1
|
Renwick AN, Whitlock BK, Nestor CC, Daniel JA, Strickland L, Lear AS, Adkins M, Griffin C, Esteller-Vico A. Chronic inflammation decreases arcuate kisspeptin expression in male sheep. Domest Anim Endocrinol 2024; 89:106868. [PMID: 38901139 PMCID: PMC11366492 DOI: 10.1016/j.domaniend.2024.106868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/17/2024] [Accepted: 06/02/2024] [Indexed: 06/22/2024]
Abstract
Lipopolysaccharide (LPS) from Gram-negative bacteria induces an immune response and impairs reproduction through suppression of gonadotropin releasing hormone (GnRH), subsequently luteinizing hormone (LH) secretion. While there is evidence that acute inflammation inhibits kisspeptin, little is known about the impact of chronic inflammation on this key reproductive neuropeptide in livestock species. Thus, we sought to examine a central mechanism whereby LPS suppresses LH secretion in sheep. Twenty wethers were randomly assigned to one of five treatment groups: control (CON; n=4), single acute IV LPS dose (SAD; n=4), daily acute IV LPS dose (DAD; n=4), daily increasing IV LPS dose (DID; n=4), and chronic subcutaneous LPS dose (CSD; n=4). On Days 1 and 7, blood samples were collected every 12 minutes for 360 minutes using jugular venipuncture. Following blood collection on Day 7, all animals were euthanized, brain tissue was perfused with 4% paraformaldehyde, and hypothalamic blocks were removed and processed for immunohistochemistry. On Day 1, LH pulse frequency was significantly lower (p=0.02) in SAD (0.25 ± 0.1 pulses/hour), DAD (0.25 ± 0.1 pulses/hour), DID (0.35 ± 0.1 pulses/hour), and CSD (0.40 ± 0.1 pulses/hour) compared to CON (0.70 ±0.1 pulses/hour). On Day 7, only DID animals (0.35 ± 0.1 pulses/hour) had significantly lower (p=0.049) LH pulse frequency compared to controls (0.85 ± 0.1 pulse/hour). Furthermore, only DID animals (33.3 ± 10.9 cells/section/animal) had significantly fewer (p=0.001) kisspeptin-immunopositive cells compared to controls (82.6 ± 13.6 cells/section/animal). Taken together, we suggest that daily increasing doses of LPS is a powerful inhibitor of kisspeptin neurons in young male sheep and a physiologically relevant model to examine the impact of chronic inflammation on the reproductive axis in livestock.
Collapse
Affiliation(s)
- A N Renwick
- Large Animal Clinical Sciences Department, University of Tennessee College of Veterinary Medicine, Knoxville, TN
| | - B K Whitlock
- Large Animal Clinical Sciences Department, University of Tennessee College of Veterinary Medicine, Knoxville, TN.
| | - C C Nestor
- Department of Animal Science, North Carolina State University, Raleigh, NC
| | - J A Daniel
- Animal Science Department, Berry College, Rome, GA
| | - L Strickland
- Large Animal Clinical Sciences Department, University of Tennessee College of Veterinary Medicine, Knoxville, TN; Department of Animal Science, University of Tennessee, Knoxville, TN
| | - A S Lear
- Large Animal Clinical Sciences Department, University of Tennessee College of Veterinary Medicine, Knoxville, TN
| | - M Adkins
- Large Animal Clinical Sciences Department, University of Tennessee College of Veterinary Medicine, Knoxville, TN
| | - C Griffin
- Large Animal Clinical Sciences Department, University of Tennessee College of Veterinary Medicine, Knoxville, TN
| | - A Esteller-Vico
- Biomedical and Diagnostic Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN
| |
Collapse
|
2
|
González-Flores O, Domínguez-Ordóñez R, Delgado-Macuil RJ, Tlachi-López JL, Luna-Hernández A, Montes-Narváez O, Pfaus JG, García-Juárez M. Participation of kisspeptin, progesterone, and GnRH receptors on lordosis behavior induced by kisspeptin. Physiol Behav 2024; 283:114609. [PMID: 38851441 DOI: 10.1016/j.physbeh.2024.114609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
The neuropeptide kisspeptin (Kiss) is crucial in regulating the hypothalamic-pituitary-gonadal axis. It is produced by two main groups of neurons in the hypothalamus: the rostral periventricular region around the third ventricle and the arcuate nucleus. Kiss is the peptide product of the KiSS-1 gene and serves as the endogenous agonist for the GPR54 receptor. The Kiss/GPR54 system functions as a critical regulator of the reproductive system. Thus, we examined the effect of intracerebroventricular administration of 3 μg of Kiss to the right lateral ventricle of ovariectomized rats primed with a dose of 5 μg subcutaneous (sc) of estradiol benzoate (EB). Kiss treatment increased the lordosis quotient at all times tested. However, the lordosis reflex score was comparatively lower yet still significant compared to the control group. To investigate receptor specificity and downstream mechanisms on lordosis, we infused 10 μg of GPR54 receptor antagonist, Kiss-234, 5 μg of the progestin receptor antagonist, RU486, or 3 μg of antide, a gonadotropin-releasing hormone-1 (GnRH-1) receptor antagonist, to the right lateral ventricle 30 min before an infusion of 3 μg of Kiss. Results demonstrated a significant reduction in the facilitation of lordosis behavior by Kiss at 60 and 120 min when Kiss-234, RU486, or antide were administered. These findings suggest that Kiss stimulates lordosis expression by activating GPR54 receptors on GnRH neurons and that Kiss/GPR54 system is an essential intermediary by which progesterone activates GnRH.
Collapse
Affiliation(s)
- Oscar González-Flores
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México
| | - Raymundo Domínguez-Ordóñez
- Licenciatura en Ingeniería Agronómica y Zootecnia, CRC, Benemérita Universidad Autónoma de, Puebla, México
| | - Raul Jacobo Delgado-Macuil
- Centro de Investigación en Biotecnología Aplicada, Instituto Politécnico Nacional, Santa Inés, Tecuexcomac, Tlaxcala, México
| | | | - Ailyn Luna-Hernández
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - Omar Montes-Narváez
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - James G Pfaus
- Center for Sexual Health and Intervention, Czech National Institute of Mental Health, Klecany, Czech Republic; Department of Psychology and Life Sciences, Charles University, Prague, Czech Republic
| | - Marcos García-Juárez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México.
| |
Collapse
|
3
|
West S, Garza V, Cardoso R. Puberty in beef heifers: effects of prenatal and postnatal nutrition on the development of the neuroendocrine axis. Anim Reprod 2024; 21:e20240048. [PMID: 39176002 PMCID: PMC11340802 DOI: 10.1590/1984-3143-ar2024-0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/29/2024] [Indexed: 08/24/2024] Open
Abstract
Reproductive maturation is a complex physiological process controlled by the neuroendocrine system and is characterized by an increase in gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) pulsatile secretion. Nutrition during early development is a key factor regulating puberty onset, which is defined as first ovulation in females. In heifers, nutrient restriction after weaning delays puberty, whereas elevated levels of nutrition and energy reserves advance reproductive maturation. Recent studies in cattle and other animal models have shown that the dam's nutrition during gestation can also program the neuroendocrine system in the developing fetus and has the potential to alter timing of puberty in the offspring. Among the metabolic signals that modulate brain development and control timing of puberty is leptin, a hormone produced primarily by adipocytes that communicates energy status to the brain. Leptin acts within the arcuate nucleus of the hypothalamus to regulate GnRH secretion via an upstream network of neurons that includes neurons that express neuropeptide Y (NPY), an orexigenic peptide with inhibitory effects on GnRH secretion, and alpha melanocyte-stimulating hormone (αMSH), an anorexigenic peptide with excitatory effects on GnRH neurons. Another important population of neurons are KNDy neurons, neurons in the arcuate nucleus that co-express the neuropeptides kisspeptin, neurokinin B, and dynorphin and have strong stimulatory effects on GnRH secretion. Our studies in beef heifers indicate that increased nutrition between 4 to 8 months of age advances puberty by diminishing NPY inhibitory tone and by increasing excitatory inputs of αMSH and kisspeptin, which collectively lead to increased GnRH/LH pulsatility. Our ongoing studies indicate that different planes of nutrition during gestation can alter maternal leptin concentrations and promote changes in the fetal brain. Nonetheless, at least in Bos indicus-influenced heifers, deficits programmed prenatally can be overcome by adequate postnatal nutrition without negatively impacting age at puberty or subsequent fertility.
Collapse
Affiliation(s)
- Sarah West
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Viviana Garza
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Rodolfo Cardoso
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| |
Collapse
|
4
|
Samir H, Elfadadny A, Radwan F, El-Sherbiny HR, Swelum AA, Khalil WA, Watanabe G. Spatial local expressions of kisspeptin in the uterus and uterine tubes and its relationship to the reproductive potential in goats. Domest Anim Endocrinol 2024; 88:106850. [PMID: 38640803 DOI: 10.1016/j.domaniend.2024.106850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
Kisspeptins are neuropeptides encoded by the Kiss1 gene that was discovered as a metastasis suppressor gene in melanoma and breast cancer. Kisspeptin has pivotal functions for gonadotropin-releasing hormone secretion and plays integrated roles in the hypothalamic-pituitary-gonadal axis. However, little is known about the peripheral expression of kisspeptin in ruminants, especially in the female reproductive tract. Here, the objectives of the current study were to investigate the spatial localization of kisspeptin and mRNA expression of Kiss1 and its receptor (Kiss1r) in the fallopian tubes (FT) and uterus of goats at varied reproductive activity (cyclic versus true anoestrous goats, n=6, each). Specimens of the uterus and FT were collected and fixed using paraformaldehyde to investigate the localizations of kisspeptin in the selected tissues by immunohistochemistry. Another set of samples was snape-frozen to identify the expressions of mRNAs encoding Kiss1 and Kiss1r using real-time PCR. Results revealed immunolocalizations of kisspeptin in the uterus and the FT. The staining of kisspeptin was found mainly in the mucosal epithelium of the uterus the FT, and the endometrial glands. Very intense staining of kisspeptin was found in the uterine and FT specimens in the true anoestrous goats compared to that in cyclic ones. The expression of mRNA encoding Kiss1 gene was significantly higher in the uterine specimen of cyclic goats (1.00±0.09) compared to that in the true anoestrous goats (0.62±0.08) (P ˂0.05), while the expression of mRNA encoding Kiss1r was significantly (P ˂0.001) higher in the uterine tissues of true anoestrous goats (1.78±0.17) compared to that in cyclic ones (1.00±0.11). In conclusion, immunohistochemical localization of kisspeptin and the expression of mRNA encoding Kiss1/Kiss1r revealed spatial changes in the uterus and FT of goats according to the reproductive potential of goats (cyclic versus true anoestrous goats). However, the definitive local role of kisspeptin in the uterus and FT need further investigation.
Collapse
Affiliation(s)
- Haney Samir
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt; Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo 183-8509, Japan.
| | - Ahmed Elfadadny
- Department of Animal Medicine, Faculty of Veterinary Medicine, Damanhour University, Damanhour, El-Beheira 22511, Egypt
| | - Faten Radwan
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo 183-8509, Japan; Veterinarian graduated from the Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt
| | - Hossam R El-Sherbiny
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Ayman A Swelum
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Wael A Khalil
- Department of Animal Production, Faculty of Agriculture, Mansoura University, Mansoura 35516, Egypt
| | - Gen Watanabe
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo 183-8509, Japan
| |
Collapse
|
5
|
Yamamoto S, Arakaki R, Noguchi H, Takeda A, Uchishiba M, Kamada S, Mineda A, Kon M, Kinouchi R, Yamamoto Y, Yoshida K, Kaji T, Shinohara N, Iwasa T. Kisspeptin administration may promote precopulatory behavior in male rats independently or supplementally to testosterone and contribute to proceptive behavior in female partners, reducing mating failure. Gen Comp Endocrinol 2024; 353:114528. [PMID: 38643848 DOI: 10.1016/j.ygcen.2024.114528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 02/22/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Kisspeptin is a peptide that plays an important role through its effects on the hypothalamus-pituitary-gonadal (HPG) axis. It has also been implicated in sexual behavior. The present study investigated whether the relationship between kisspeptin and sexual behavior is independent of the HPG axis, i.e., testosterone. Sexual behavior was examined after the administration of kisspeptin to gonadally intact male rats and gonadectomized male rats that received testosterone supplementation. Other male rats were also observed for sexual behavior once a week from 2 to 5 weeks after gonadectomy and receiving kisspeptin for the sixth postoperative week. Sexual behavior in female rats serving as the partner for each male was also observed. Female rats were not administered kisspeptin in the present study. The results obtained showed that the administration of kisspeptin increased precopulatory behavior in gonadally intact male rats and gonadectomized male rats that received testosterone supplementation and proceptive behavior in their female partners. Precopulatory behavior in males and receptive behavior in females increased, while copulatory behavior in males and receptive behavior in females remained unchanged. Furthermore, the administration of kisspeptin increased precopulatory behavior in gonadectomized males, but did not affect receptive behavior in females. These results suggest that kisspeptin affected males independently and/or supplementally to testosterone, and also that changes in the presence of testosterone in males had an impact on proceptive behavior in their female partners. In conclusion, kisspeptin may involve an as-yet-unidentified neural pathway in sexual desire independently of the HPG axis.
Collapse
Affiliation(s)
- Shota Yamamoto
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Graduate School, Tokushima University, Tokushima 770-8501, Japan; Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-0808, Japan
| | - Ryosuke Arakaki
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Graduate School, Tokushima University, Tokushima 770-8501, Japan
| | - Hiroki Noguchi
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Graduate School, Tokushima University, Tokushima 770-8501, Japan
| | - Asuka Takeda
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Graduate School, Tokushima University, Tokushima 770-8501, Japan
| | - Maimi Uchishiba
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Graduate School, Tokushima University, Tokushima 770-8501, Japan
| | - Shuhei Kamada
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Graduate School, Tokushima University, Tokushima 770-8501, Japan
| | - Ayuka Mineda
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Graduate School, Tokushima University, Tokushima 770-8501, Japan
| | - Masafumi Kon
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-0808, Japan
| | - Riyo Kinouchi
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Graduate School, Tokushima University, Tokushima 770-8501, Japan
| | - Yuri Yamamoto
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Graduate School, Tokushima University, Tokushima 770-8501, Japan
| | - Kanako Yoshida
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Graduate School, Tokushima University, Tokushima 770-8501, Japan
| | - Takashi Kaji
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Graduate School, Tokushima University, Tokushima 770-8501, Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-0808, Japan
| | - Takeshi Iwasa
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Graduate School, Tokushima University, Tokushima 770-8501, Japan.
| |
Collapse
|
6
|
Dardente H, Lomet D, Robert V, Lasserre O, Gonzalez AA, Mialhe X, Beltramo M. Photoperiod, but not progesterone, has a strong impact upon the transcriptome of the medio-basal hypothalamus in female goats and ewes. Mol Cell Endocrinol 2024; 588:112216. [PMID: 38556161 DOI: 10.1016/j.mce.2024.112216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
Photoperiod is the main environmental driver of seasonal responses in organisms living at temperate and polar latitudes. Other external cues such as food and temperature, and internal cues including hormones, intervene to fine-tune phasing of physiological functions to the solar year. In mammals, the medio-basal hypothalamus (MBH) is the key integrator of these cues, which orchestrates a wide array of seasonal functions, including breeding. Here, using RNAseq and RT-qPCR, we demonstrate that molecular components of the photoperiodic response previously identified in ewes are broadly conserved in does (female goats, Capra hircus), with a common core of ∼50 genes. This core group can be defined as the "MBH seasonal trancriptome", which includes key players of the pars tuberalis-tanycytes neuroendocrine retrograde pathway that governs intra-MBH photoperiodic switches of triiodothyronine (T3) production (Tshb, Eya3, Dio2 and SlcO1c1), the two histone methyltransferases Suv39H2 and Ezh2 and the secreted protein Vmo1. Prior data in ewes revealed that T3 and estradiol (E2), both key hormones for the proper timing of seasonal breeding, differentially impact the MBH seasonal transcriptome, and identified cellular and molecular targets through which these hormones might act. In contrast, information regarding the potential impact of progesterone (P4) upon the MBH transcriptome was nonexistent. Here, we demonstrate that P4 has no discernible transcriptional impact in either does or ewes. Taken together, our data show that does and ewes possess a common core set of photoperiod-responsive genes in the MBH and conclusively demonstrate that P4 is not a key regulator of the MBH transcriptome.
Collapse
Affiliation(s)
- Hugues Dardente
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France.
| | - Didier Lomet
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France
| | - Vincent Robert
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France
| | | | - Anne-Alicia Gonzalez
- MGX-Montpellier GenomiX, Univ. Montpellier, CNRS, INSERM, 34094, Montpellier, France
| | - Xavier Mialhe
- MGX-Montpellier GenomiX, Univ. Montpellier, CNRS, INSERM, 34094, Montpellier, France
| | | |
Collapse
|
7
|
Przybył BJ, Szlis M, Wysoczański B, Wójcik-Gładysz A. The role of QRFP43 in the secretory activity of the gonadotrophic axis in female sheep. Sci Rep 2024; 14:8989. [PMID: 38637687 PMCID: PMC11026372 DOI: 10.1038/s41598-024-59801-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024] Open
Abstract
In mammals reproduction is regulated by many factors, among others by the peptides belonging to the RFamide peptide family. However, the knowledge concerning on the impact of recently identified member of this family (QRFP43) on the modulation of the gonadotrophic axis activity is still not fully understood and current research results are ambiguous. In the present study we tested the in vivo effect of QRFP43 on the secretory activity of the gonadotrophic axis at the hypothalamic-pituitary level in Polish Merino sheep. The animals (n = 48) were randomly divided into three experimental groups: controls receiving an icv infusion of Ringer-Locke solution, group receiving icv infusion of QRFP43 at 10 μg per day and 50 μg per day. All sheep received four 50 min icv infusions at 30 min intervals, on each of three consecutive days. Hypothalamic and pituitaries were collected and secured for further immunohistochemical and molecular biological analysis. In addition, during the experiment a blood samples have been collected for subsequent RIA determinations. QRFP43 was found to downregulate Kiss mRNA expression in the MBH and reduce the level of IR material in ME. This resulted in a reduction of GnRH IR material in the ME. QRFP43 increased plasma FSH levels while decreasing LH levels. Our findings indicate that QRFP43 inhibits the activity of the gonadotropic axis in the ovine at the level of the hypothalamus and may represent another neuromodulator of reproductive processes in animals.
Collapse
Affiliation(s)
- Bartosz Jarosław Przybył
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110, Jabłonna, Poland
| | - Michał Szlis
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110, Jabłonna, Poland.
| | - Bartłomiej Wysoczański
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110, Jabłonna, Poland
| | - Anna Wójcik-Gładysz
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110, Jabłonna, Poland
| |
Collapse
|
8
|
Nakajo M, Kanda S, Oka Y. Involvement of the kisspeptin system in regulation of sexual behaviors in medaka. iScience 2024; 27:108971. [PMID: 38333699 PMCID: PMC10850746 DOI: 10.1016/j.isci.2024.108971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 11/09/2023] [Accepted: 01/16/2024] [Indexed: 02/10/2024] Open
Abstract
In mammals, kisspeptin (Kiss1) neurons are generally considered as a sex steroid-dependent key regulator of hypothalamic-pituitary-gonadal (HPG) axis. In contrast, previous studies in non-mammalian species, especially in teleosts, propose that Kiss1 is not directly involved in the HPG axis regulation, which suggests some sex-steroid-dependent functions of kisspeptin(s) other than the HPG axis regulation in non-mammals. Here, we used knockout (KO) medaka of kisspeptin receptor-coding genes (gpr54-1 and gpr54-2) and examined possible roles of kisspeptin in the regulation of sexual behaviors. We found that the KO pairs of gpr54-1, but not gpr54-2, spawned fewer eggs and exhibited delayed spawning than wild type pairs. Detailed behavior analysis suggested that the KO females are responsible for the delayed spawning and that the KO males showed hyper-motivation for courtship. Taken together, the present finding suggests that one of the reproductive-state-dependent functions of the Kiss1 may be the control of successful sexual behaviors.
Collapse
Affiliation(s)
- Mikoto Nakajo
- Department of Physiology, Division of Life Sciences, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Shinji Kanda
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Yoshitaka Oka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| |
Collapse
|
9
|
Aerts EG, Griesgraber MJ, Shuping SL, Bowdridge EC, Hardy SL, Goodman RL, Nestor CC, Hileman SM. The effect of NK3-Saporin injection within the arcuate nucleus on puberty, the LH surge, and the response to Senktide in female sheep†. Biol Reprod 2024; 110:275-287. [PMID: 37930247 DOI: 10.1093/biolre/ioad147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/09/2023] [Accepted: 10/24/2023] [Indexed: 11/07/2023] Open
Abstract
The timing of puberty onset is reliant on increased gonadotropin-releasing hormone (GnRH). This elicits a corresponding increase in luteinizing hormone (LH) due to a lessening of sensitivity to the inhibitory actions of estradiol (E2). The mechanisms underlying the increase in GnRH release likely involve a subset of neurons within the arcuate (ARC) nucleus of the hypothalamus that contain kisspeptin, neurokinin B (NKB), and dynorphin (KNDy neurons). We aimed to determine if KNDy neurons in female sheep are critical for: timely puberty onset; the LH surge; and the response to an intravenous injection of the neurokinin-3 receptor (NK3R) agonist, senktide. Prepubertal ewes received injections aimed at the ARC containing blank-saporin (control, n = 5) or NK3-saporin (NK3-SAP, n = 6) to ablate neurons expressing NK3R. Blood samples taken 3/week for 65 days following surgery were assessed for progesterone to determine onset of puberty. Control ewes exhibited onset of puberty at 33.2 ± 3.9 days post sampling initiation, whereas 5/6 NK3-SAP treated ewes didn't display an increase in progesterone. After an artificial LH surge protocol, surge amplitude was lower in NK3-SAP ewes. Finally, ewes were treated with senktide to determine if an LH response was elicited. LH pulses were evident in both groups in the absence of injections, but the response to senktide vs saline was similar between groups. These results show that KNDy cells are necessary for timely puberty onset and for full expresson of the LH surge. The occurrence of LH pulses in NK3-SAP treated ewes may indicate a recovery from an apulsatile state.
Collapse
Affiliation(s)
- Eliana G Aerts
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
| | - Max J Griesgraber
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
| | - Sydney L Shuping
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | | | - Steven L Hardy
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
| | - Robert L Goodman
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
- Department of Neuroscience, West Virginia University, Morgantown, WV 26506, USA
| | - Casey C Nestor
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Stanley M Hileman
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
- Department of Neuroscience, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
10
|
Tesema B, Liu GQ, Jiang XP. Active kisspeptin DNA vaccines oral immunization disrupt mRNA hormone receptors expression in ram lambs. Anim Biotechnol 2023; 34:2285-2294. [PMID: 35714982 DOI: 10.1080/10495398.2022.2087665] [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] [Indexed: 11/01/2022]
Abstract
To evaluate the efficacy of oral immunization with active kisspeptin DNA vaccine on the expression of hormone receptor mRNA. For this study, ten 56-day-old Hu breed ram lambs were randomly assigned to the treatment and control groups (n = 5). Treatment Experimental group received C500/pKS-asd and the control group received C500/pVAX-asd (aspartate-β semialdehyde dehydrogenase orally on days 0, 28, and 56, and blood samples were taken at each immunization interval (14-day) and tissues samples were collected at the end of the experimental period (day 98). The collected samples were stored in the refrigerator at -20 °C and liquid nitrogen, respectively, for laboratory examination. Total RNA was extracted from samples using TRIzol reagent and quantitative real-time polymerase chain reaction (QPCR) was used to quantify the levels of KISS1, G protein-coupled receptor-54 (Kiss1r), and gonadotrophin-releasing hormone (GnRH) mRNA in the hypothalamus. Levels of luteinizing hormone receptor (LHR) and luteinizing hormone beta (LHβ) mRNA, and follicle-stimulating hormone receptor (FSHR) and follicle-stimulating hormone beta (FSHβ) mRNA in the testes and pituitary were analyzed, respectively. Further, gonadotropin-releasing hormone receptor (GnRHR) mRNA expression level in the pituitary was measured. Moreover, the Kiss1r concentration level in the blood was measured using an indirect ELISA. The concentration of Kiss1r in the blood was lower in the treatment group than in the control group (p < 0.05). The levels of testicular FSHR and LHR mRNA were significantly lower in the treatment group (p < 0.05) when compared to the control group. Furthermore, the treatment group's levels of hypothalamic KISS1, Kiss1r, and GnRH mRNA were significantly lower (p < 0.05) than the controls. LH, FSH, and GnRHR mRNA expression in the pituitary were also significantly lower in the treatment group (p < 0.01 and p < 0.05, respectively). These findings imply that oral immunization with active kisspeptin DNA vaccine suppresses hormone receptor mRNA expression in the ram lambs.
Collapse
Affiliation(s)
- Birhanu Tesema
- Key Laboratory of Agricultural - Animal Genetics, Breeding, and Reproduction, Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
- Department of Animal Science, College of Agricultural Sciences, Bule Hora University, Bule Hora, Ethiopia
| | - Gui-Qiang Liu
- Key Laboratory of Agricultural - Animal Genetics, Breeding, and Reproduction, Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Xun-Ping Jiang
- Key Laboratory of Agricultural - Animal Genetics, Breeding, and Reproduction, Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| |
Collapse
|
11
|
Constantin S, Sokanovic SJ, Mochimaru Y, Smiljanic K, Sivcev S, Prévide RM, Wray S, Balla T, Stojilkovic SS. Postnatal Development and Maintenance of Functional Pituitary Gonadotrophs Is Dependent on PI4-Kinase A. Endocrinology 2023; 164:bqad168. [PMID: 37935042 PMCID: PMC10652335 DOI: 10.1210/endocr/bqad168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023]
Abstract
Postnatal development of functional pituitary gonadotrophs is necessary for maturation of the hypothalamic-pituitary-gonadal axis, puberty, and reproduction. Here we examined the role of PI4-kinase A, which catalyzes the biosynthesis of PI4P in mouse reproduction by knocking out this enzyme in cells expressing the gonadotropin-releasing hormone (GnRH) receptor. Knockout (KO) mice were infertile, reflecting underdeveloped gonads and reproductive tracts and lack of puberty. The number and distribution of hypothalamic GnRH neurons and Gnrh1 expression in postnatal KOs were not affected, whereas Kiss1/kisspeptin expression was increased. KO of PI4-kinase A also did not alter embryonic establishment and neonatal development and function of the gonadotroph population. However, during the postnatal period, there was a progressive loss of expression of gonadotroph-specific genes, including Fshb, Lhb, and Gnrhr, accompanied by low gonadotropin synthesis. The postnatal gonadotroph population also progressively declined, reaching approximately one-third of that observed in controls at 3 months of age. In these residual gonadotrophs, GnRH-dependent calcium signaling and calcium-dependent membrane potential changes were lost, but intracellular administration of inositol-14,5-trisphosphate rescued this signaling. These results indicate a key role for PI4-kinase A in the postnatal development and maintenance of a functional gonadotroph population.
Collapse
Affiliation(s)
- Stephanie Constantin
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Srdjan J Sokanovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yuta Mochimaru
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kosara Smiljanic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sonja Sivcev
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rafael M Prévide
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Susan Wray
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tamas Balla
- Section on Molecular Signal Transduction, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stanko S Stojilkovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
12
|
Zhang Y, Yang Y, Tao Y, Guo X, Cui Y, Li Z. Phthalates (PAEs) and reproductive toxicity: Hypothalamic-pituitary-gonadal (HPG) axis aspects. JOURNAL OF HAZARDOUS MATERIALS 2023; 459:132182. [PMID: 37557049 DOI: 10.1016/j.jhazmat.2023.132182] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023]
Abstract
Phthalates (PAEs) are widely used for their excellent ability to improve plastic products. As an essential endocrine axis that regulates the reproductive system, whether dysfunction of the hypothalamic-pituitary-gonadal (HPG) axis is involved in reproductive toxicity mediated by environmental endocrine disruptors PAEs has become a hot topic of widespread concern. This study systematically reviewed the adverse effects of multiple PAEs on the HPG axis in different models and objectively discussed the possible underlying mechanisms. The abnormal release of gonadotropin-releasing hormone and gonadotropin, dysfunction of sex hormone receptors and steroid hormone synthesis, and general damage, including cell proliferation, oxidative stress, apoptosis, and autophagy have been confirmed to be involved in this process. Although it is widely established that PAEs induce HPG axis dysfunction, the specific mechanisms involved remain unclear. From a systematic review of relevant publications, it appears that the abnormal expression of peroxisome proliferator-activated, aryl hydrocarbon, and insulin receptors mediated by PAEs is key upstream event that induces these adverse outcomes; however, this inference needs to be further verified. Overall, this study aimed to provide reliable potential biomarkers for future environmental risk assessment and epidemiological investigation of PAEs.
Collapse
Affiliation(s)
- Ying Zhang
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China.
| | - Yang Yang
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| | - Yue Tao
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiangyong Guo
- Fuyu County Agricultural Technology Extension Center, Qiqihar 161200, PR China
| | - Yunhe Cui
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| | - Zixu Li
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| |
Collapse
|
13
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 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
|
14
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 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
|
15
|
Carrasco RA, Breen KM. Allostasis in Neuroendocrine Systems Controlling Reproduction. Endocrinology 2023; 164:bqad125. [PMID: 37586095 PMCID: PMC10461221 DOI: 10.1210/endocr/bqad125] [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: 05/30/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023]
Abstract
Allostasis provides a supporting role to the homeostatic control of biological variables in mammalian species. While the concept of homeostasis is related to the control of variables within a set point or range that are essential to life, allostasis refers to systems that facilitate adaptation to challenges that the organism faces and the new requirements for survival. Essential for such adaptation is the role played by the brain in eliciting neural and neuroendocrine responses. Reproductive function is fundamental for the survival of species but is costly in energetic terms and requires a synchrony with an ever-changing environment. Thus, in many species reproductive function is blocked or delayed over immediate challenges. This review will cover the physiological systems and neuroendocrine pathways that supply allostatic control over reproductive neuroendocrine systems. Light, hypoxia, temperature, nutrition, psychosocial, and immune mediators influence the neuroendocrine control of reproductive functions through pathways that are confluent at the paraventricular nucleus; however, understanding of the integrative responses to these stimuli has not been clarified. Likely, the ultimate consequence of these allostatic mechanisms is the modification of kisspeptin and gonadotropin-releasing hormone neuronal activity, thus compromising reproduction function in the short term, while preserving species survivability.
Collapse
Affiliation(s)
- Rodrigo A Carrasco
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093-0674, USA
| | - Kellie M Breen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093-0674, USA
| |
Collapse
|
16
|
Zimerman J, Niño OMS, da Costa CS, Zanol JF, Comério M, da Gama de Souza LN, Miranda-Alves L, Miranda RA, Lisboa PC, Camilo TA, Rorato R, Alves GA, Frazão R, Zomer HD, Freitas-Lima LC, Graceli JB. Subacute high-refined carbohydrate diet leads to abnormal reproductive control of the hypothalamic-pituitary axis in female rats. Reprod Toxicol 2023; 119:108410. [PMID: 37211340 DOI: 10.1016/j.reprotox.2023.108410] [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: 03/12/2023] [Revised: 05/05/2023] [Accepted: 05/18/2023] [Indexed: 05/23/2023]
Abstract
We previously reported that female rats placed on a diet containing refined carbohydrates (HCD) resulted in obesity and reproductive abnormalities, such as high serum LH concentration and abnormal ovarian function. However, the impacts at the hypothalamic-pituitary (HP) function, specifically regarding pathways linked to reproductive axis modulation are unknown. In this study, we assessed whether subacute feeding with HCD results in abnormal reproductive control in the HP axis. Female rats were fed with HCD for 15 days and reproductive HP axis morphophysiology was assessed. HCD reduced hypothalamic mRNA expression (Kiss1, Lepr, and Amhr2) and increased pituitary LHβ+ cells. These changes likely contribute to the increase in serum LH concentration observed in HCD. Blunted estrogen negative feedback was observed in HCD, with increased kisspeptin protein expression in the arcuate nucleus of the hypothalamus (ARH), lower LHβ+ cells and LH concentration in ovariectomized (OVX)+HCD rats. Thus, these data suggest that HCD feeding led to female abnormal reproductive control of HP axis.
Collapse
Affiliation(s)
- Jeanini Zimerman
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | - Oscar M S Niño
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil; Faculty of Human Sciences and Education, Universidad de los Llanos, Villavicencio, Meta, Colombia
| | - Charles S da Costa
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | - Jordana F Zanol
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | - Milena Comério
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | | | - Leandro Miranda-Alves
- Experimental Endocrinology Research, Development and Innovation Group, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil; Postgraduate Program in Endocrinology, School of Medicine, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, Ilha do Governador, Cidade Universitária, UFRJ, RJ, Brazil
| | - Rosiane A Miranda
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - Patrícia C Lisboa
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - Tays A Camilo
- Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Rodrigo Rorato
- Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Guilherme Andrade Alves
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Renata Frazão
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Helena D Zomer
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | | | - Jones B Graceli
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil.
| |
Collapse
|
17
|
Kumar SS, Bouwer GT, Jackson MK, Perkinson MR, McDonald FJ, Brown CH, Augustine RA. Kisspeptin neuron projections to oxytocin neurons are not necessary for parturition in the mouse. Brain Struct Funct 2023; 228:1535-1548. [PMID: 37389617 PMCID: PMC10335956 DOI: 10.1007/s00429-023-02670-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/13/2023] [Indexed: 07/01/2023]
Abstract
Oxytocin is synthesized by hypothalamic supraoptic nucleus (SON) and paraventricular nucleus (PVN) neurons and is released from the posterior pituitary gland to trigger uterine contractions during parturition. In rats, oxytocin neuron innervation by periventricular nucleus (PeN) kisspeptin neurons increases over pregnancy and intra-SON kisspeptin administration excites oxytocin neurons only in late pregnancy. To test the hypothesis that kisspeptin neurons excite oxytocin neurons to trigger uterine contractions during birth in C57/B6J mice, double-label immunohistochemistry for kisspeptin and oxytocin first confirmed that kisspeptin neurons project to the SON and PVN. Furthermore, kisspeptin fibers expressed synaptophysin and formed close appositions with oxytocin neurons in the mouse SON and PVN before and during pregnancy. Stereotaxic viral delivery of caspase-3 into the AVPV/PeN of Kiss-Cre mice before mating reduced kisspeptin expression in the AVPV, PeN, SON and PVN by > 90% but did not affect the duration of pregnancy or the timing of delivery of each pup during parturition. Therefore, it appears that AVPV/PeN kisspeptin neuron projections to oxytocin neurons are not necessary for parturition in the mouse.
Collapse
Affiliation(s)
- Shalini S Kumar
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Physiology, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Gregory T Bouwer
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Physiology, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Meliame K Jackson
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Physiology, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Michael R Perkinson
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Physiology, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Fiona J McDonald
- Department of Physiology, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Colin H Brown
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Physiology, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Rachael A Augustine
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand.
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand.
- Department of Physiology, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand.
| |
Collapse
|
18
|
Huang H, Xiao K, Shu T, Liu X, Yang J. Effects of Kisspeptin on the reproductive function in the Dabry's sturgeon (Acipenser dabrynus). Gen Comp Endocrinol 2023; 336:114244. [PMID: 36841441 DOI: 10.1016/j.ygcen.2023.114244] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/26/2022] [Accepted: 02/18/2023] [Indexed: 02/27/2023]
Abstract
Kisspeptin, a kind of neuropeptide, is involved in various physiological processes such as tumor metastasis inhibition and reproductive regulation due to its ability to interact with Kisspeptin receptor-Kissr. In teleost, Kisspeptin/Kissr system stimulates the hypothalamus-pituitary-gonadal axis (HPG axis), which is crucial for the reproductive regulation. Compared to one Kisspeptin protein Kiss1 was existed in mammals, two Kisspeptin were identified in sturgeon species, including Kiss1 and Kiss2, with specific receptors of Kissr1 and Kissr2, respectively. However, few reports described the effects of the two isoforms of Kisspeptin on the reproductive regulation in sturgeon. The core peptides of Kiss1 and Kiss2 (Kiss1-10 and Kiss2-10) of Dabry's sturgeon were successfully synthesized to explore the functional influence of Kisspeptin on the sturgeon HPG axis in the present study. The present findings suggested that intraperitoneal injection of Kiss1-10 and Kiss2-10 could significantly up-regulate the mRNA expression of Gnrh、Fsh and Lh in the hypothalamus and pituitary and the content of Lh protein in the serum. Assays of Kisspeptin-treated cells demonstrated that Kiss1-10 and Kiss2-10 can significantly promote the expression of Gnrh in hypothalamus cells and Lh and Fsh in pituitary cells of Dabry's sturgeon, indicating their direct-acting effect on pituitary cells and regulatory function on the reproductive development of sturgeon. This study described the reproductive function of the Kisspeptin in the Dabry's sturgeon for the first time, and provided supportive reference for the development of high-efficiency ripening technologies of artificially breeding sturgeon.
Collapse
Affiliation(s)
- Hongtao Huang
- Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, Hubei 443100, China; Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, Hubei 443100, China.
| | - Kan Xiao
- Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, Hubei 443100, China; Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, Hubei 443100, China.
| | - Tingting Shu
- Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, Hubei 443100, China; Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, Hubei 443100, China.
| | - Xueqing Liu
- Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, Hubei 443100, China; Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, Hubei 443100, China.
| | - Jing Yang
- Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, Hubei 443100, China; Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, Hubei 443100, China.
| |
Collapse
|
19
|
Garza V, West SM, Cardoso RC. Review: Gestational and postnatal nutritional effects on the neuroendocrine control of puberty and subsequent reproductive performance in heifers. Animal 2023; 17 Suppl 1:100782. [PMID: 37567667 DOI: 10.1016/j.animal.2023.100782] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 08/13/2023] Open
Abstract
Pubertal attainment is an intricate biological process that involves maturation of the reproductive neuroendocrine axis and increased pulsatile release of gonadotropin-releasing hormone (GnRH) and luteinizing hormone. Nutrition is a critical environmental factor controlling the timing of puberty attainment. Nutrient restriction during early postnatal development delays puberty, whereas increased feed intake and adiposity during this period hasten pubertal maturation by imprinting the hypothalamus. Moreover, the dam's nutrition during gestation can program the neuroendocrine system in the developing fetus and has the potential to advance or delay puberty in the offspring. Leptin, a hormone produced primarily by adipose cells, plays an important role in communicating energy status to the brain and regulating sexual maturation. Leptin's regulation of GnRH release is mediated by an upstream neuronal network since GnRH neurons do not contain the leptin receptor. Two groups of neurons located in the arcuate nucleus of the hypothalamus that express neuropeptide Y (NPY), an orexigenic peptide, and alpha melanocyte-stimulating hormone (αMSH), an anorexigenic peptide, are central elements of the neural circuitry that relay inhibitory (NPY) and excitatory (αMSH) inputs to GnRH neurons. Moreover, KNDy neurons, neurons in the arcuate nucleus that co-express kisspeptin, neurokinin B (NKB), and dynorphin, also play a role in the metabolic regulation of puberty. Our studies in beef heifers demonstrate that increased rates of BW gain during early postweaning (4-9 mo of age) result in reduced expression of NPY mRNA, increased expression of proopiomelanocortin and kisspeptin receptor mRNA, reduced NPY inhibitory inputs to GnRH neurons, and increased excitatory αMSH inputs to KNDy neurons. Finally, our most recent data demonstrate that nutrition of the cow during the last two trimesters of gestation can also induce transcriptional and structural changes in hypothalamic neurocircuitries in the heifer progeny that likely persist long-term after birth. Managerial approaches, such as supplementation of the dam during gestation (fetal programming), creep feeding, early weaning, and stair-step nutritional regimens have been developed to exploit brain plasticity and advance pubertal maturation in heifers.
Collapse
Affiliation(s)
- Viviana Garza
- Department of Animal Science, Texas A&M University, 2471 TAMU, College Station, TX 77843, USA
| | - Sarah M West
- Department of Animal Science, Texas A&M University, 2471 TAMU, College Station, TX 77843, USA
| | - Rodolfo C Cardoso
- Department of Animal Science, Texas A&M University, 2471 TAMU, College Station, TX 77843, USA.
| |
Collapse
|
20
|
Velasco I, Franssen D, Daza-Dueñas S, Skrapits K, Takács S, Torres E, Rodríguez-Vazquez E, Ruiz-Cruz M, León S, Kukoricza K, Zhang FP, Ruohonen S, Luque-Cordoba D, Priego-Capote F, Gaytan F, Ruiz-Pino F, Hrabovszky E, Poutanen M, Vázquez MJ, Tena-Sempere M. Dissecting the KNDy hypothesis: KNDy neuron-derived kisspeptins are dispensable for puberty but essential for preserved female fertility and gonadotropin pulsatility. Metabolism 2023; 144:155556. [PMID: 37121307 DOI: 10.1016/j.metabol.2023.155556] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/31/2023] [Accepted: 04/02/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND Kiss1 neurons in the hypothalamic arcuate-nucleus (ARC) play key roles in the control of GnRH pulsatility and fertility. A fraction of ARC Kiss1 neurons, termed KNDy, co-express neurokinin B (NKB; encoded by Tac2). Yet, NKB- and Kiss1-only neurons are also found in the ARC, while a second major Kiss1-neuronal population is present in the rostral hypothalamus. The specific contribution of different Kiss1 neuron sub-sets and kisspeptins originating from them to the control of reproduction and eventually other bodily functions remains to be fully determined. METHODS To tease apart the physiological roles of KNDy-born kisspeptins, conditional ablation of Kiss1 in Tac2-expressing cells was implemented in vivo. To this end, mice with Tac2 cell-specific Kiss1 KO (TaKKO) were generated and subjected to extensive reproductive and metabolic characterization. RESULTS TaKKO mice displayed reduced ARC kisspeptin content and Kiss1 expression, with greater suppression in females, which was detectable at infantile-pubertal age. In contrast, Tac2/NKB levels were fully preserved. Despite the drop of ARC Kiss1/kisspeptin, pubertal timing was normal in TaKKO mice of both sexes. However, young-adult TaKKO females displayed disturbed LH pulsatility and sex steroid levels, with suppressed basal LH and pre-ovulatory LH surges, early-onset subfertility and premature ovarian insufficiency. Conversely, testicular histology and fertility were grossly conserved in TaKKO males. Ablation of Kiss1 in Tac2-cells led also to sex-dependent alterations in body composition, glucose homeostasis, especially in males, and locomotor activity, specifically in females. CONCLUSIONS Our data document that KNDy-born kisspeptins are dispensable/compensable for puberty in both sexes, but required for maintenance of female gonadotropin pulsatility and fertility, as well as for adult metabolic homeostasis. SIGNIFICANCE STATEMENT Neurons in the hypothalamic arcuate nucleus (ARC) co-expressing kisspeptins and NKB, named KNDy, have been recently suggested to play a key role in pulsatile secretion of gonadotropins, and hence reproduction. However, the relative contribution of this Kiss1 neuronal-subset, vs. ARC Kiss1-only and NKB-only neurons, as well as other Kiss1 neuronal populations, has not been assessed in physiological settings. We report here findings in a novel mouse-model with elimination of KNDy-born kisspeptins, without altering other kisspeptin compartments. Our data highlights the heterogeneity of ARC Kiss1 populations and document that, while dispensable/compensable for puberty, KNDy-born kisspeptins are required for proper gonadotropin pulsatility and fertility, specifically in females, and adult metabolic homeostasis. Characterization of this functional diversity is especially relevant, considering the potential of kisspeptin-based therapies for management of human reproductive disorders.
Collapse
Affiliation(s)
- Inmaculada Velasco
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain; Hospital Universitario Reina Sofía, Cordoba, Spain
| | - Delphine Franssen
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain; GIGA-Neurosciences Unit, University of Liège, Liège, Belgium
| | - Silvia Daza-Dueñas
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain; Hospital Universitario Reina Sofía, Cordoba, Spain
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Szabolcs Takács
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Encarnación Torres
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain; Hospital Universitario Reina Sofía, Cordoba, Spain
| | - Elvira Rodríguez-Vazquez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain; Hospital Universitario Reina Sofía, Cordoba, Spain
| | - Miguel Ruiz-Cruz
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain; Hospital Universitario Reina Sofía, Cordoba, Spain
| | - Silvia León
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain; Hospital Universitario Reina Sofía, Cordoba, Spain
| | - Krisztina Kukoricza
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Finland
| | - Fu-Ping Zhang
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Finland
| | - Suvi Ruohonen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Finland
| | - Diego Luque-Cordoba
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain; Department of Analytical Chemistry, University of Córdoba, Spain; CIBER Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III, Spain
| | - Feliciano Priego-Capote
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain; Department of Analytical Chemistry, University of Córdoba, Spain; CIBER Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III, Spain
| | - Francisco Gaytan
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain; Hospital Universitario Reina Sofía, Cordoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| | - Francisco Ruiz-Pino
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Matti Poutanen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Finland
| | - María J Vázquez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain; Hospital Universitario Reina Sofía, Cordoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain; Hospital Universitario Reina Sofía, Cordoba, Spain; Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Finland; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain.
| |
Collapse
|
21
|
Meunier MA, Porte C, Poissenot K, Vacher H, Brachet M, Chamero P, Beltramo M, Abecia JA, Delgadillo JA, Chemineau P, Keller M. Male-induced early puberty correlates with the maturation of arcuate nucleus kisspeptin neurons in does. J Neuroendocrinol 2023; 35:e13284. [PMID: 37157154 DOI: 10.1111/jne.13284] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/09/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023]
Abstract
In goats, early exposure of spring-born females to sexually active bucks induces an early puberty onset assessed by the first ovulation. This effect is found when females are continuously exposed well before the male breeding season starting in September. The first aim of this study was to evaluate whether a shortened exposure of females to males could also lead to early puberty. We assessed the onset of puberty in Alpine does isolated from bucks (ISOL), exposed to wethers (CAS), exposed to intact bucks from the end of June (INT1), or mid-August (INT2). Intact bucks became sexually active in mid-September. At the beginning of October, 100% of INT1 and 90% of INT2 exposed does ovulated, in contrast to the ISOL (0%) and CAS (20%) groups. This demonstrated that contact with males that become sexually active is the main factor prompting precocious puberty in females. Furthermore, a reduced male exposure during a short window before the breeding season is sufficient to induce this phenomenon. The second aim was to investigate the neuroendocrine changes induced by male exposure. We found a significant increase in kisspeptin immunoreactivity (fiber density and number of cell bodies) in the caudal part of the arcuate nucleus of INT1 and INT2 exposed females. Thus, our results suggest that sensory stimuli from sexually active bucks (e.g., chemosignals) may trigger an early maturation of the ARC kisspeptin neuronal network leading to gonadotropin-releasing hormone secretion and first ovulation.
Collapse
Affiliation(s)
- Maxime A Meunier
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Chantal Porte
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Kévin Poissenot
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Hélène Vacher
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Morgane Brachet
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Pablo Chamero
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Massimiliano Beltramo
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - José A Abecia
- Departamento de Producción Animal y Ciencia de los Alimentos, IUCA, Universidad de Zaragoza, Zaragoza, Spain
| | - José A Delgadillo
- Centro de Investigación en Reproducción Caprina, Universidad Autónoma Agraria Antonio Narro, Torreón, Mexico
| | - Philippe Chemineau
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Matthieu Keller
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| |
Collapse
|
22
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [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
|
23
|
Prashar V, Arora T, Singh R, Sharma A, Parkash J. Hypothalamic Kisspeptin Neurons: Integral Elements of the GnRH System. Reprod Sci 2023; 30:802-822. [PMID: 35799018 DOI: 10.1007/s43032-022-01027-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/23/2022] [Indexed: 12/19/2022]
Abstract
Highly sophisticated and synchronized interactions of various cells and hormonal signals are required to make organisms competent for reproduction. GnRH neurons act as a common pathway for multiple cues for the onset of puberty and attaining reproductive function. GnRH is not directly receptive to most of the signals required for the GnRH secretion during the various phases of the ovarian cycle. Kisspeptin neurons of the hypothalamus convey these signals required for the synchronized release of the GnRH. The steroid-sensitive anteroventral periventricular nucleus (AVPV) kisspeptin and arcuate nucleus (ARC) KNDy neurons convey steroid feedback during the reproductive cycle necessary for GnRH surge and pulse, respectively. AVPV region kisspeptin neurons also communicate with nNOS synthesizing neurons and suprachiasmatic nucleus (SCN) neurons to coordinate the process of the ovarian cycle. Neurokinin B (NKB) and dynorphin play roles in the GnRH pulse stimulation and inhibition, respectively. The loss of NKB and kisspeptin function results in the development of neuroendocrine disorders such as hypogonadotropic hypogonadism (HH) and infertility. Ca2+ signaling is essential for GnRH pulse generation, which is propagated through gap junctions between astrocytes-KNDy and KNDy-KNDy neurons. Impaired functioning of KNDy neurons could develop the characteristics associated with polycystic ovarian syndrome (PCOS) in rodents. Kisspeptin-increased synthesis led to excessive secretion of the LH associated with PCOS. This review provides the latest insights and understanding into the role of the KNDy and AVPV/POA kisspeptin neurons in GnRH secretion and PCOS.
Collapse
Affiliation(s)
- Vikash Prashar
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Tania Arora
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Randeep Singh
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Arti Sharma
- Department of Computational Sciences, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Jyoti Parkash
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India.
| |
Collapse
|
24
|
Power EM, Ganeshan D, Iremonger KJ. Estradiol regulates voltage-gated potassium currents in corticotropin-releasing hormone neurons. J Exp Biol 2023; 226:287072. [PMID: 36805713 PMCID: PMC10038157 DOI: 10.1242/jeb.245222] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/08/2023] [Indexed: 02/22/2023]
Abstract
Corticotropin-releasing hormone (CRH) neurons are the primary neural population controlling the hypothalamic-pituitary-adrenal (HPA) axis and the secretion of adrenal stress hormones. Previous work has demonstrated that stress hormone secretion can be regulated by circulating levels of estradiol. However, the effect of estradiol on CRH neuron excitability is less clear. Here, we show that chronic estradiol replacement following ovariectomy increases two types of potassium channel currents in CRH neurons: fast inactivating voltage-gated A-type K+ channel currents (IA) and non-inactivating M-type K+ channel currents (IM). Despite the increase in K+ currents following estradiol replacement, there was no overall change in CRH neuron spiking excitability assessed with either frequency-current curves or current ramps. Together, these data reveal a complex picture whereby ovariectomy and estradiol replacement differentially modulate distinct aspects of CRH neuron and HPA axis function.
Collapse
Affiliation(s)
- Emmet M Power
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin9016, New Zealand
| | - Dharshini Ganeshan
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin9016, New Zealand
| | - Karl J Iremonger
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin9016, New Zealand
| |
Collapse
|
25
|
Hellier V, Dardente H, Lomet D, Cognié J, Dufourny L. Interactions between β-endorphin and kisspeptin neurons of the ewe arcuate nucleus are modulated by photoperiod. J Neuroendocrinol 2023; 35:e13242. [PMID: 36880357 DOI: 10.1111/jne.13242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023]
Abstract
Opioid peptides are well-known modulators of the central control of reproduction. Among them, dynorphin coexpressed in kisspeptin (KP) neurons of the arcuate nucleus (ARC) has been thoroughly studied for its autocrine effect on KP release through κ opioid receptors. Other studies have suggested a role for β-endorphin (BEND), a peptide cleaved from the pro-opiomelanocortin precursor, on food intake and central control of reproduction. Similar to KP, BEND content in the ARC of sheep is modulated by day length and BEND modulates food intake in a dose-dependent manner. Because KP levels in the ARC vary with photoperiodic and metabolic status, a photoperiod-driven influence of BEND neurons on neighboring KP neurons is plausible. The present study aimed to investigate a possible modulatory action of BEND on KP neurons located in the ovine ARC. Using confocal microscopy, numerous KP appositions on BEND neurons were found but there was no photoperiodic variation of the number of these interactions in ovariectomized, estradiol-replaced ewes. By contrast, BEND terminals on KP neurons were twice as numerous under short days, in ewes having an activated gonadotropic axis, compared to anestrus ewes under long days. Injection of 5 μg BEND into the third ventricle of short-day ewes induced a significant and specific increase of activated KP neurons (16% vs. 9% in controls), whereas the percentage of overall activated (c-Fos positive) neurons, was similar between both groups. These data suggest a photoperiod-dependent influence of BEND on KP neurons of the ARC, which may influence gonadotropin-releasing hormone pulsatile secretion and inform KP neurons about metabolic status.
Collapse
Affiliation(s)
- Vincent Hellier
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Hugues Dardente
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Didier Lomet
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Juliette Cognié
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | | |
Collapse
|
26
|
De Jesus AN, Henry BA. The role of oestrogen in determining sexual dimorphism in energy balance. J Physiol 2023; 601:435-449. [PMID: 36117117 PMCID: PMC10092637 DOI: 10.1113/jp279501] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/26/2022] [Indexed: 02/03/2023] Open
Abstract
Energy balance is determined by caloric intake and the rate at which energy is expended, with the latter comprising resting energy expenditure, physical activity and adaptive thermogenesis. The regulation of both energy intake and expenditure exhibits clear sexual dimorphism, with young women being relatively protected against weight gain and the development of cardiometabolic diseases. Preclinical studies have indicated that females are more sensitive to the satiety effects of leptin and insulin compared to males. Furthermore, females have greater thermogenic activity than males, whereas resting energy expenditure is generally higher in males than females. In addition to this, in post-menopausal women, the decline in sex steroid concentration, particularly in oestrogen, is associated with a shift in the distribution of adipose tissue and overall increased propensity to gain weight. Oestrogens are known to regulate energy balance and weight homeostasis via effects on both food intake and energy expenditure. Indeed, 17β-oestradiol treatment increases melanocortin signalling in the hypothalamus to cause satiety. Furthermore, oestrogenic action at the ventromedial hypothalamus has been linked with increased energy expenditure in female mice. We propose that oestrogen action on energy balance is multi-faceted and is fundamental to determining sexual dimorphism in weight control. Furthermore, evidence suggests that the decline in oestrogen levels leads to increased risk of weight gain and development of cardiometabolic disease in women across the menopausal transition.
Collapse
Affiliation(s)
- Anne Nicole De Jesus
- Metabolism, Obesity and Diabetes Program, Biomedicine, Discovery Institute, Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Belinda A Henry
- Metabolism, Obesity and Diabetes Program, Biomedicine, Discovery Institute, Department of Physiology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
27
|
Gurule S, Sustaita-Monroe J, Padmanabhan V, Cardoso R. Developmental programming of the neuroendocrine axis by steroid hormones: Insights from the sheep model of PCOS. Front Endocrinol (Lausanne) 2023; 14:1096187. [PMID: 36755919 PMCID: PMC9899912 DOI: 10.3389/fendo.2023.1096187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/06/2023] [Indexed: 01/25/2023] Open
Abstract
The reproductive neuroendocrine system is a key target for the developmental programming effects of steroid hormones during early life. While gonadal steroids play an important role in controlling the physiological development of the neuroendocrine axis, human fetuses are susceptible to adverse programming due to exposure to endocrine disrupting chemicals with steroidal activity, inadvertent use of contraceptive pills during pregnancy, as well as from disease states that result in abnormal steroid production. Animal models provide an unparalleled resource to understand the effects of steroid hormones on the development of the neuroendocrine axis and their role on the developmental origins of health and disease. In female sheep, exposure to testosterone (T) excess during fetal development results in an array of reproductive disorders that recapitulate those seen in women with polycystic ovary syndrome (PCOS), including disrupted neuroendocrine feedback mechanisms, increased pituitary responsiveness to gonadotropin-releasing hormone (GnRH), luteinizing hormone (LH) hypersecretion, functional hyperandrogenism, multifollicular ovarian morphology, and premature reproductive failure. Similar to a large proportion of women with PCOS, these prenatally T-treated sheep also manifest insulin resistance and cardiovascular alterations, including hypertension. This review article focuses on the effects of prenatal androgens on the developmental programming of hypothalamic and pituitary alterations in the sheep model of PCOS phenotype, centering specifically on key neurons, neuropeptides, and regulatory pathways controlling GnRH and LH secretion. Insights obtained from the sheep model as well as other animal models of perinatal androgen excess can have important translational relevance to treat and prevent neuroendocrine dysfunction in women with PCOS and other fertility disorders.
Collapse
Affiliation(s)
- Sara Gurule
- Department of Animal Science, Texas A&M University, College Station, TX, United States
| | | | | | - Rodolfo Cardoso
- Department of Animal Science, Texas A&M University, College Station, TX, United States
| |
Collapse
|
28
|
Xu G, Zhao X, Li Z, Hu J, Li X, Li J, Chen Y. Effects of electroacupuncture on the kisspeptin-gonadotropin-releasing hormone (GnRH) /luteinizing hormone (LH) neural circuit abnormalities and androgen receptor expression of kisspeptin/neurokinin B/dynorphin neurons in PCOS rats. J Ovarian Res 2023; 16:15. [PMID: 36650561 PMCID: PMC9844031 DOI: 10.1186/s13048-022-01078-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 12/13/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is characterized by hyperandrogenism, anovulation, and polycystic ovaries. Electroacupuncture (EA) can effectively improve hyperandrogenism and increase ovulation frequency in patients with PCOS. Pieces of suggest that androgen activity in the brain is associated with impaired steroid negative feedback in such patients. Studies have shown that EA regulated androgen receptor (AR) expression and local factor levels (such as anti-Müllerian hormone and inhibin B) in the ovary of PCOS rats. However, few studies have explored the effect of EA on androgen activity in the brain. OBJECTIVE This study investigated the effect of EA on the kisspeptin-gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) neural circuit and sex hormone receptor expression in the hypothalamus of PCOS rats. METHODS PCOS signs were induced by letrozole administration, and the induced rats were treated with low-frequency EA at Guan Yuan acupoint (CV4). The effect of EA on PCOS-like signs was evaluated by observing changes in the body weight, ovarian quality, ovarian morphology, and serum sex hormone levels in rats. To explore the mechanism of the effect of EA on PCOS-like signs, the neuropeptide content of the kisspeptin-GnRH/LH neural circuit was assessed using enzyme-linked immunosorbent assay(ELISA); AR and estrogen receptor α (ERα) coexpression on kisspeptin/neurokinin B/dynorphin (KNDy) neurons was determined via triple-label immunofluorescence; and protein and mRNA expression of Kiss1, Ar, Esr1, and kisspeptin receptor (Kiss1r) was evaluated via western blotting and Reverse Transcription-Polymerase Chain Reaction (RT-PCR). RESULTS The results revealed that the estrous cycle of rats in the EA treatment group recovered, and their body and ovary weight reduced; ovarian morphology improved; serum testosterone and LH levels significantly decreased; and kisspeptin, GnRH, and dynorphin levels in hypothalamic arcuate nucleus significantly decreased. Compared with controls, the number of AR/Kiss1-positive cells increased, number of ERα/Kiss1-positive cells decreased, and protein and mRNA expression of Kiss1, Ar, and Kiss1r significantly increased in PCOS rats. However, EA treatment reversed these changes and reduced the expression of Kiss1, Ar, and Kiss1r significantly. CONCLUSION Improvement in the reproductive hallmarks of PCOS rats via EA may be achieved by regulating the kisspeptin-GnRH/LH circuit via androgen activity attenuation. Thus, the results provide an experimental basis for acupuncture as an adjuvant medical therapy on PCOS.
Collapse
Affiliation(s)
- Ge Xu
- grid.411480.80000 0004 1799 1816Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China ,grid.412540.60000 0001 2372 7462Shanghai University of Traditional Chinese Medicine, Shanghai, 201210 China
| | - Xuedan Zhao
- grid.411480.80000 0004 1799 1816Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China ,grid.412540.60000 0001 2372 7462Shanghai University of Traditional Chinese Medicine, Shanghai, 201210 China
| | - Zhihao Li
- grid.411480.80000 0004 1799 1816Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
| | - Junwei Hu
- grid.411480.80000 0004 1799 1816Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
| | - Xu Li
- grid.411480.80000 0004 1799 1816Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
| | - Juanjuan Li
- grid.411480.80000 0004 1799 1816Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
| | - Yuelai Chen
- grid.411480.80000 0004 1799 1816Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
| |
Collapse
|
29
|
Hatcher KM, Costanza L, Kauffman AS, Stephens SBZ. The molecular phenotype of kisspeptin neurons in the medial amygdala of female mice. Front Endocrinol (Lausanne) 2023; 14:1093592. [PMID: 36843592 PMCID: PMC9951589 DOI: 10.3389/fendo.2023.1093592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/13/2023] [Indexed: 02/12/2023] Open
Abstract
Reproduction is regulated through the hypothalamic-pituitary-gonadal (HPG) axis, largely via the action of kisspeptin neurons in the hypothalamus. Importantly, Kiss1 neurons have been identified in other brain regions, including the medial amygdala (MeA). Though the MeA is implicated in regulating aspects of both reproductive physiology and behavior, as well as non-reproductive processes, the functional roles of MeA Kiss1 neurons are largely unknown. Additionally, besides their stimulation by estrogen, little is known about how MeA Kiss1 neurons are regulated. Using a RiboTag mouse model in conjunction with RNA-seq, we examined the molecular profile of MeA Kiss1 neurons to identify transcripts that are co-expressed in MeA Kiss1 neurons of female mice and whether these transcripts are modulated by estradiol (E2) treatment. RNA-seq identified >13,800 gene transcripts co-expressed in female MeA Kiss1 neurons, including genes for neuropeptides and receptors implicated in reproduction, metabolism, and other neuroendocrine functions. Of the >13,800 genes co-expressed in MeA Kiss1 neurons, only 45 genes demonstrated significantly different expression levels due to E2 treatment. Gene transcripts such as Kiss1, Gal, and Oxtr increased in response to E2 treatment, while fewer transcripts, such as Esr1 and Cyp26b1, were downregulated by E2. Dual RNAscope and immunohistochemistry was performed to validate co-expression of MeA Kiss1 with Cck and Cartpt. These results are the first to establish a profile of genes actively expressed by MeA Kiss1 neurons, including a subset of genes regulated by E2, which provides a useful foundation for future investigations into the regulation and function of MeA Kiss1 neurons.
Collapse
Affiliation(s)
- Katherine M. Hatcher
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Leah Costanza
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Alexander S. Kauffman
- Department of OBGYN and Reproductive Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Shannon B. Z. Stephens
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
- Department of OBGYN and Reproductive Sciences, University of California, San Diego, La Jolla, CA, United States
- *Correspondence: Shannon B. Z. Stephens,
| |
Collapse
|
30
|
Leonardi CEP, Carrasco RA, Dias FCF, Zwiefelhofer EM, Adams GP, Singh J. Mechanism of LH release after peripheral administration of kisspeptin in cattle. PLoS One 2022; 17:e0278564. [PMID: 36459509 PMCID: PMC9718405 DOI: 10.1371/journal.pone.0278564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 11/18/2022] [Indexed: 12/04/2022] Open
Abstract
Kisspeptin modulates GnRH secretion in mammals and peripheral administration of 10-amino acid fragment of kisspeptin (Kp10) induces LH release and ovulation in cattle. Experiments were done to determine if iv administration of kisspeptin will activate GnRH neurons (i.e., after crossing the blood-brain barrier) and if pre-treatment with a GnRH receptor blocker will alter kisspeptin-induced LH release (from gonadotrophs) and ovulation. In Experiment 1, cows (n = 3 per group) were given human-Kisspeptin10 (hKp10; 3 x 15 mg iv at 60-min intervals) or normal saline and euthanized 150 min after treatment was initiated. Every 20th free-floating section (50 μm thickness) from the preoptic area to hypothalamus was double immunostained to colocalize GnRH- (DAB) and activated neurons (cFOS; Nickel-DAB). Kisspeptin induced plasma LH release from 15 to 150 min (P = 0.01) but the proportion of activated GnRH neurons did not differ between groups (5.8% and 3.5%, respectively; P = 0.11). Immunogold electron microscopy detected close contacts between kisspeptin fibers and GnRH terminals in the median eminence. In Experiment 2, pubertal heifers (n = 5 per group) were treated with 1) hKp10 iv, 2) Cetrorelix (GnRH antagonist; im) + hKp10 iv or 3) saline on Day 6 of the follicular wave under low-progesterone condition. A rise in plasma LH concentration was detected from 15 to 240 min in the hKp10 group but not in cetrorelix or control group (P<0.001). Ovulations were detected only in the hKp10 group (4/5; P = 0.02). Cetrorelix treatment was associated with regression of the preovulatory dominant follicle and emergence of a new follicular wave 3.4±0.75 days after the treatment in all five heifers. Results support the hypothesis that the effect of peripheral kisspeptin is mediated downstream of GnRH synthesis and does not involve GnRH-independent LH release from gonadotrophs. Peripheral kisspeptin may release pre-synthesized GnRH from the nerve terminals in areas outside the blood-brain barrier.
Collapse
Affiliation(s)
- Carlos E. P. Leonardi
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Rodrigo A. Carrasco
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Fernanda C. F. Dias
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Eric M. Zwiefelhofer
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Gregg P. Adams
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Jaswant Singh
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
- * E-mail:
| |
Collapse
|
31
|
Mills EG, Dhillo WS. Invited review: Translating kisspeptin and neurokinin B biology into new therapies for reproductive health. J Neuroendocrinol 2022; 34:e13201. [PMID: 36262016 PMCID: PMC9788075 DOI: 10.1111/jne.13201] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 07/26/2022] [Accepted: 09/06/2022] [Indexed: 12/30/2022]
Abstract
The reproductive neuropeptide kisspeptin has emerged as the master regulator of mammalian reproduction due to its key roles in the initiation of puberty and the control of fertility. Alongside the tachykinin neurokinin B and the endogenous opioid dynorphin, these peptides are central to the hormonal control of reproduction. Building on the expanding body of experimental animal models, interest has flourished with human studies revealing that kisspeptin administration stimulates physiological reproductive hormone secretion in both healthy men and women, as well as patients with common reproductive disorders. In addition, emerging therapeutic roles based on neurokinin B for the management of menopausal flushing, endometriosis and uterine fibroids are increasingly recognised. In this review, we focus on kisspeptin and neurokinin B and their potential application as novel clinical strategies for the management of reproductive disorders.
Collapse
Affiliation(s)
- Edouard G. Mills
- Section of Endocrinology and Investigative MedicineImperial College LondonLondonUK
- Department of EndocrinologyImperial College Healthcare NHS TrustLondonUK
| | - Waljit S. Dhillo
- Section of Endocrinology and Investigative MedicineImperial College LondonLondonUK
- Department of EndocrinologyImperial College Healthcare NHS TrustLondonUK
| |
Collapse
|
32
|
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
|
33
|
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: 12] [Impact Index Per Article: 6.0] [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
|
34
|
Stincic TL, Kelly MJ. Estrogenic regulation of reproduction and energy homeostasis by a triumvirate of hypothalamic arcuate neurons. J Neuroendocrinol 2022; 34:e13145. [PMID: 35581942 DOI: 10.1111/jne.13145] [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: 12/18/2021] [Revised: 03/31/2022] [Accepted: 04/15/2022] [Indexed: 11/29/2022]
Abstract
Pregnancy is energetically demanding and therefore, by necessity, reproduction and energy balance are inextricably linked. With insufficient or excessive energy stores a female is liable to suffer complications during pregnancy or produce unhealthy offspring. Gonadotropin-releasing hormone neurons are responsible for initiating both the pulsatile and subsequent surge release of luteinizing hormone to control ovulation. Meticulous work has identified two hypothalamic populations of kisspeptin (Kiss1) neurons that are critical for this pattern of release. The involvement of the hypothalamus is unsurprising because its quintessential function is to couple the endocrine and nervous systems, coordinating energy balance and reproduction. Estrogens, more specifically 17β-estradiol (E2 ), orchestrate the activity of a triumvirate of hypothalamic neurons within the arcuate nucleus (ARH) that govern the physiological underpinnings of these behavioral dynamics. Arising from a common progenitor pool, these cells differentiate into ARH kisspeptin, pro-opiomelanocortin (POMC), and agouti related peptide/neuropeptide Y (AgRP) neurons. Although the excitability of all these subpopulations is subject to genomic and rapid estrogenic regulation, Kiss1 neurons are the most sensitive, reflecting their integral function in female fertility. Based on the premise that E2 coordinates autonomic functions around reproduction, we review recent findings on how Kiss1 neurons interact with gonadotropin-releasing hormone, AgRP and POMC neurons, as well as how the rapid membrane-initiated and intracellular signaling cascades activated by E2 in these neurons are critical for control of homeostatic functions supporting reproduction. In particular, we highlight how Kiss1 and POMC neurons conspire to inhibit AgRP neurons and diminish food motivation in service of reproductive success.
Collapse
Affiliation(s)
- Todd L Stincic
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
| | - Martin J Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| |
Collapse
|
35
|
Harlow K, Griesgraber MJ, Seman AD, Shuping SL, Sommer JR, Griffith EH, Hileman SM, Nestor CC. The impact of undernutrition on KNDy (kisspeptin/neurokinin B/dynorphin) neurons in female lambs. J Neuroendocrinol 2022; 34:e13135. [PMID: 35579068 PMCID: PMC9286635 DOI: 10.1111/jne.13135] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 03/25/2022] [Accepted: 03/27/2022] [Indexed: 11/30/2022]
Abstract
Undernutrition limits reproduction through inhibition of gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) secretion. Because KNDy neurons coexpress neuropeptides that play stimulatory (kisspeptin and neurokinin B [NKB]) and inhibitory (dynorphin) roles in pulsatile GnRH/LH release, we hypothesized that undernutrition would inhibit kisspeptin and NKB expression at the same time as increasing dynorphin expression. Fifteen ovariectomized lambs were either fed to maintain pre-study body weight (controls) or feed-restricted to lose 20% of pre-study body weight (FR) over 13 weeks. Blood samples were collected and plasma from weeks 0 and 13 were assessed for LH by radioimmunoassay. At week 13, animals were killed, and brain tissue was processed for assessment of KNDy peptide mRNA or protein expression. Mean LH and LH pulse amplitude were lower in FR lambs compared to controls. We observed lower mRNA abundance for kisspeptin within KNDy neurons of FR lambs compared to controls with no significant change in mRNA for NKB or dynorphin. We also observed that FR lambs had fewer numbers of arcuate nucleus kisspeptin and NKB perikarya compared to controls. These findings support the idea that KNDy neurons are important for regulating reproduction during undernutrition in female sheep.
Collapse
Affiliation(s)
- KaLynn Harlow
- Department of Animal ScienceNorth Carolina State UniversityRaleighNCUSA
| | - Max J. Griesgraber
- Department of Physiology and PharmacologyWest Virginia UniversityMorgantownWVUSA
| | - Andrew D. Seman
- Department of Physiology and PharmacologyWest Virginia UniversityMorgantownWVUSA
| | - Sydney L. Shuping
- Department of Animal ScienceNorth Carolina State UniversityRaleighNCUSA
| | - Jeffrey R. Sommer
- Department of Animal ScienceNorth Carolina State UniversityRaleighNCUSA
| | | | - Stanley M. Hileman
- Department of Physiology and PharmacologyWest Virginia UniversityMorgantownWVUSA
- Department of NeuroscienceWest Virginia UniversityMorgantownWVUSA
| | - Casey C Nestor
- Department of Animal ScienceNorth Carolina State UniversityRaleighNCUSA
| |
Collapse
|
36
|
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: 11] [Impact Index Per Article: 5.5] [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
|
37
|
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: 14] [Impact Index Per Article: 7.0] [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
|
38
|
Grassi D, Marraudino M, Garcia-Segura LM, Panzica GC. The hypothalamic paraventricular nucleus as a central hub for the estrogenic modulation of neuroendocrine function and behavior. Front Neuroendocrinol 2022; 65:100974. [PMID: 34995643 DOI: 10.1016/j.yfrne.2021.100974] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/17/2022]
Abstract
Estradiol and hypothalamic paraventricular nucleus (PVN) help coordinate reproduction with body physiology, growth and metabolism. PVN integrates hormonal and neural signals originating in the periphery, generating an output mediated both by its long-distance neuronal projections, and by a variety of neurohormones produced by its magnocellular and parvocellular neurosecretory cells. Here we review the cyto-and chemo-architecture, the connectivity and function of PVN and the sex-specific regulation exerted by estradiol on PVN neurons and on the expression of neurotransmitters, neuromodulators, neuropeptides and neurohormones in PVN. Classical and non-classical estrogen receptors (ERs) are expressed in neuronal afferents to PVN and in specific PVN interneurons, projecting neurons, neurosecretory neurons and glial cells that are involved in the input-output integration and coordination of neurohormonal signals. Indeed, PVN ERs are known to modulate body homeostatic processes such as autonomic functions, stress response, reproduction, and metabolic control. Finally, the functional implications of the estrogenic modulation of the PVN for body homeostasis are discussed.
Collapse
Affiliation(s)
- D Grassi
- Department of Anatomy, Histology and Neuroscience, Universidad Autonoma de Madrid, Madrid, Spain
| | - M Marraudino
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy
| | - L M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - G C Panzica
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy; Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy.
| |
Collapse
|
39
|
Lee EB, Dilower I, Marsh CA, Wolfe MW, Masumi S, Upadhyaya S, Rumi MAK. Sexual Dimorphism in Kisspeptin Signaling. Cells 2022; 11:1146. [PMID: 35406710 PMCID: PMC8997554 DOI: 10.3390/cells11071146] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 02/05/2023] Open
Abstract
Kisspeptin (KP) and kisspeptin receptor (KPR) are essential for the onset of puberty, development of gonads, and maintenance of gonadal function in both males and females. Hypothalamic KPs and KPR display a high degree of sexual dimorphism in expression and function. KPs act on KPR in gonadotropin releasing hormone (GnRH) neurons and induce distinct patterns of GnRH secretion in males and females. GnRH acts on the anterior pituitary to secrete gonadotropins, which are required for steroidogenesis and gametogenesis in testes and ovaries. Gonadal steroid hormones in turn regulate the KP neurons. Gonadal hormones inhibit the KP neurons within the arcuate nucleus and generate pulsatile GnRH mediated gonadotropin (GPN) secretion in both sexes. However, the numbers of KP neurons in the anteroventral periventricular nucleus and preoptic area are greater in females, which release a large amount of KPs in response to a high estrogen level and induce the preovulatory GPN surge. In addition to the hypothalamus, KPs and KPR are also expressed in various extrahypothalamic tissues including the liver, pancreas, fat, and gonads. There is a remarkable difference in circulating KP levels between males and females. An increased level of KPs in females can be linked to increased numbers of KP neurons in female hypothalamus and more KP production in the ovaries and adipose tissues. Although the sexually dimorphic features are well characterized for hypothalamic KPs, very little is known about the extrahypothalamic KPs. This review article summarizes current knowledge regarding the sexual dimorphism in hypothalamic as well as extrahypothalamic KP and KPR system in primates and rodents.
Collapse
Affiliation(s)
- Eun Bee Lee
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Iman Dilower
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Courtney A. Marsh
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Michael W. Wolfe
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Saeed Masumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| | - Sameer Upadhyaya
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| | - Mohammad A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| |
Collapse
|
40
|
Dai Pra R, Mohr SM, Merriman DK, Bagriantsev SN, Gracheva EO. Ground squirrels initiate sexual maturation during hibernation. Curr Biol 2022; 32:1822-1828.e4. [PMID: 35245461 DOI: 10.1016/j.cub.2022.02.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/15/2021] [Accepted: 02/09/2022] [Indexed: 12/01/2022]
Abstract
Adequate nutrition is essential for normal reproductive function, which is vital for species to survive. In humans and other mammals, starvation and undernutrition deplete fat reserves and cause weight loss, attenuating the function of the reproductive axis and causing hypogonadism.1-4 Thirteen-lined ground squirrels (Ictidomys tridecemlineatus) spend 7 months of every year in hibernation without food and water. Hibernating squirrels alternate between periods of torpor and interbout arousal (IBA), when animals temporarily return to an active-like state.5 The physiological significance of IBA is unclear, but it is thought to be essential for hibernation in animals that drop their body temperature to 2°C-4°C during torpor. Here, we report that juvenile male ground squirrels initiate reproductive maturation during their first hibernation season, despite prolonged undernutrition and profound weight loss. We show that the hypothalamic reproductive axis undergoes activation during interbout arousals in the middle of hibernation, triggering production of luteinizing hormone and testosterone, and promoting testicular growth. Initiation of sexual maturation is circannually entrained and is independent of physiological state, ambient temperature, and food availability. Our study suggests a role for interbout arousals during hibernation and uncovers the neurophysiological mechanism of reproductive axis activation during conditions of extreme negative energy balance.
Collapse
Affiliation(s)
- Rafael Dai Pra
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Sarah M Mohr
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Dana K Merriman
- Department of Biology, University of Wisconsin-Oshkosh, 800 Algoma Boulevard, Oshkosh, WI 54901, USA
| | - Sviatoslav N Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA.
| | - Elena O Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA.
| |
Collapse
|
41
|
Sivalingam M, Ogawa S, Trudeau VL, Parhar IS. Conserved functions of hypothalamic kisspeptin in vertebrates. Gen Comp Endocrinol 2022; 317:113973. [PMID: 34971635 DOI: 10.1016/j.ygcen.2021.113973] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022]
Abstract
Hypothalamic kisspeptin encoded by KISS1/Kiss1 gene emerged as a regulator of the reproductive axis in mammals following the discovery of the kisspeptin receptor (Kissr) and its role in reproduction. Kisspeptin-Kissr systems have been investigated in various vertebrates, and a conserved sequence of kisspeptin-Kissr has been identified in most vertebrate species except in the avian linage. In addition, multiple paralogs of kisspeptin sequences have been identified in the non-mammalian vertebrates. The allegedly conserved role of kisspeptin-Kissr in reproduction became debatable when kiss/kissr genes-deficient zebrafish and medaka showed no apparent effect on the onset of puberty, sexual development, maturation and reproductive capacity. Therefore, it is questionable whether the role of kisspeptin in reproduction is conserved among vertebrate species. Here we discuss from a comparative and evolutional aspect the diverse functions of kisspeptin and its receptor in vertebrates. Primarily this review focuses on the role of hypothalamic kisspeptin in reproductive and non-reproductive functions that are conserved in vertebrate species.
Collapse
Affiliation(s)
- Mageswary Sivalingam
- Brain Research Institute, Jeffery Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Petaling Jaya, Selangor, Malaysia
| | - Satoshi Ogawa
- Brain Research Institute, Jeffery Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Petaling Jaya, Selangor, Malaysia
| | - Vance L Trudeau
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Ishwar S Parhar
- Brain Research Institute, Jeffery Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Petaling Jaya, Selangor, Malaysia.
| |
Collapse
|
42
|
Flay H, Reed C, Kuhn-Sherlock B, Phyn C, Burke C, Meier S, Clarke I. Response to kisspeptin and GnRH agonist administration in Holstein-Friesian dairy heifers with positive or negative genetic merit for fertility traits. J Dairy Sci 2022; 105:3601-3614. [DOI: 10.3168/jds.2021-21394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/13/2021] [Indexed: 11/19/2022]
|
43
|
Rønnekleiv OK, Qiu J, Kelly MJ. Hypothalamic Kisspeptin Neurons and the Control of Homeostasis. Endocrinology 2022; 163:bqab253. [PMID: 34953135 PMCID: PMC8758343 DOI: 10.1210/endocr/bqab253] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Indexed: 12/27/2022]
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
|
44
|
Gołyszny M, Obuchowicz E, Zieliński M. Neuropeptides as regulators of the hypothalamus-pituitary-gonadal (HPG) axis activity and their putative roles in stress-induced fertility disorders. Neuropeptides 2022; 91:102216. [PMID: 34974357 DOI: 10.1016/j.npep.2021.102216] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/21/2021] [Accepted: 10/05/2021] [Indexed: 11/24/2022]
Abstract
Neuropeptides being regulators of the hypothalamus-pituitary-adrenal (HPA) axis activity, also affect the function of the hypothalamus-pituitary-gonadal (HPG) axis by regulating gonadotrophin-releasing hormone (GnRH) secretion from hypothalamic neurons. Here, we review the available data on how neuropeptides affect HPG axis activity directly or indirectly via their influence on the HPA axis. The putative role of neuropeptides in stress-induced infertility, such as polycystic ovary syndrome, is also described. This review discusses both well-known neuropeptides (i.e., kisspeptin, Kp; oxytocin, OT; arginine-vasopressin, AVP) and more recently discovered peptides (i.e., relaxin-3, RLN-3; nesfatin-1, NEFA; phoenixin, PNX; spexin, SPX). For the first time, we present an up-to-date review of all published data regarding interactions between the aforementioned neuropeptide systems. The reviewed literature suggest new pathophysiological mechanisms leading to fertility disturbances that are induced by stress.
Collapse
Affiliation(s)
- Miłosz Gołyszny
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18 Street, 40-752 Katowice, Poland.
| | - Ewa Obuchowicz
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18 Street, 40-752 Katowice, Poland.
| | - Michał Zieliński
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18 Street, 40-752 Katowice, Poland.
| |
Collapse
|
45
|
Tsukamura H. Kobayashi Award 2019: The neuroendocrine regulation of the mammalian reproduction. Gen Comp Endocrinol 2022; 315:113755. [PMID: 33711315 DOI: 10.1016/j.ygcen.2021.113755] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/13/2021] [Accepted: 02/23/2021] [Indexed: 02/05/2023]
Abstract
Mammalian reproductive function is a complex system of many players orchestrated by the hypothalamus-pituitary-gonadal (HPG) axis. The hypothalamic gonadotropin-releasing hormone (GnRH) and the consequent pituitary gonadotropin release show two modes of secretory patterns, namely the surge and pulse modes. The surge mode is triggered by the positive feedback action of estrogen secreted from the mature ovarian follicle to induce ovulation in females of most mammalian species. The pulse mode of GnRH release is required for stimulating tonic gonadotropin secretion to drive folliculogenesis, spermatogenesis and steroidogenesis and is negatively fine-tuned by the sex steroids. Accumulating evidence suggests that hypothalamic kisspeptin neurons are the master regulator for animal reproduction to govern the HPG axis. Specifically, kisspeptin neurons located in the anterior hypothalamus, such as the anteroventral periventricular nucleus (AVPV) in rodents and preoptic nucleus (POA) in ruminants, primates and others, and the neurons located in the arcuate nucleus (ARC) in posterior hypothalamus in most mammals are considered to play a key role in generating the surge and pulse modes of GnRH release, respectively. The present article focuses on the role of AVPV (or POA) kisspeptin neurons as a center for GnRH surge generation and of the ARC kisspeptin neurons as a center for GnRH pulse generation to mediate estrogen positive and negative feedback mechanisms, respectively, and discusses how the estrogen epigenetically regulates kisspeptin gene expression in these two populations of neurons. This article also provides the mechanism how malnutrition and lactation suppress GnRH/gonadotropin pulses through an inhibition of the ARC kisspeptin neurons. Further, the article discusses the programming effect of estrogen on kisspeptin neurons in the developmental brain to uncover the mechanism underlying the sex difference in GnRH/gonadotropin release as well as an irreversible infertility induced by supra-physiological estrogen exposure in rodent models.
Collapse
Affiliation(s)
- Hiroko Tsukamura
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan.
| |
Collapse
|
46
|
Hypothalamic kisspeptin and kisspeptin receptors: Species variation in reproduction and reproductive behaviours. Front Neuroendocrinol 2022; 64:100951. [PMID: 34757093 DOI: 10.1016/j.yfrne.2021.100951] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/22/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023]
Abstract
Kisspeptin, encoded by the KISS1 gene, was first discovered as a potential metastasis suppressor gene. The prepro-kisspeptin precursor is cleaved into shorter mature bioactive peptides of varying sizes that bind to the G protein-coupled receptor GPR54 (=KISS1R). Over the last two decades, multiple types of Kiss and KissR genes have been discovered in mammalian and non-mammalian vertebrate species, but they are remarkably absent in birds. Kiss neuronal populations are distributed mainly in the hypothalamus. The KissRs are widely distributed in the brain, including the hypothalamic and non-hypothalamic regions, such as the hippocampus, amygdala, and habenula. The role of KISS1-KISS1R in humans and Kiss1-Kiss1R in rodents is associated with puberty, gonadal maturation, and the reproductive axis. However, recent gene deletion studies in zebrafish and medaka have provided controversial results, suggesting that the reproductive role of kiss is dispensable. This review highlights the evolutionary history, localisation, and significance of Kiss-KissR in reproduction and reproductive behaviours in mammalian and non-mammalian vertebrates.
Collapse
|
47
|
Ozawa H. Kisspeptin neurons as an integration center of reproductive regulation: Observation of reproductive function based on a new concept of reproductive regulatory nervous system. Reprod Med Biol 2021; 21:e12419. [PMID: 34934400 PMCID: PMC8656200 DOI: 10.1002/rmb2.12419] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 11/29/2022] Open
Abstract
Background Regulation of the reproductive system has been explained by the actions and feedback of gonadotropin releasing hormone‐luteinizing hormone/follicle stimulating hormone (GnRH‐LH/FSH) ‐sex steroids; however, the discovery of kisspeptin neurons and a kisspeptin‐GnRH‐LH/FSH axis has prompted this regulation to be reviewed. Methods We investigated changes in kisspeptin neurons and associated changes in the hypothalamic‐pituitary‐gonadal (HPG) axis under various situations and experimental conditions using histochemical methods. Main findings (Results) Kisspeptin neurons play an important role in receiving and integrating information from internal and external environmental factors and communicating it to the conventional HPG axis. Conclusion The recently described Kisspeptin‐GnRH‐LH/FSH‐gonad system regulates reproductive function via mechanisms that until recently were not completely understood.
Collapse
Affiliation(s)
- Hitoshi Ozawa
- Department of Anatomy and Neurobiology Graduate School of Medicine Nippon Medical School Tokyo Japan
| |
Collapse
|
48
|
Moore AM, Lohr DB, Coolen LM, Lehman MN. Prenatal Androgen Exposure Alters KNDy Neurons and Their Afferent Network in a Model of Polycystic Ovarian Syndrome. Endocrinology 2021; 162:bqab158. [PMID: 34346492 PMCID: PMC8402932 DOI: 10.1210/endocr/bqab158] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Indexed: 02/08/2023]
Abstract
Polycystic ovarian syndrome (PCOS), the most common endocrinopathy affecting women worldwide, is characterized by elevated luteinizing hormone (LH) pulse frequency due to the impaired suppression of gonadotrophin-releasing hormone (GnRH) release by steroid hormone negative feedback. Although neurons that co-express kisspeptin, neurokinin B, and dynorphin (KNDy cells) were recently defined as the GnRH/LH pulse generator, little is understood about their role in the pathogenesis of PCOS. We used a prenatal androgen-treated (PNA) mouse model of PCOS to determine whether changes in KNDy neurons or their afferent network underlie altered negative feedback. First, we identified elevated androgen receptor gene expression in KNDy cells of PNA mice, whereas progesterone receptor and dynorphin gene expression was significantly reduced, suggesting elevated androgens in PCOS disrupt progesterone negative feedback via direct actions upon KNDy cells. Second, we discovered GABAergic and glutamatergic synaptic input to KNDy neurons was reduced in PNA mice. Retrograde monosynaptic tract-tracing revealed a dramatic reduction in input originates from sexually dimorphic afferents in the preoptic area, anteroventral periventricular nucleus, anterior hypothalamic area and lateral hypothalamus. These results reveal 2 sites of neuronal alterations potentially responsible for defects in negative feedback in PCOS: changes in gene expression within KNDy neurons, and changes in synaptic inputs from steroid hormone-responsive hypothalamic regions. How each of these changes contribute to the neuroendocrine phenotype seen in in PCOS, and the role of specific sets of upstream KNDy afferents in the process, remains to be determined.
Collapse
Affiliation(s)
- Aleisha M Moore
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| | - Dayanara B Lohr
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| | - Lique M Coolen
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| | - Michael N Lehman
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
49
|
Harlow K, Renwick AN, Shuping SL, Sommer JR, Lents CA, Knauer MT, Nestor CC. Evidence that pubertal status impacts KNDy neurons in the gilt. Biol Reprod 2021; 105:1533-1544. [PMID: 34643223 DOI: 10.1093/biolre/ioab189] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/26/2022] Open
Abstract
Puberty onset is a complex physiological process which enables the capacity for reproduction through increased gonadotropin-releasing hormone (GnRH), and subsequently luteinizing hormone (LH), secretion. While cells that coexpress kisspeptin, neurokinin B (NKB), and dynorphin in the hypothalamic arcuate nucleus (ARC) are believed to govern the timing of puberty, the degree to which KNDy neurons exist and are regulated by pubertal status remains to be determined in the gilt. Hypothalamic tissue from prepubertal and postpubertal, early follicular phase gilts was used to determine the expression of kisspeptin, NKB, and dynorphin within the ARC. Fluorescent in situ hybridization revealed that the majority (> 74%) of ARC neurons that express mRNA for kisspeptin coexpressed mRNA for NKB and dynorphin. There were fewer ARC cells that expressed mRNA for dynorphin in postpubertal gilts compared to prepubertal gilts (P < 0.05), but the number of ARC cells expressing mRNA for kisspeptin or NKB was not different between groups. Within KNDy neurons, mRNA abundance for kisspeptin, NKB, and dynorphin of postpubertal gilts was the same as, less than, and greater than, respectively, prepubertal gilts. Immunostaining for kisspeptin did not differ between prepubertal and postpubertal gilts, but there were fewer NKB immunoreactive fibers in postpubertal gilts compared to prepubertal gilts (P < 0.05). Together, these data reveal novel information about KNDy neurons in gilts and supports the idea that NKB and dynorphin play a role in puberty onset in the female pig.
Collapse
Affiliation(s)
- KaLynn Harlow
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695 USA
| | - Allison N Renwick
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695 USA
| | - Sydney L Shuping
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695 USA
| | - Jeffrey R Sommer
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695 USA
| | - Clay A Lents
- USDA, ARS, U.S. Meat Animal Research Center, Livestock Biosystems Research Unit, Clay Center, NE 68966-0166, USA
| | - Mark T Knauer
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695 USA
| | - Casey C Nestor
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695 USA
| |
Collapse
|
50
|
Poissenot K, Chorfa A, Moussu C, Trouillet AC, Brachet M, Chesneau D, Chemineau P, Ramadier E, Pinot A, Benoit E, Lattard V, Dardente H, Drevet J, Saez F, Keller M. Photoperiod is involved in the regulation of seasonal breeding in male water voles (Arvicola terrestris). J Exp Biol 2021; 224:272112. [PMID: 34494651 DOI: 10.1242/jeb.242792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/26/2021] [Indexed: 12/27/2022]
Abstract
Mammals living at temperate latitudes typically display annual cyclicity in their reproductive activity: births are synchronized when environmental conditions are most favorable. In a majority of these species, day length is the main proximate factor used to anticipate seasonal changes and to adapt physiology. The brain integrates this photoperiodic signal through key hypothalamic structures, which regulate the reproductive axis. In this context, our study aimed to characterize regulations that occur along the hypothalamo-pituitary-gonadal (HPG) axis in male fossorial water voles (Arvicola terrestris, also known as Arvicola amphibius) throughout the year and to further probe the implication of photoperiod in these seasonal regulations. Our monthly field monitoring showed dramatic seasonal changes in the morphology and activity of reproductive organs, as well as in the androgen-dependent lateral scent glands. Moreover, our data uncovered seasonal variations at the hypothalamic level. During the breeding season, kisspeptin expression in the arcuate nucleus (ARC) decreases, while RFRP3 expression in the dorsomedial hypothalamic nucleus (DMH) increases. Our follow-up laboratory study revealed activation of the reproductive axis and confirmed a decrease in kisspeptin expression in males exposed to a long photoperiod (summer condition) compared with those maintained under a short photoperiod (winter condition) that retain all features reminiscent of sexual inhibition. Altogether, our study characterizes neuroendocrine and anatomical markers of seasonal reproductive rhythmicity in male water voles and further suggests that these seasonal changes are strongly impacted by photoperiod.
Collapse
Affiliation(s)
- Kevin Poissenot
- Physiologie de la Reproduction et des Comportements, UMR INRAE, CNRS, Université de Tours, IFCE, 37380 Nouzilly, France
| | - Areski Chorfa
- GReD Laboratory, CNRS UMR 6293 - INSERM U1103 - Université Clermont Auvergne, 28 place Henri Dunant, 63001, Clermont-Ferrand Cedex, France
| | - Chantal Moussu
- Physiologie de la Reproduction et des Comportements, UMR INRAE, CNRS, Université de Tours, IFCE, 37380 Nouzilly, France
| | - Anne-Charlotte Trouillet
- Physiologie de la Reproduction et des Comportements, UMR INRAE, CNRS, Université de Tours, IFCE, 37380 Nouzilly, France
| | - Morgane Brachet
- Physiologie de la Reproduction et des Comportements, UMR INRAE, CNRS, Université de Tours, IFCE, 37380 Nouzilly, France
| | - Didier Chesneau
- Physiologie de la Reproduction et des Comportements, UMR INRAE, CNRS, Université de Tours, IFCE, 37380 Nouzilly, France
| | - Philippe Chemineau
- Physiologie de la Reproduction et des Comportements, UMR INRAE, CNRS, Université de Tours, IFCE, 37380 Nouzilly, France
| | - Etienne Ramadier
- USC 1233 RS2GP, INRAE, VetAgro Sup, Université de Lyon, F-69280 Marcy l'Etoile, France
| | - Adrien Pinot
- USC 1233 RS2GP, INRAE, VetAgro Sup, Université de Lyon, F-69280 Marcy l'Etoile, France
| | - Etienne Benoit
- USC 1233 RS2GP, INRAE, VetAgro Sup, Université de Lyon, F-69280 Marcy l'Etoile, France
| | - Virginie Lattard
- USC 1233 RS2GP, INRAE, VetAgro Sup, Université de Lyon, F-69280 Marcy l'Etoile, France
| | - Hugues Dardente
- Physiologie de la Reproduction et des Comportements, UMR INRAE, CNRS, Université de Tours, IFCE, 37380 Nouzilly, France
| | - Joël Drevet
- GReD Laboratory, CNRS UMR 6293 - INSERM U1103 - Université Clermont Auvergne, 28 place Henri Dunant, 63001, Clermont-Ferrand Cedex, France
| | - Fabrice Saez
- GReD Laboratory, CNRS UMR 6293 - INSERM U1103 - Université Clermont Auvergne, 28 place Henri Dunant, 63001, Clermont-Ferrand Cedex, France
| | - Matthieu Keller
- Physiologie de la Reproduction et des Comportements, UMR INRAE, CNRS, Université de Tours, IFCE, 37380 Nouzilly, France
| |
Collapse
|