be timed by an increase in pulsatile gonadotropin-releasing hormone (GnRH)/gonadotropin secretion in mammals. The underlying mechanism of pubertal onset in mammals is still an open question. Evidence accumulated in the last 15 years suggests that kisspeptin/neurokinin B/dynorphin A (KNDy) neurons in the hypothalamic arcuate nucleus play a key role in pubertal onset by triggering pulsatile GnRH/gonadotropin secretin in mammals. Specifically, KNDy neurons are now considered a part of GnRH pulse generator, in which neurokinin B facilitates and dynorphin A inhibits, the synchronized discharge of KNDy neurons in autocrine and/or paracrine manners. Kisspeptin serves as a potent secretagogue of GnRH secretion and thus its release is fundamental to pubertal increase in GnRH/gonadotropin secretion in mammals. Proposed mechanisms inhibiting Kiss1 (kisspeptin gene) expression during childhood to juvenile varies from species to species: we envisage that negative feedback action of estrogen plays a key role in the inhibition of Kiss1 expression in KNDy neurons in rodents and sheep, whereas estrogen-independent inhibition of kisspeptin secretion by γ-amino butyric acid or neuropeptide Y are suggested to be responsible for the pre-pubertal suppression of GnRH/gonadotropin secretion in primates. Taken together, the timing of pubertal onset is postulated to be controlled by upstream regulators for kisspeptin biosynthesis and secretion in mammals.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
- *Correspondence: Yoshihisa Uenoyama
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Sho Nakamura
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
36
|
Simonneaux V. A Kiss to drive rhythms in reproduction. Eur J Neurosci 2018; 51:509-530. [DOI: 10.1111/ejn.14287] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/08/2018] [Accepted: 11/19/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Valérie Simonneaux
- Institut des Neurosciences Cellulaires et IntégrativesCNRSUniversité de Strasbourg Strasbourg France
| |
Collapse
|
37
|
Ahmad Pampori Z, Ahmad Sheikh A, Aarif O, Hasin D, Ahmad Bhat I. Physiology of reproductive seasonality in sheep – an update. BIOL RHYTHM RES 2018. [DOI: 10.1080/09291016.2018.1548112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Zahoor Ahmad Pampori
- Division of Veterinary Physiology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, India
| | - Aasif Ahmad Sheikh
- Division of Veterinary Physiology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, India
| | - Ovais Aarif
- Division of Veterinary Physiology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, India
| | - Dilruba Hasin
- Division of Veterinary Physiology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, India
| | - Irfan Ahmad Bhat
- Division of Veterinary Physiology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, India
| |
Collapse
|
38
|
Hugon-Rodin J, Yoshii K, Lahlou N, Flandrin J, Gompel A, de Roux N. Complete Kisspeptin Receptor Inactivation Does Not Impede Exogenous GnRH-Induced LH Surge in Humans. J Clin Endocrinol Metab 2018; 103:4482-4490. [PMID: 30124894 DOI: 10.1210/jc.2018-00410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 08/13/2018] [Indexed: 02/13/2023]
Abstract
CONTEXT Mutations in the kisspeptin receptor (KISS1R) gene have been reported in a few patients with normosmic congenital hypogonadotropic hypogonadism (nCHH) (OMIM #146110). OBJECTIVES To describe a female patient with nCHH and a novel homozygous KISS1R mutation and to assess the role of kisspeptin pathway to induce an ovulation by GnRH pulse therapy. DESIGN, SETTING, AND INTERVENTION Observational study of a patient including genetic and kisspeptin receptor functions and treatment efficiency using a GnRH pump. MAIN OUTCOME MEASURE Response to pulsatile GnRH therapy. RESULTS A partial isolated gonadotropic deficiency was diagnosed in a 28-year-old woman with primary amenorrhea and no breast development. A novel homozygous c.953T>C variant was identified in KISS1R. This mutation led to substitution of leucine 318 for proline (p.Leu318Pro) in the seventh transmembrane domain of KISS1R. Signaling via the mutated receptor was profoundly impaired in HEK293-transfected cells. The mutated receptor was not detected on the membrane of HEK293-transfected cells. After several pulsatile GnRH therapy cycles, an LH surge with ovulation and pregnancy was obtained. CONCLUSION GnRH pulsatile therapy can induce an LH surge in a woman with a mutated KISS1R, which was previously thought to be completely inactivated in vivo.
Collapse
Affiliation(s)
- Justine Hugon-Rodin
- Paris Descartes University, Gynecology Endocrinology Unit, Port-Royal Teaching Hospital, AP-HP, Paris, France
| | - Keisuke Yoshii
- Paris Diderot University, Sorbonne Paris Cité, U1141, Inserm, Paris, France
| | | | - Jennifer Flandrin
- Paris Descartes University, Gynecology Endocrinology Unit, Port-Royal Teaching Hospital, AP-HP, Paris, France
| | - Anne Gompel
- Paris Descartes University, Gynecology Endocrinology Unit, Port-Royal Teaching Hospital, AP-HP, Paris, France
| | - Nicolas de Roux
- Paris Diderot University, Sorbonne Paris Cité, U1141, Inserm, Paris, France
- AP-HP, Biochemistry Laboratory, Robert Debré Teaching Hospital, Paris, France
| |
Collapse
|
39
|
Ubuka T, Tsutsui K. Comparative and Evolutionary Aspects of Gonadotropin-Inhibitory Hormone and FMRFamide-Like Peptide Systems. Front Neurosci 2018; 12:747. [PMID: 30405335 PMCID: PMC6200920 DOI: 10.3389/fnins.2018.00747] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 09/28/2018] [Indexed: 11/13/2022] Open
Abstract
Gonadotropin-inhibitory hormone (GnIH) is a hypothalamic neuropeptide that was found in the brain of Japanese quail when investigating the existence of RFamide peptides in birds. GnIH was named because it decreased gonadotropin release from cultured anterior pituitary, which was located in the hypothalamo-hypophysial system. GnIH and GnIH precursor gene related peptides have a characteristic C-terminal LPXRFamide (X = L or Q) motif that is conserved in jawed vertebrates. Orthologous peptides to GnIH are also named RFamide related peptide or LPXRFamide peptide from their structure. A G-protein coupled receptor GPR147 is the primary receptor for GnIH. Similarity-based clustering of neuropeptide precursors in metazoan species indicates that GnIH precursor of vertebrates is evolutionarily related to FMRFamide precursor of mollusk and nematode. FMRFamide peptide is the first RFamide peptide that was identified from the ganglia of the venus clam. In order to infer the evolutionary history of the GnIH-GnIH receptor system we investigate the structural similarities between GnIH and its receptor and well-studied nematode Caenorhabditis elegans (C. elegans) FMRFamide-like peptides (FLPs) and their receptors. We also compare the functions of FLPs of nematode with GnIH of chordates. A multiple sequence alignment and phylogenetic analyses of GnIH, neuropeptide FF (NPFF), a paralogous peptide of GnIH, and FLP precursors have shown that GnIH and NPFF precursors belong to different clades and some FLP precursors have structural similarities to either precursor. The peptide coding regions of FLP precursors in the same clade align well with those of GnIH or NPFF precursors. Alignment of GnIH (LPXRFa) peptides of chordates and FLPs of C. elegans grouped the peptides into five groups according to the last C-terminal amino acid sequences, which were MRFa, LRFa, VRFa, IRFa, and PQRFa. Phylogenetic analysis of receptors suggested that GPR147 has evolutionary relationships with FLP receptors, which regulate reproduction, aggression, locomotion, and feeding. GnIH and some FLPs mediate the effect of stress on reproduction and behavior, which may also be a conserved property of these peptide systems. Future studies are needed to investigate the mechanism of how neuropeptide precursor genes are mutated to evolve new neuropeptides and their inheritance.
Collapse
Affiliation(s)
- Takayoshi Ubuka
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Shinjuku, Japan
| | - Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Shinjuku, Japan
| |
Collapse
|
40
|
Kriegsfeld LJ, Jennings KJ, Bentley GE, Tsutsui K. Gonadotrophin-inhibitory hormone and its mammalian orthologue RFamide-related peptide-3: Discovery and functional implications for reproduction and stress. J Neuroendocrinol 2018; 30:e12597. [PMID: 29624758 PMCID: PMC6263162 DOI: 10.1111/jne.12597] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 03/26/2018] [Accepted: 03/28/2018] [Indexed: 02/06/2023]
Abstract
At the turn of the millennium, a neuropeptide with pronounced inhibitory actions on avian pituitary gonadotrophin secretion was identified and named gonadotrophin-inhibitory hormone (GnIH). Across bird species, GnIH acts at the level of the pituitary and the gonadotrophin-releasing hormone (GnRH) neuronal system to inhibit reproduction. Subsequent to this initial discovery, orthologues of GnIH have been identified and characterised across a broad range of species. In many vertebrates, the actions of GnIH and its orthologues serve functional roles analogous to those seen in birds. In other cases, GnIH and its orthologues exhibit more diverse actions dependent on sex, species, season and reproductive condition. The present review highlights the discovery and functional implications of GnIH across species, focusing on research domains in which the significance of this neuropeptide has been explored most.
Collapse
Affiliation(s)
- Lance J. Kriegsfeld
- Department of Psychology, University of California, Berkeley, California 94720, USA
- The Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720, USA
- Corresponding Author: Lance J. Kriegsfeld, Ph.D. Neurobiology Laboratory, Department of Psychology and The Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, California 94720-1650, USA, Phone: (510) 642-5148; Fax: (510) 642-5293;
| | - Kimberly J. Jennings
- Department of Psychology, University of California, Berkeley, California 94720, USA
| | - George E. Bentley
- The Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720, USA
- Department of Integrative Biology, University of California, Berkeley, California 94720, USA
| | - Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences, Department of Biology, Waseda University, and Center for Medical Life Science of Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| |
Collapse
|
41
|
Weems PW, Lehman MN, Coolen LM, Goodman RL. The Roles of Neurokinins and Endogenous Opioid Peptides in Control of Pulsatile LH Secretion. VITAMINS AND HORMONES 2018; 107:89-135. [PMID: 29544644 DOI: 10.1016/bs.vh.2018.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Work over the last 15 years on the control of pulsatile LH secretion has focused largely on a set of neurons in the arcuate nucleus (ARC) that contains two stimulatory neuropeptides, critical for fertility in humans (kisspeptin and neurokinin B (NKB)) and the inhibitory endogenous opioid peptide (EOP), dynorphin, and are now known as KNDy (kisspeptin-NKB-dynorphin) neurons. In this review, we consider the role of each of the KNDy peptides in the generation of GnRH pulses and the negative feedback actions of ovarian steroids, with an emphasis on NKB and dynorphin. With regard to negative feedback, there appear to be important species differences. In sheep, progesterone inhibits GnRH pulse frequency by stimulating dynorphin release, and estradiol inhibits pulse amplitude by suppressing kisspeptin. In rodents, the role of KNDy neurons in estrogen negative feedback remains controversial, progesterone may inhibit GnRH via dynorphin, but the physiological significance of this action is unclear. In primates, an EOP, probably dynorphin, mediates progesterone negative feedback, and estrogen inhibits kisspeptin expression. In contrast, there is now compelling evidence from several species that kisspeptin is the output signal from KNDy neurons that drives GnRH release during a pulse and may also act within the KNDy network to affect pulse frequency. NKB is thought to act within this network to initiate each pulse, although there is some redundancy in tachykinin signaling in rodents. In ruminants, dynorphin terminates GnRH secretion at the end of pulse, most likely acting on both KNDy and GnRH neurons, but the data on the role of this EOP in rodents are conflicting.
Collapse
Affiliation(s)
- Peyton W Weems
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Michael N Lehman
- University of Mississippi Medical Center, Jackson, MS, United States
| | - Lique M Coolen
- University of Mississippi Medical Center, Jackson, MS, United States
| | | |
Collapse
|
42
|
Yeo SH, Colledge WH. The Role of Kiss1 Neurons As Integrators of Endocrine, Metabolic, and Environmental Factors in the Hypothalamic-Pituitary-Gonadal Axis. Front Endocrinol (Lausanne) 2018; 9:188. [PMID: 29755406 PMCID: PMC5932150 DOI: 10.3389/fendo.2018.00188] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/06/2018] [Indexed: 01/06/2023] Open
Abstract
Kisspeptin-GPR54 signaling in the hypothalamus is required for reproduction and fertility in mammals. Kiss1 neurons are key regulators of gonadotropin-releasing hormone (GnRH) release and modulation of the hypothalamic-pituitary-gonadal (HPG) axis. Arcuate Kiss1 neurons project to GnRH nerve terminals in the median eminence, orchestrating the pulsatile secretion of luteinizing hormone (LH) through the intricate interaction between GnRH pulse frequency and the pituitary gonadotrophs. Arcuate Kiss1 neurons, also known as KNDy neurons in rodents and ruminants because of their co-expression of neurokinin B and dynorphin represent an ideal hub to receive afferent inputs from other brain regions in response to physiological and environmental changes, which can regulate the HPG axis. This review will focus on studies performed primarily in rodent and ruminant species to explore potential afferent inputs to Kiss1 neurons with emphasis on the arcuate region but also considering the rostral periventricular region of the third ventricle (RP3V). Specifically, we will discuss how these inputs can be modulated by hormonal, metabolic, and environmental factors to control gonadotropin secretion and fertility. We also summarize the methods and techniques that can be used to study functional inputs into Kiss1 neurons.
Collapse
|
43
|
He Y, Sun W, Yu J. Is precocious puberty linked to hypothalamic expression of arginine-phenylalanine-amide-related peptide? IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2017; 20:1074-1078. [PMID: 29147481 PMCID: PMC5673690 DOI: 10.22038/ijbms.2017.9397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The up-regulation and down-regulation of gonadotropin-releasing hormone (GnRH) in central precocious puberty is not yet known. However, recent advances in neuroendocrinology have shown the controlling role of arginine-phenylalanine RF-amide-related peptides (RFRPs) on GnRH secretion in different phenomenon of reproduction such as estrus cycle and pregnancy, but the exact role of RFRPs in puberty and its related pathologic condition, precocious puberty, is not clear yet. This paper hypothesizes that RFRP is a regulatory peptide of puberty and might prevent the precocious puberty. On the basis of previous studies on hormonal fluctuations at the time of puberty, RFRP might have a role on controlling of premature secretion of GnRH and avoiding central precocious puberty.
Collapse
Affiliation(s)
- Yuanyuan He
- Department of Traditional Chinese Medicine, Children's Hospital of Fudan University, Shanghai, 200032, China
| | - Wen Sun
- Department of Traditional Chinese Medicine, Children's Hospital of Fudan University, Shanghai, 200032, China
| | - Jian Yu
- Department of Traditional Chinese Medicine, Children's Hospital of Fudan University, Shanghai, 200032, China
| |
Collapse
|
44
|
George JT, Hendrikse M, Veldhuis JD, Clarke IJ, Anderson RA, Millar RP. Effect of gonadotropin-inhibitory hormone on luteinizing hormone secretion in humans. Clin Endocrinol (Oxf) 2017; 86:731-738. [PMID: 28186349 DOI: 10.1111/cen.13308] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/09/2017] [Accepted: 02/03/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Gonadotropin-inhibitory hormone (GnIH, human homologue of RFRP-3) suppresses gonadotropin secretion in animal models, but its effects have not been studied in the human. OBJECTIVE We tested the hypotheses that exogenous GnIH inhibits LH secretion (i) in postmenopausal women and (ii) in men concurrently administered exogenous kisspeptin. DESIGN Following in vitro and in vivo preclinical studies to functionally characterize the GnIH peptide, a dose-finding study (human GnIH: 1·5-150 μg/kg/h, iv for 3 h) was undertaken, and 50 μg/kg/h selected for further evaluation. Five postmenopausal women were administered 50 μg/kg/h iv infusion for 3 h or vehicle on two separate days. Four men were administered kisspeptin-10 (0·3 μg/kg iv bolus) with simultaneous infusion of GnIH (50 μg/kg/h, iv for 3 h) or vehicle. PARTICIPANTS Healthy postmenopausal women (mean age 58 ± 2 years, LH: 30·8 ± 2·9 IU/l, FSH: 78·7 ± 6·4 IU/l, oestradiol: <50 pmol/l) and men (39·8 ± 2·1 years, mean total testosterone 12·1 ± 1·8 nmol/l, LH 2·2 ± 0·2 IU/l). PRIMARY OUTCOME Change in area under curve (AUC) of LH during GnIHvs vehicle. RESULTS During GnIH administration in postmenopausal women, LH secretion decreased (ΔAUC: -9·9 ± 1·8 IU/3 h) vs vehicle (ΔAUC: -0·5 ± 1·7 IU/3 h; P = 0·02). Kisspeptin-10-stimulated LH responses in men were not affected by GnIH co-administration (60-min AUC of LH 6·2 ± 0·8 IU/h with kisspeptin-10 alone, 6·3 ± 1·0 IU/h, kisspeptin-10 with GnIH, P = 0·72). Exogenous GnIH was well tolerated, with no adverse events reported. CONCLUSIONS Gonadotropin-inhibitory hormone decreased LH secretion in postmenopausal women in this first-in-human study. Kisspeptin-stimulated LH secretion in men was not inhibited during concomitant administration of GnIH.
Collapse
Affiliation(s)
- J T George
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Diabetes Trials Unit, Endocrinology and Metabolism, Oxford Centre for Diabetes, Oxford, UK
| | - M Hendrikse
- Department of Medical Biochemistry and Institute for Infectious Diseases and Molecular Medicine, University of Cape Town, Observatory, South Africa
| | - J D Veldhuis
- Endocrine Research Unit, Center for Translational Science Activities, Mayo Clinic, Rochester, MN, USA
| | - I J Clarke
- Department of Physiology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| | - R A Anderson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - R P Millar
- Department of Medical Biochemistry and Institute for Infectious Diseases and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Mammal Research Institute and Centre for Neuroendocrinology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
45
|
Stevenson TJ. Environmental and hormonal regulation of epigenetic enzymes in the hypothalamus. J Neuroendocrinol 2017; 29. [PMID: 28370682 DOI: 10.1111/jne.12471] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 03/09/2017] [Accepted: 03/25/2017] [Indexed: 12/13/2022]
Abstract
Neuroendocrine structures integrate a vast range of external cues and internal signals that, in turn, result in adaptive physiological responses. Emerging data indicate that light, social cues, stress and energy balance stimulate relatively short- and long-term genomic modifications in discrete neuroendocrine structures, which are mediated by epigenetic mechanisms. Moreover, environmentally-induced fluctuations in the synthesis of local hypothalamic and circulating hormones provide an internal signal that contributes to the extensive neuroendocrine genomic plasticity. This review examines the impact of environmental stimuli and endogenous hormonal signals on the regulation of epigenetic enzymes in key neuroendocrine structures. The data discussed are predominantly derived from studies in the neuroendocrine control of seasonal reproduction and the impact of social stress in rodent models. The perspective presented considers the role of oestrogen and glucocorticoids as the primary catalysts for inducing epigenetic modifications (eg, DNA methylation) in specific neuroendocrine structures. Oestrogen and glucocorticoid actions suggest: (i) a preferential action for specific epigenetic enzymes and (ii) nucleus- and cell-specific modifications. Untangling the complex web of hormonal regulation of methylation and acetylation will enhance our understanding of short- and long-term changes in epigenetic enzymes that generate adaptive and pathological neuroendocrine responses.
Collapse
Affiliation(s)
- T J Stevenson
- Institute for Biological and Environmental Sciences, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
46
|
Marraudino M, Miceli D, Farinetti A, Ponti G, Panzica G, Gotti S. Kisspeptin innervation of the hypothalamic paraventricular nucleus: sexual dimorphism and effect of estrous cycle in female mice. J Anat 2017; 230:775-786. [PMID: 28295274 DOI: 10.1111/joa.12603] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2017] [Indexed: 01/11/2023] Open
Abstract
The hypothalamic paraventricular nucleus (PVN) is the major autonomic output area of the hypothalamus and a critical regulatory center for energy homeostasis. The organism's energetic balance is very important for both the regular onset of puberty and regulation of fertility. Several studies have suggested a relationship among neural circuits controlling food intake, energy homeostasis and the kisspeptin peptide. The kisspeptin system is clustered in two main groups of cell bodies [the anterior ventral periventricular region (AVPV) and the arcuate nucleus (ARC)] projecting mainly to gonadotropin-releasing hormone (GnRH) neurons and to a few other locations, including the PVN. In the present study, we investigated the distribution of the kisspeptin fibers within the PVN of adult CD1 mice. We observed a significant sexual dimorphism for AVPV and ARC, as well as for the PVN innervation. Kisspeptin fibers showed a different density within the PVN, being denser in the medial part than in the lateral one; moreover, in female, the density changed, according to different phases of the estrous cycle (the highest density being in estrus phase). The presence of a profound effect of estrous cycle on the kisspeptin immunoreactivity in AVPV (with a higher signal in estrus) and ARC, and the strong co-localization between kisspeptin and NkB only in ARC and not in PVN suggested that the majority of the kisspeptin fibers found in the PVN might arise directly from AVPV.
Collapse
Affiliation(s)
- Marilena Marraudino
- Department of Neuroscience, Laboratory of Neuroendocrinology, University of Torino, Torino, Italy.,Neuroscience Institute Cavalieri-Ottolenghi (NICO), Orbassano, Italy
| | - Dèsirèe Miceli
- Department of Neuroscience, Laboratory of Neuroendocrinology, University of Torino, Torino, Italy.,Neuroscience Institute Cavalieri-Ottolenghi (NICO), Orbassano, Italy
| | - Alice Farinetti
- Department of Neuroscience, Laboratory of Neuroendocrinology, University of Torino, Torino, Italy.,Neuroscience Institute Cavalieri-Ottolenghi (NICO), Orbassano, Italy
| | - Giovanna Ponti
- Neuroscience Institute Cavalieri-Ottolenghi (NICO), Orbassano, Italy.,Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| | - GianCarlo Panzica
- Department of Neuroscience, Laboratory of Neuroendocrinology, University of Torino, Torino, Italy.,Neuroscience Institute Cavalieri-Ottolenghi (NICO), Orbassano, Italy
| | - Stefano Gotti
- Department of Neuroscience, Laboratory of Neuroendocrinology, University of Torino, Torino, Italy.,Neuroscience Institute Cavalieri-Ottolenghi (NICO), Orbassano, Italy
| |
Collapse
|
47
|
RFamide-related peptide-3 (RFRP-3) suppresses sexual maturation in a eusocial mammal. Proc Natl Acad Sci U S A 2017; 114:1207-1212. [PMID: 28096421 DOI: 10.1073/pnas.1616913114] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Neuroendocrine mechanisms underlying social inhibition of puberty are not well understood. Here, we use a model exhibiting the most profound case of pubertal suppression among mammals to explore a role for RFamide-related peptide-3 [RFRP-3; mammalian ortholog to gonadotropin-inhibitory hormone (GnIH)] in neuroendocrine control of reproductive development. Naked mole rats (NMRs) live in sizable colonies where breeding is monopolized by two to four dominant animals, and no other members exhibit signs of puberty throughout their lives unless they are removed from the colony. Because of its inhibitory action on the reproductive axis in other vertebrates, we investigated the role of RFRP-3 in social reproductive suppression in NMRs. We report that RFRP-3 immunofluorescence expression patterns and RFRP-3/GnRH cross-talk are largely conserved in the NMR brain, with the exception of the unique presence of RFRP-3 cell bodies in the arcuate nucleus (Arc). Immunofluorescence comparisons revealed that central expression of RFRP-3 is altered by reproductive status, with RFRP-3 immunoreactivity enhanced in the paraventricular nucleus, dorsomedial nucleus, and Arc of reproductively quiescent NMRs. We further observed that exogenous RFRP-3 suppresses gonadal steroidogenesis and mating behavior in NMRs given the opportunity to undergo puberty. Together, our findings establish a role for RFRP-3 in preserving reproductive immaturity, and challenge the view that stimulatory peptides are the ultimate gatekeepers of puberty.
Collapse
|
48
|
Beymer M, Henningsen J, Bahougne T, Simonneaux V. The role of kisspeptin and RFRP in the circadian control of female reproduction. Mol Cell Endocrinol 2016; 438:89-99. [PMID: 27364888 DOI: 10.1016/j.mce.2016.06.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 06/24/2016] [Accepted: 06/26/2016] [Indexed: 12/14/2022]
Abstract
In female mammals, reproduction shows ovarian and daily rhythms ensuring that the timing of the greatest fertility coincides with maximal activity and arousal. The ovarian cycle, which lasts from a few days to a few weeks, depends on the rhythm of follicle maturation and ovarian hormone production, whereas the daily cycle depends on a network of circadian clocks of which the main one is located in the suprachiasmatic nuclei (SCN). In the last ten years, major progress has been made in the understanding of the neuronal mechanisms governing mammalian reproduction with the finding that two hypothalamic Arg-Phe-amide peptides, kisspeptin (Kp) and RFRP, regulate GnRH neurons. In this review we discuss the pivotal role of Kp and RFRP neurons at the interface between the SCN clock signal and GnRH neurons to properly time gonadotropin-induced ovulation. We also report recent findings indicating that these neurons may be part of the multi-oscillatory circadian system that times female fertility. Finally, we will discuss recent investigations indicating a role, and putative therapeutic use, of these neuropeptides in human reproduction.
Collapse
Affiliation(s)
- Matthew Beymer
- Institut des Neurosciences Cellulaires et Intégratives (CNRS UPR 3212), 5 rue Blaise Pascal, 67084 Strasbourg, France
| | - Jo Henningsen
- Institut des Neurosciences Cellulaires et Intégratives (CNRS UPR 3212), 5 rue Blaise Pascal, 67084 Strasbourg, France
| | - Thibault Bahougne
- Institut des Neurosciences Cellulaires et Intégratives (CNRS UPR 3212), 5 rue Blaise Pascal, 67084 Strasbourg, France; Service d'Endocrinologie et Diabète, Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Valérie Simonneaux
- Institut des Neurosciences Cellulaires et Intégratives (CNRS UPR 3212), 5 rue Blaise Pascal, 67084 Strasbourg, France.
| |
Collapse
|
49
|
Ullah R, Shen Y, Zhou YD, Huang K, Fu JF, Wahab F, Shahab M. Expression and actions of GnIH and its orthologs in vertebrates: Current status and advanced knowledge. Neuropeptides 2016; 59:9-20. [PMID: 27255391 DOI: 10.1016/j.npep.2016.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/01/2016] [Accepted: 05/23/2016] [Indexed: 12/20/2022]
Abstract
The physiology of reproduction is very complex and is regulated by multiple factors, including a number of hypothalamic neuropeptides. In last few decades, various neuropeptides have been discovered to be involved in stimulation or inhibition of reproduction. In 2000, Tsutsui and colleagues uncovered gonadotropin-inhibitory hormone (GnIH), a neuropeptide generating inhibitory drive to the reproductive axis, in the brain of Coturnix quail. Afterward, GnIH orthologs were discovered in other vertebrates from fish to mammals including human. In these vertebrates, all the discovered GnIH and its ortholgs have LPXRFamide (X=L or Q) sequence at C-terminus. GnIH orthologs of mammals and primates are also termed as RFamide-related peptide (RFRP)-1 and -3 that too have an LPXRFamide (X=L or Q) motif at their C-terminus. GnIH and its orthologs form a member of the RFamide peptide family. GnIH signals via its canonical G protein coupled receptor 147 (GPR147). Both GnIH and GPR147 are expressed in hypothalamus and other brain regions. Besides actions through the hypothalamic GnRH and kisspeptinergic neurons, GnIH-GPR147 signaling exerts inhibitory effect on the reproductive axis via pituitary gonadotropes and directly at gonadal level. Various factors including availability and quality of food, photoperiod, temperature, social interaction, various stresses and some diseases modulate GnIH-GPR147 signaling. In this review, we have discussed expression and actions of GnIH and its orthologs in vertebrates. Special emphasis is given on the role of GnIH-GPR147 signaling pathway in the regulation of reproduction. We have also reviewed and discussed currently available literature on the participation of GnIH-GPR147 signaling pathway in the stress modulation of reproduction.
Collapse
Affiliation(s)
- Rahim Ullah
- Department of Endocrinology, Children Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China; Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan; Key Laboratory of Medical Neurobiology of Ministry of Health and Zhejiang Province, Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, PR China
| | - Yi Shen
- Key Laboratory of Medical Neurobiology of Ministry of Health and Zhejiang Province, Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, PR China
| | - Yu-Dong Zhou
- Key Laboratory of Medical Neurobiology of Ministry of Health and Zhejiang Province, Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, PR China
| | - Ke Huang
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan; Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Jun-Fen Fu
- Department of Endocrinology, Children Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Fazal Wahab
- Stem Cell Biology Unit, German Primate Center, Leibniz-Institute for Primate Research, Kellnerweg 4, 37077 Gottingen, Germany
| | - Muhammad Shahab
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
50
|
Abstract
The gonadotropin-releasing hormone (GnRH) neuronal network generates pulse and surge modes of gonadotropin secretion critical for puberty and fertility. The arcuate nucleus kisspeptin neurons that innervate the projections of GnRH neurons in and around their neurosecretory zone are key components of the pulse generator in all mammals. By contrast, kisspeptin neurons located in the preoptic area project to GnRH neuron cell bodies and proximal dendrites and are involved in surge generation in female rodents (and possibly other species). The hypothalamic-pituitary-gonadal axis develops embryonically but, apart from short periods of activation immediately after birth, remains suppressed through a combination of gonadal and non-gonadal mechanisms. At puberty onset, the pulse generator reactivates, probably owing to progressive stimulatory influences on GnRH neurons from glial and neurotransmitter signalling, and the re-emergence of stimulatory arcuate kisspeptin input. In females, the development of pulsatile gonadotropin secretion enables final maturation of the surge generator that ultimately triggers the first ovulation. Representation of the GnRH neuronal network as a series of interlocking functional modules could help conceptualization of its functioning in different species. Insights into pulse and surge generation are expected to aid development of therapeutic strategies ameliorating pubertal disorders and infertility in the clinic.
Collapse
Affiliation(s)
- Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| |
Collapse
|