1
|
Hemminger Z, Sanchez-Tam G, Ocampo HD, Wang A, Underwood T, Xie F, Zhao Q, Song D, Li JJ, Dong H, Wollman R. Spatial Single-Cell Mapping of Transcriptional Differences Across Genetic Backgrounds in Mouse Brains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617260. [PMID: 39416191 PMCID: PMC11483037 DOI: 10.1101/2024.10.08.617260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Genetic variation can alter brain structure and, consequently, function. Comparative statistical analysis of mouse brains across genetic backgrounds requires spatial, single-cell, atlas-scale data, in replicates-a challenge for current technologies. We introduce Atlas-scale Transcriptome Localization using Aggregate Signatures (ATLAS), a scalable tissue mapping method. ATLAS learns transcriptional signatures from scRNAseq data, encodes them in situ with tens of thousands of oligonucleotide probes, and decodes them to infer cell types and imputed transcriptomes. We validated ATLAS by comparing its cell type inferences with direct MERFISH measurements of marker genes and quantitative comparisons to four other technologies. Using ATLAS, we mapped the central brains of five male and five female C57BL/6J (B6) mice and five male BTBR T+ tf/J (BTBR) mice, an idiopathic model of autism, collectively profiling over 40 million cells across over 400 coronal sections. Our analysis revealed over 40 significant differences in cell type distributions and identified 16 regional composition changes across male-female and B6-BTBR comparisons. ATLAS thus enables systematic comparative studies, facilitating organ-level structure-function analysis of disease models.
Collapse
Affiliation(s)
| | | | | | - Aihui Wang
- Department of Chemistry and Biochemistry, UCLA
| | | | - Fangming Xie
- Department of Chemical Biology, David Geffen School of Medicine at UCLA
| | - Qiuying Zhao
- Department of Neurobiology, David Geffen School of Medicine at UCLA
| | | | - Jingyi Jessica Li
- Department of Statistics and Data Science, UCLA
- Institute of Quantitative Biosciences, UCLA
| | - Hongwei Dong
- Department of Neurobiology, David Geffen School of Medicine at UCLA
| | - Roy Wollman
- Department of Chemistry and Biochemistry, UCLA
- Institute of Quantitative Biosciences, UCLA
- Department of Integrative Biology and Physiology, UCLA
| |
Collapse
|
2
|
Patel AH, Koysombat K, Pierret A, Young M, Comninos AN, Dhillo WS, Abbara A. Kisspeptin in functional hypothalamic amenorrhea: Pathophysiology and therapeutic potential. Ann N Y Acad Sci 2024; 1540:21-46. [PMID: 39287750 DOI: 10.1111/nyas.15220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Functional hypothalamic amenorrhea (FHA) is one of the most common causes of secondary amenorrhea, resulting in anovulation and infertility, and is a low estrogen state that increases the risk of cardiovascular disease and impairs bone health. FHA is characterized by acquired suppression of physiological pulsatile gonadotropin-releasing hormone (GnRH) release by the hypothalamus in the absence of an identifiable structural cause, resulting in a functional hypogonadotropic hypogonadism. FHA results from either decreased energy intake and/or excessive exercise, leading to low energy availability and weight loss-often in combination with psychological stress on top of a background of genetic susceptibility. The hypothalamic neuropeptide kisspeptin is a key component of the GnRH pulse generator, tightly regulating pulsatile GnRH secretion and the downstream reproductive axis. Here, we review the physiological regulation of pulsatile GnRH secretion by hypothalamic kisspeptin neurons and how their activity is modulated by signals of energy status to affect reproductive function. We explore endocrine factors contributing to the suppression of GnRH pulsatility in the pathophysiology of FHA and how hypothalamic kisspeptin neurons likely represent a final common pathway through which these factors affect GnRH pulse generation. Finally, we discuss the therapeutic potential of kisspeptin as a novel treatment for women with FHA.
Collapse
Affiliation(s)
- Aaran H Patel
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Chelsea and Westminster Hospital, London, UK
| | - Kanyada Koysombat
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Aureliane Pierret
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
| | - Megan Young
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
| | - Alexander N Comninos
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Ali Abbara
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
3
|
Leng D, Zeng B, Wang T, Chen BL, Li DY, Li ZJ. Single nucleus/cell RNA-seq of the chicken hypothalamic-pituitary-ovarian axis offers new insights into the molecular regulatory mechanisms of ovarian development. Zool Res 2024; 45:1088-1107. [PMID: 39245652 PMCID: PMC11491784 DOI: 10.24272/j.issn.2095-8137.2024.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 06/17/2024] [Indexed: 09/10/2024] Open
Abstract
The hypothalamic-pituitary-ovarian (HPO) axis represents a central neuroendocrine network essential for reproductive function. Despite its critical role, the intrinsic heterogeneity within the HPO axis across vertebrates and the complex intercellular interactions remain poorly defined. This study provides the first comprehensive, unbiased, cell type-specific molecular profiling of all three components of the HPO axis in adult Lohmann layers and Liangshan Yanying chickens. Within the hypothalamus, pituitary, and ovary, seven, 12, and 13 distinct cell types were identified, respectively. Results indicated that the pituitary adenylate cyclase activating polypeptide (PACAP), follicle-stimulating hormone (FSH), and prolactin (PRL) signaling pathways may modulate the synthesis and secretion of gonadotropin-releasing hormone (GnRH), FSH, and luteinizing hormone (LH) within the hypothalamus and pituitary. In the ovary, interactions between granulosa cells and oocytes involved the KIT, CD99, LIFR, FN1, and ANGPTL signaling pathways, which collectively regulate follicular maturation. The SEMA4 signaling pathway emerged as a critical mediator across all three tissues of the HPO axis. Additionally, gene expression analysis revealed that relaxin 3 (RLN3), gastrin-releasing peptide (GRP), and cocaine- and amphetamine regulated transcripts (CART, also known as CARTPT) may function as novel endocrine hormones, influencing the HPO axis through autocrine, paracrine, and endocrine pathways. Comparative analyses between Lohmann layers and Liangshan Yanying chickens demonstrated higher expression levels of GRP, RLN3, CARTPT, LHCGR, FSHR, and GRPR in the ovaries of Lohmann layers, potentially contributing to their superior reproductive performance. In conclusion, this study provides a detailed molecular characterization of the HPO axis, offering novel insights into the regulatory mechanisms underlying reproductive biology.
Collapse
Affiliation(s)
- Dong Leng
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Bo Zeng
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Tao Wang
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China
| | - Bin-Long Chen
- College of Animal Science, Xichang University, Xichang, Sichuan 615000, China. E-mail:
| | - Di-Yan Li
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China. E-mail:
| | - Zhuan-Jian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China. E-mail:
| |
Collapse
|
4
|
Anaya ES, de Groot EL, Lydon JP, Pangas SA, Hartig SM. Contributions of white adipose tissue to energy requirements for female reproduction. Trends Endocrinol Metab 2024; 35:809-820. [PMID: 38749883 PMCID: PMC11387141 DOI: 10.1016/j.tem.2024.04.012] [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: 02/14/2024] [Revised: 04/13/2024] [Accepted: 04/15/2024] [Indexed: 09/12/2024]
Abstract
Body composition impacts female fertility and there are established relationships between adipose tissue and the reproductive system. Maintaining functional adipose tissue is vital for meeting the energetic demands during the reproductive process, from ovulation to delivery and lactation. White adipose tissue (WAT) shows plastic responses to daily physiology and secretes diverse adipokines that affect the hypothalamic-pituitary-ovarian axis, but many other interorgan interactions remain to be determined. This review summarizes the current state of research on the dialogue between WAT and the female reproductive system, focusing on the impact of this crosstalk on ovarian and endometrial factors essential for fecundity.
Collapse
Affiliation(s)
- Elizabeth S Anaya
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Cancer and Cellular Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - Evelyn L de Groot
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Cancer and Cellular Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Stephanie A Pangas
- Cancer and Cellular Biology Program, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Sean M Hartig
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
5
|
Sáenz de Miera C, Bellefontaine N, Allen SJ, Myers MG, Elias CF. Glutamate neurotransmission from leptin receptor cells is required for typical puberty and reproductive function in female mice. eLife 2024; 13:RP93204. [PMID: 39007235 PMCID: PMC11249761 DOI: 10.7554/elife.93204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
Abstract
The hypothalamic ventral premammillary nucleus (PMv) is a glutamatergic nucleus essential for the metabolic control of reproduction. However, conditional deletion of leptin receptor long form (LepRb) in vesicular glutamate transporter 2 (Vglut2) expressing neurons results in virtually no reproductive deficits. In this study, we determined the role of glutamatergic neurotransmission from leptin responsive PMv neurons on puberty and fertility. We first assessed if stimulation of PMv neurons induces luteinizing hormone (LH) release in fed adult females. We used the stimulatory form of designer receptor exclusively activated by designer drugs (DREADDs) in LeprCre (LepRb-Cre) mice. We collected blood sequentially before and for 1 hr after intravenous clozapine-N-oxide injection. LH level increased in animals correctly targeted to the PMv, and LH level was correlated to the number of Fos immunoreactive neurons in the PMv. Next, females with deletion of Slc17a6 (Vglut2) in LepRb neurons (LeprΔVGlut2) showed delayed age of puberty, disrupted estrous cycles, increased gonadotropin-releasing hormone (GnRH) concentration in the axon terminals, and disrupted LH secretion, suggesting impaired GnRH release. To assess if glutamate is required for PMv actions in pubertal development, we generated a Cre-induced reexpression of endogenous LepRb (LeprloxTB) with concomitant deletion of Slc17a6 (Vglut2flox) mice. Rescue of Lepr and deletion of Slc17a6 in the PMv was obtained by stereotaxic injection of an adeno-associated virus vector expressing Cre recombinase. Control LeprloxTB mice with PMv LepRb rescue showed vaginal opening, follicle maturation, and became pregnant, while LeprloxTB;Vglut2flox mice showed no pubertal development. Our results indicate that glutamatergic neurotransmission from leptin sensitive neurons regulates the reproductive axis, and that leptin action on pubertal development via PMv neurons requires Vglut2.
Collapse
Affiliation(s)
- Cristina Sáenz de Miera
- Department of Molecular and Integrative Physiology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Nicole Bellefontaine
- Department of Molecular and Integrative Physiology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Susan J Allen
- Department of Molecular and Integrative Physiology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Martin G Myers
- Department of Molecular and Integrative Physiology, University of Michigan–Ann ArborAnn ArborUnited States
- Elizabeth W. Caswell Diabetes Institute, University of Michigan–Ann ArborAnn ArborUnited States
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan–Ann ArborAnn ArborUnited States
| | - Carol F Elias
- Department of Molecular and Integrative Physiology, University of Michigan–Ann ArborAnn ArborUnited States
- Elizabeth W. Caswell Diabetes Institute, University of Michigan–Ann ArborAnn ArborUnited States
- Department of Obstetrics and Gynecology, University of Michigan–Ann ArborAnn ArborUnited States
| |
Collapse
|
6
|
de Miera CS, Bellefontaine N, Allen SJ, Myers MG, Elias CF. Glutamate neurotransmission from leptin receptor cells is required for typical puberty and reproductive function in female mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.21.558865. [PMID: 37790549 PMCID: PMC10542178 DOI: 10.1101/2023.09.21.558865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
The hypothalamic ventral premammillary nucleus (PMv) is a glutamatergic nucleus essential for the metabolic control of reproduction. However, conditional deletion of leptin receptor (LepRb) in vesicular glutamate transporter 2 (Vglut2) expressing neurons results in virtually no reproductive deficits. In this study, we determine the role of glutamatergic signaling from leptin responsive PMv neurons on puberty and fertility. We first assessed if stimulation of PMv neurons induces LH release in fed adult females. We used the stimulatory form of designer receptor exclusively activated by designer drugs (DREADDs) in LepRb-Cre mice. We collected blood sequentially before and for 1h after iv. clozapine-N-oxide injection. LH level increased in animals correctly targeted to the PMv, and LH level was correlated to the number of cFos immunoreactive neurons in the PMv. Next, females with deletion of Vglut2 in LepRb neurons (LepR∆VGlut2) showed delayed age of puberty, disrupted estrous cycles, increased GnRH concentration in the axon terminals and disrupted LH responses, suggesting impaired GnRH release. To assess if glutamate is required for PMv actions in pubertal development, we generated a Cre-induced reexpression of endogenous LepRb (LepRloxTB) with concomitant deletion of Vglut2 (Vglut2-floxed) mice. Rescue of Lepr and deletion of Vglut2 in the PMv was obtained by stereotaxic injection of an adeno-associated virus vector expressing Cre recombinase. Control LepRloxTB mice with PMv LepRb rescue showed vaginal opening, follicle maturation and became pregnant, while LepRloxTB;Vglut2flox mice showed no pubertal development. Our results indicate that glutamatergic signaling from leptin sensitive neurons regulates the reproductive axis, and that leptin action on pubertal development via PMv neurons requires Vglut2.
Collapse
Affiliation(s)
- Cristina Sáenz de Miera
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109-5622, USA
| | - Nicole Bellefontaine
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109-5622, USA
| | - Susan J. Allen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109-5622, USA
| | - Martin G. Myers
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109-5622, USA
- Elizabeth W. Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI, 48109-5622, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, 48109-5622, USA
| | - Carol F. Elias
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109-5622, USA
- Elizabeth W. Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI, 48109-5622, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109-5622, USA
| |
Collapse
|
7
|
Anderson GM, Hill JW, Kaiser UB, Navarro VM, Ong KK, Perry JRB, Prevot V, Tena-Sempere M, Elias CF. Metabolic control of puberty: 60 years in the footsteps of Kennedy and Mitra's seminal work. Nat Rev Endocrinol 2024; 20:111-123. [PMID: 38049643 PMCID: PMC10843588 DOI: 10.1038/s41574-023-00919-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/19/2023] [Indexed: 12/06/2023]
Abstract
An individual's nutritional status has a powerful effect on sexual maturation. Puberty onset is delayed in response to chronic energy insufficiency and is advanced under energy abundance. The consequences of altered pubertal timing for human health are profound. Late puberty increases the chances of cardiometabolic, musculoskeletal and neurocognitive disorders, whereas early puberty is associated with increased risks of adult obesity, type 2 diabetes mellitus, cardiovascular diseases and various cancers, such as breast, endometrial and prostate cancer. Kennedy and Mitra's trailblazing studies, published in 1963 and using experimental models, were the first to demonstrate that nutrition is a key factor in puberty onset. Building on this work, the field has advanced substantially in the past decade, which is largely due to the impressive development of molecular tools for experimentation and population genetics. In this Review, we discuss the latest advances in basic and translational sciences underlying the nutritional and metabolic control of pubertal development, with a focus on perspectives and future directions.
Collapse
Affiliation(s)
- Greg M Anderson
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH, USA
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Victor M Navarro
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken K Ong
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - John R B Perry
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Vincent Prevot
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain.
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain.
| | - Carol F Elias
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
- Department of Obstetrics & Gynecology, University of Michigan, Ann Arbor, MI, USA.
- Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Lepeak L, Miracle S, Ferragud A, Seiglie MP, Shafique S, Ozturk Z, Minnig MA, Medeiros G, Cottone P, Sabino V. Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) of the Bed Nucleus of the Stria Terminalis Mediates Heavy Alcohol Drinking in Mice. eNeuro 2023; 10:ENEURO.0424-23.2023. [PMID: 38053471 PMCID: PMC10755645 DOI: 10.1523/eneuro.0424-23.2023] [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: 10/19/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 12/07/2023] Open
Abstract
Alcohol use disorder (AUD) is a complex psychiatric disease characterized by periods of heavy drinking and periods of withdrawal. Chronic exposure to ethanol causes profound neuroadaptations in the extended amygdala, which cause allostatic changes promoting excessive drinking. The bed nucleus of the stria terminalis (BNST), a brain region involved in both excessive drinking and anxiety-like behavior, shows particularly high levels of pituitary adenylate cyclase-activating polypeptide (PACAP), a key mediator of the stress response. Recently, a role for PACAP in withdrawal-induced alcohol drinking and anxiety-like behavior in alcohol-dependent rats has been proposed; whether the PACAP system of the BNST is also recruited in other models of alcohol addiction and whether it is of local or nonlocal origin is currently unknown. Here, we show that PACAP immunoreactivity is increased selectively in the BNST of C57BL/6J mice exposed to a chronic, intermittent access to ethanol. While pituitary adenylate cyclase-activating polypeptide (PACAP) type 1 receptor-expressing cells were unchanged by chronic alcohol, the levels of a peptide closely related to PACAP, the calcitonin gene-related neuropeptide, were found to also be increased in the BNST. Finally, using a retrograde chemogenetic approach in PACAP-ires-Cre mice, we found that the inhibition of PACAP neuronal afferents to the BNST reduced heavy ethanol drinking. Our data suggest that the PACAP system of the BNST is recruited by chronic, voluntary alcohol drinking in mice and that nonlocally originating PACAP projections to the BNST regulate heavy alcohol intake, indicating that this system may represent a promising target for novel AUD therapies.
Collapse
Affiliation(s)
| | | | - Antonio Ferragud
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University Chobanian & Avedisian, School of Medicine, Boston, Massachusetts 02118
| | - Mariel P. Seiglie
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University Chobanian & Avedisian, School of Medicine, Boston, Massachusetts 02118
| | - Samih Shafique
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University Chobanian & Avedisian, School of Medicine, Boston, Massachusetts 02118
| | - Zeynep Ozturk
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University Chobanian & Avedisian, School of Medicine, Boston, Massachusetts 02118
| | - Margaret A. Minnig
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University Chobanian & Avedisian, School of Medicine, Boston, Massachusetts 02118
| | - Gianna Medeiros
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University Chobanian & Avedisian, School of Medicine, Boston, Massachusetts 02118
| | | | | |
Collapse
|
9
|
Mei L, Osakada T, Lin D. Hypothalamic control of innate social behaviors. Science 2023; 382:399-404. [PMID: 37883550 PMCID: PMC11105421 DOI: 10.1126/science.adh8489] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023]
Abstract
Sexual, parental, and aggressive behaviors are central to the reproductive success of individuals and species survival and thus are supported by hardwired neural circuits. The reproductive behavior control column (RBCC), which comprises the medial preoptic nucleus (MPN), the ventrolateral part of the ventromedial hypothalamus (VMHvl), and the ventral premammillary nucleus (PMv), is essential for all social behaviors. The RBCC integrates diverse hormonal and metabolic cues and adjusts an animal's physical activity, hence the chance of social encounters. The RBCC further engages the mesolimbic dopamine system to maintain social interest and reinforces cues and actions that are time-locked with social behaviors. We propose that the RBCC and brainstem form a dual-control system for generating moment-to-moment social actions. This Review summarizes recent progress regarding the identities of RBCC cells and their pathways that drive different aspects of social behaviors.
Collapse
Affiliation(s)
- Long Mei
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Takuya Osakada
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Dayu Lin
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
- Department of Psychiatry, New York University Langone Medical Center, New York, NY 10016, USA
- Department of Neuroscience and Physiology, New York University Langone Medical Center, New York, NY 10016, USA
- Center for Neural Science, New York University, New York, NY 10016, USA
| |
Collapse
|
10
|
Mansano NDS, Vieira HR, Araujo-Lopes R, Szawka RE, Donato J, Frazao R. Fasting Modulates GABAergic Synaptic Transmission to Arcuate Kisspeptin Neurons in Female Mice. Endocrinology 2023; 164:bqad150. [PMID: 37793082 DOI: 10.1210/endocr/bqad150] [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: 06/20/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/06/2023]
Abstract
It is well-established that the hypothalamic-pituitary-gonadal (HPG) axis is suppressed due to negative energy balance. However, less information is available on whether kisspeptin neuronal activity contributes to fasting-induced responses. In the present study, female and male mice were fasted for 24 hours or provided food ad libitum (fed group) to determine whether acute fasting is sufficient to modulate kisspeptin neuronal activity. In female mice, fasting attenuated luteinizing hormone (LH) and prolactin (PRL) serum levels and increased follicle-stimulating hormone levels compared with the fed group. In contrast, fasting did not affect gonadotropin or PRL secretion in male mice. By measuring genes related to LH pulse generation in micropunches obtained from the arcuate nucleus of the hypothalamus (ARH), we observed that fasting reduced Kiss1 mRNA levels in female and male mice. In contrast, Pdyn expression was upregulated only in fasted female mice, whereas no changes in the Tac2 mRNA levels were observed in both sexes. Interestingly, the frequency and amplitude of the GABAergic postsynaptic currents recorded from ARH kisspeptin neurons (ARHKisspeptin) were reduced in 24-hour fasted female mice but not in males. Additionally, neuropeptide Y induced a hyperpolarization in the resting membrane potential of ARHKisspeptin neurons of fed female mice but not in males. Thus, the response of ARHKisspeptin neurons to fasting is sexually dependent with a female bias, associated with changes in gonadotropins and PRL secretion. Our findings suggest that GABAergic transmission to ARHKisspeptin neurons modulates the activity of the HPG axis during situations of negative energy balance.
Collapse
Affiliation(s)
- Naira da Silva Mansano
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Anatomia, São Paulo, SP 05508-000, Brazil
| | - Henrique Rodrigues Vieira
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Anatomia, São Paulo, SP 05508-000, Brazil
| | - Roberta Araujo-Lopes
- Universidade Federal de Minas Gerais, Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Belo Horizonte, MG 31270-901, Brazil
| | - Raphael Escorsim Szawka
- Universidade Federal de Minas Gerais, Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Belo Horizonte, MG 31270-901, Brazil
| | - Jose Donato
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, São Paulo, SP 05508-000, Brazil
| | - Renata Frazao
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Anatomia, São Paulo, SP 05508-000, Brazil
| |
Collapse
|
11
|
Yin C, Wang Y, Zhou P, Shi H, Ma X, Yin Z, Liu Y. Genomic Scan for Runs of Homozygosity and Selective Signature Analysis to Identify Candidate Genes in Large White Pigs. Int J Mol Sci 2023; 24:12914. [PMID: 37629094 PMCID: PMC10454931 DOI: 10.3390/ijms241612914] [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: 06/28/2023] [Revised: 08/09/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Large White pigs are extensively utilized in China for their remarkable characteristics of rapid growth and the high proportion of lean meat. The economic traits of pigs, comprising reproductive and meat quality traits, play a vital role in swine production. In this study, 2295 individuals, representing three different genetic backgrounds Large White pig populations were used: 500 from the Canadian line, 295 from the Danish line, and 1500 from the American line. The GeneSeek 50K GGP porcine HD array was employed to genotype the three pig populations. Firstly, genomic selective signature regions were identified using the pairwise fixation index (FST) and locus-specific branch length (LSBL). By applying a top 1% threshold for both parameters, a total of 888 candidate selective windows were identified, harbouring 1571 genes. Secondly, the investigation of regions of homozygosity (ROH) was performed utilizing the PLINK software. In total, 25 genomic regions exhibiting a high frequency of ROHs were detected, leading to the identification of 1216 genes. Finally, the identified potential functional genes from candidate genomic regions were annotated, and several important candidate genes associated with reproductive traits (ADCYAP1, U2, U6, CETN1, Thoc1, Usp14, GREB1L, FGF12) and meat quality traits (MiR-133, PLEKHO1, LPIN2, SHANK2, FLVCR1, MYL4, SFRP1, miR-486, MYH3, STYX) were identified. The findings of this study provide valuable insights into the genetic basis of economic traits in Large White pigs and may have potential use in future pig breeding programs.
Collapse
Affiliation(s)
- Chang Yin
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (C.Y.); (Y.W.); (P.Z.); (H.S.); (X.M.)
| | - Yuwei Wang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (C.Y.); (Y.W.); (P.Z.); (H.S.); (X.M.)
| | - Peng Zhou
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (C.Y.); (Y.W.); (P.Z.); (H.S.); (X.M.)
| | - Haoran Shi
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (C.Y.); (Y.W.); (P.Z.); (H.S.); (X.M.)
| | - Xinyu Ma
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (C.Y.); (Y.W.); (P.Z.); (H.S.); (X.M.)
| | - Zongjun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China;
| | - Yang Liu
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (C.Y.); (Y.W.); (P.Z.); (H.S.); (X.M.)
| |
Collapse
|
12
|
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
|
13
|
Ruiz-Cruz M, Torres-Granados C, Tena-Sempere M, Roa J. Central and peripheral mechanisms involved in the control of GnRH neuronal function by metabolic factors. Curr Opin Pharmacol 2023; 71:102382. [PMID: 37307655 DOI: 10.1016/j.coph.2023.102382] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 06/14/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons are the final output pathway for the brain control of reproduction. The activity of this neuronal population, mainly located at the preoptic area of the hypothalamus, is controlled by a plethora of metabolic signals. However, it has been documented that most of these signal impact on GnRH neurons through indirect neuronal circuits, Kiss1, proopiomelanocortin, and neuropeptide Y/agouti-related peptide neurons being some of the most prominent mediators. In this context, compelling evidence has been gathered in recent years on the role of a large range of neuropeptides and energy sensors in the regulation of GnRH neuronal activity through both direct and indirect mechanisms. The present review summarizes some of the most prominent recent advances in our understanding of the peripheral factors and central mechanisms involved in the metabolic control of GnRH neurons.
Collapse
Affiliation(s)
- Miguel Ruiz-Cruz
- Instituto Maimónides de Investigación Biomédica de Córdoba, Department of Cell Biology, Physiology and Immunology, University of Córdoba; Hospital Universitario Reina Sofia (IMIBIC/HURS), 14004 Córdoba, Spain
| | - Carmen Torres-Granados
- Instituto Maimónides de Investigación Biomédica de Córdoba, Department of Cell Biology, Physiology and Immunology, University of Córdoba; Hospital Universitario Reina Sofia (IMIBIC/HURS), 14004 Córdoba, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba, Department of Cell Biology, Physiology and Immunology, University of Córdoba; Hospital Universitario Reina Sofia (IMIBIC/HURS), 14004 Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - Juan Roa
- Instituto Maimónides de Investigación Biomédica de Córdoba, Department of Cell Biology, Physiology and Immunology, University of Córdoba; Hospital Universitario Reina Sofia (IMIBIC/HURS), 14004 Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004 Córdoba, Spain.
| |
Collapse
|
14
|
Abstract
Reproduction is the biological process by which new individuals are produced by their parents. It is the fundamental feature of all known life and is required for the existence of all species. All mammals reproduce sexually, a process that involves the union of two reproductive cells, one from a male and one from a female. Sexual behaviors are a series of actions leading to reproduction. They are composed of appetitive, action, and refractory phases, each supported by dedicated developmentally-wired neural circuits to ensure high reproduction success. In rodents, successful reproduction can only occur during female ovulation. Thus, female sexual behavior is tightly coupled with ovarian activity, namely the estrous cycle. This is achieved through the close interaction between the female sexual behavior circuit and the hypothalamic-pituitary-gonadal (HPG) axis. In this review, we will summarize our current understanding, learned mainly in rodents, regarding the neural circuits underlying each phase of the female sexual behaviors and their interaction with the HPG axis, highlighting the gaps in our knowledge that require future investigation.
Collapse
Affiliation(s)
- Luping Yin
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Dayu Lin
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA; Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA.
| |
Collapse
|
15
|
Argente J, Dunkel L, Kaiser UB, Latronico AC, Lomniczi A, Soriano-Guillén L, Tena-Sempere M. Molecular basis of normal and pathological puberty: from basic mechanisms to clinical implications. Lancet Diabetes Endocrinol 2023; 11:203-216. [PMID: 36620967 PMCID: PMC10198266 DOI: 10.1016/s2213-8587(22)00339-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 01/07/2023]
Abstract
Puberty is a major maturational event; its mechanisms and timing are driven by genetic determinants, but also controlled by endogenous and environmental cues. Substantial progress towards elucidation of the neuroendocrine networks governing puberty has taken place. However, key aspects of the mechanisms responsible for the precise timing of puberty and its alterations have only recently begun to be deciphered, propelled by epidemiological data suggesting that pubertal timing is changing in humans, via mechanisms that are not yet understood. By integrating basic and clinical data, we provide a comprehensive overview of current advances on the physiological basis of puberty, with a particular focus on the roles of kisspeptins and other central transmitters, the underlying molecular and endocrine mechanisms, and the pathways involved in pubertal modulation by nutritional and metabolic cues. Additionally, we have summarised molecular features of precocious and delayed puberty in both sexes, as revealed by clinical and genetic studies. This Review is a synoptic up-to-date view of how puberty is controlled and of the pathogenesis of major pubertal alterations, from both a clinical and translational perspective. We also highlight unsolved challenges that will seemingly concentrate future research efforts in this active domain of endocrinology.
Collapse
Affiliation(s)
- Jesús Argente
- Department of Pediatrics & Pediatric Endocrinology, Universidad Autónoma de Madrid, University Hospital Niño Jesús, Instituto de Investigación Sanitaria La Princesa, Madrid, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; IMDEA Food Institute, Madrid, Spain.
| | - Leo Dunkel
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London Medical School, London, UK
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ana C Latronico
- Developmental Endocrinology Unit, Laboratory of Hormones and Molecular Genetics, LIM42, Department of Endocrinology and Metabolism, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Alejandro Lomniczi
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Leandro Soriano-Guillén
- Service of Pediatrics, University Hospital Fundación Jiménez Díaz, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Tena-Sempere
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain; Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofia, Córdoba, Spain; Institute of Biomedicine, University of Turku, Turku, Finland.
| |
Collapse
|
16
|
Evans MC, Campbell RE, Anderson GM. Physiological regulation of leptin as an integrative signal of reproductive readiness. Curr Opin Pharmacol 2022; 67:102321. [PMID: 36427399 DOI: 10.1016/j.coph.2022.102321] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 10/24/2022] [Indexed: 11/23/2022]
Abstract
Reproductive function is tightly regulated by both environmental and physiological factors. The adipose-derived hormone leptin has been identified as one such critical factor that relays information about peripheral energy availability to the centrally-governed HPG axis to ensure there is sufficient energy availability to support the high energy demands of mammalian reproduction. In the absence of adequate central leptin signaling, reproductive function is suppressed. While leptin levels are predominantly regulated by adiposity, circulating leptin levels are also under the modulatory influence of other factors, such as stress system activation, circadian rhythmicity, and immune activation and the inflammatory response. Furthermore, changes in leptin sensitivity can affect the degree to which leptin exerts its influence on the neuroendocrine reproductive axis. This review will discuss the different mechanisms by which leptin serves to integrate and relay information about metabolic, psychological, environmental and immune conditions to the central neuronal network that governs reproductive function.
Collapse
Affiliation(s)
- Maggie C Evans
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Greg M Anderson
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand.
| |
Collapse
|
17
|
Hajdarovic KH, Yu D, Webb AE. Understanding the aging hypothalamus, one cell at a time. Trends Neurosci 2022; 45:942-954. [PMID: 36272823 PMCID: PMC9671837 DOI: 10.1016/j.tins.2022.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/21/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022]
Abstract
The hypothalamus is a brain region that integrates signals from the periphery and the environment to maintain organismal homeostasis. To do so, specialized hypothalamic neuropeptidergic neurons control a range of processes, such as sleep, feeding, the stress response, and hormone release. These processes are altered with age, which can affect longevity and contribute to disease status. Technological advances, such as single-cell RNA sequencing, are upending assumptions about the transcriptional identity of cell types in the hypothalamus and revealing how distinct cell types change with age. In this review, we summarize current knowledge about the contribution of hypothalamic functions to aging. We highlight recent single-cell studies interrogating distinct cell types of the mouse hypothalamus and suggest ways in which single-cell 'omics technologies can be used to further understand the aging hypothalamus and its role in longevity.
Collapse
Affiliation(s)
| | - Doudou Yu
- Graduate program in Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Ashley E Webb
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA; Center on the Biology of Aging, Brown University, Providence, RI 02912, USA; Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA; Center for Translational Neuroscience, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
18
|
Patko E, Szabo E, Toth D, Tornoczky T, Bosnyak I, Vaczy A, Atlasz T, Reglodi D. Distribution of PACAP and PAC1 Receptor in the Human Eye. J Mol Neurosci 2022; 72:2176-2187. [PMID: 35253081 PMCID: PMC9726800 DOI: 10.1007/s12031-022-01985-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 02/08/2022] [Indexed: 12/16/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide with widespread distribution and diverse biological functions. Several studies show that PACAP has strong cytoprotective effects mediated mostly through its specific PAC1 receptor (PAC1-R) and it plays important roles in several pathological conditions. Its distribution and altered expression are known in various human tissues, but there is no descriptive data about PACAP and its receptors in the human eyebulb. Since PACAP38 is the dominant form of the naturally occurring PACAP, our aim was to investigate the distribution of PACAP38-like immunoreactivity in the human eye and to describe the presence of PAC1-R. Semiquantitative evaluation was performed after routine histology and immunohistochemical labeling on human eye sections. Our results showed high level of immunopositivity in the corneal epithelium and endothelium. Within the vascular layer, the iris and the ciliary body had strong immunopositivity for both PACAP and PAC1-R. Several layers of the retina showed immunoreactivity for PACAP and PAC1-R, but the ganglion cell layer had a special pattern in the immunolabeling. Labeling was observed in the neuropil within the optic nerve in both cases and glial cells displayed immunoreactivity for PAC1-R. In summary, our study indicates the widespread occurrence of PACAP and its specific receptor in the human eye, implying that the results from in vitro and animal studies have translational value and most probably are also present in the human eye.
Collapse
Affiliation(s)
- Evelin Patko
- Department of Anatomy, Medical School, MTA-PTE PACAP Research Team, University of Pecs, 7624, Pecs, Hungary
| | - Edina Szabo
- Department of Anatomy, Medical School, MTA-PTE PACAP Research Team, University of Pecs, 7624, Pecs, Hungary
| | - Denes Toth
- Department of Forensic Medicine, Medical School, University of Pecs, 7624, Pecs, Hungary
| | - Tamas Tornoczky
- Department of Pathology, Medical School and Clinical Center, University of Pecs, 7624, Pecs, Hungary
| | - Inez Bosnyak
- Department of Anatomy, Medical School, MTA-PTE PACAP Research Team, University of Pecs, 7624, Pecs, Hungary
| | - Alexandra Vaczy
- Department of Anatomy, Medical School, MTA-PTE PACAP Research Team, University of Pecs, 7624, Pecs, Hungary
| | - Tamas Atlasz
- Department of Anatomy, Medical School, MTA-PTE PACAP Research Team, University of Pecs, 7624, Pecs, Hungary.
- Szentagothai Research Center, Medical School, University of Pecs, 7624, Pecs, Hungary.
- Department of Sportbiology, University of Pecs, 7624, Pecs, Hungary.
| | - Dora Reglodi
- Department of Anatomy, Medical School, MTA-PTE PACAP Research Team, University of Pecs, 7624, Pecs, Hungary
- Szentagothai Research Center, Medical School, University of Pecs, 7624, Pecs, Hungary
| |
Collapse
|
19
|
Gao F, Ma J, Yu YQ, Gao XF, Bai Y, Sun Y, Liu J, Liu X, Barry DM, Wilhelm S, Piccinni-Ash T, Wang N, Liu D, Ross RA, Hao Y, Huang X, Jia JJ, Yang Q, Zheng H, van Nispen J, Chen J, Li H, Zhang J, Li YQ, Chen ZF. A non-canonical retina-ipRGCs-SCN-PVT visual pathway for mediating contagious itch behavior. Cell Rep 2022; 41:111444. [PMID: 36198265 PMCID: PMC9595067 DOI: 10.1016/j.celrep.2022.111444] [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: 10/22/2021] [Revised: 08/10/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022] Open
Abstract
Contagious itch behavior informs conspecifics of adverse environment and is crucial for the survival of social animals. Gastrin-releasing peptide (GRP) and its receptor (GRPR) in the suprachiasmatic nucleus (SCN) of the hypothalamus mediates contagious itch behavior in mice. Here, we show that intrinsically photosensitive retina ganglion cells (ipRGCs) convey visual itch information, independently of melanopsin, from the retina to GRP neurons via PACAP-PAC1R signaling. Moreover, GRPR neurons relay itch information to the paraventricular nucleus of the thalamus (PVT). Surprisingly, neither the visual cortex nor superior colliculus is involved in contagious itch. In vivo calcium imaging and extracellular recordings reveal contagious itch-specific neural dynamics of GRPR neurons. Thus, we propose that the retina-ipRGC-SCN-PVT pathway constitutes a previously unknown visual pathway that probably evolved for motion vision that encodes salient environmental cues and enables animals to imitate behaviors of conspecifics as an anticipatory mechanism to cope with adverse conditions. It has been shown that GRP-GRPR neuropeptide signaling in the SCN is important for contagious itch behavior in mice. Gao et al. find that SCN-projecting ipRGCs are sufficient to relay itch information from the retina to the SCN by releasing neuropeptide PACAP to activate the GRP-GRPR pathway.
Collapse
Affiliation(s)
- Fang Gao
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jun Ma
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yao-Qing Yu
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Institute for Biomedical Sciences of Pain, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, P. R. China
| | - Xiao-Fei Gao
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Present address: Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, P. R. China
| | - Yang Bai
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Department of Anatomy, Histology and Embryology & K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, P. R. China,Present address: Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang 110016, P. R. China
| | - Yi Sun
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Department of Anatomy, Histology and Embryology & K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, P. R. China,Present address: Binzhou Medical University, Yantai 264003, P. R. China
| | - Juan Liu
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xianyu Liu
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Devin M. Barry
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven Wilhelm
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tyler Piccinni-Ash
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Na Wang
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Present address: Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, P. R. China
| | - Dongyang Liu
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Department of Pain Management, the State Key Clinical Specialty in Pain Medicine, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, P.R. China
| | - Rachel A. Ross
- Department of Neuroscience, Psychiatry and Medicine, Albert Einstein College of Medicine Rose F. Kennedy Center, Bronx, NY, USA
| | - Yan Hao
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Present address: Department of Pediatrics, Tongji Hospital, Tongji Medical College, HuaZhong University of Science and Technology, Wuhan 430030, P. R. China
| | - Xu Huang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institute for Medical and Engineering Innovation, Eye & ENT Hospital, Fudan University, Shanghai 200031, P.R. China
| | - Jin-Jing Jia
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Present address: College of Life Sciences, Xinyang Normal University, Xinyang 464000, P. R. China
| | - Qianyi Yang
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hao Zheng
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institute for Medical and Engineering Innovation, Eye & ENT Hospital, Fudan University, Shanghai 200031, P.R. China
| | - Johan van Nispen
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Present address: Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Jun Chen
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, P. R. China
| | - Hui Li
- Department of Anatomy, Histology and Embryology & K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, P. R. China
| | - Jiayi Zhang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institute for Medical and Engineering Innovation, Eye & ENT Hospital, Fudan University, Shanghai 200031, P.R. China
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology & K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, P. R. China
| | - Zhou-Feng Chen
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Lead contact,Correspondence:
| |
Collapse
|
20
|
Saengkaew T, Howard SR. Genetics of pubertal delay. Clin Endocrinol (Oxf) 2022; 97:473-482. [PMID: 34617615 PMCID: PMC9543006 DOI: 10.1111/cen.14606] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 12/23/2022]
Abstract
The timing of pubertal development is strongly influenced by the genetic background, and clinical presentations of delayed puberty are often found within families with clear patterns of inheritance. The discovery of the underlying genetic regulators of such conditions, in recent years through next generation sequencing, has advanced the understanding of the pathogenesis of disorders of pubertal timing and the potential for genetic testing to assist diagnosis for patients with these conditions. This review covers the significant advances in the understanding of the biological mechanisms of delayed puberty that have occurred in the last two decades.
Collapse
Affiliation(s)
- Tansit Saengkaew
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Endocrinology Unit, Department of Paediatrics, Faculty of MedicinePrince of Songkla UniversitySongkhlaThailand
| | - Sasha R. Howard
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
21
|
Koppan M, Nagy Z, Bosnyak I, Reglodi D. Female reproductive functions of the neuropeptide PACAP. Front Endocrinol (Lausanne) 2022; 13:982551. [PMID: 36204113 PMCID: PMC9531758 DOI: 10.3389/fendo.2022.982551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/31/2022] [Indexed: 12/28/2022] Open
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuropeptide originally isolated as a hypothalamic peptide. It has a widespread distribution in the body and has a diverse spectrum of actions. Among other processes, PACAP has been shown to be involved in reproduction. In this review we summarize findings related to the entire spectrum of female reproduction. PACAP is a regulatory factor in gonadal hormone production, influences follicular development and plays a role in fertilization and embryonic/placental development. Furthermore, PACAP is involved in hormonal changes during and after birth and affects maternal behavior. Although most data come from cell cultures and animal experiments, increasing number of evidence suggests that similar effects of PACAP can be found in humans. Among other instances, PACAP levels show changes in the serum during pregnancy and birth. PACAP is also present in the human follicular and amniotic fluids and in the milk. Levels of PACAP in follicular fluid correlate with the number of retrieved oocytes in hyperstimulated women. Human milk contains very high levels of PACAP compared to plasma levels, with colostrum showing the highest concentration, remaining steady thereafter for the first 7 months of lactation. All these data imply that PACAP has important functions in reproduction both under physiological and pathological conditions.
Collapse
Affiliation(s)
| | - Zsuzsanna Nagy
- Department of Physiology, University of Pecs Medical School, Pécs, Hungary
| | - Inez Bosnyak
- Department of Anatomy, ELKH-PTE PACAP Research Group and Szentagothai Research Center, University of Pecs Medical School, Pécs, Hungary
| | - Dora Reglodi
- Department of Anatomy, ELKH-PTE PACAP Research Group and Szentagothai Research Center, University of Pecs Medical School, Pécs, Hungary
| |
Collapse
|
22
|
Socs3 ablation in kisspeptin cells partially prevents lipopolysaccharide-induced body weight loss. Cytokine 2022; 158:155999. [PMID: 35985175 DOI: 10.1016/j.cyto.2022.155999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/25/2022] [Accepted: 08/02/2022] [Indexed: 11/22/2022]
Abstract
Many cytokines have been proposed to regulate reproduction due to their actions on hypothalamic kisspeptin cells, the main modulators of gonadotropin-releasing hormone (GnRH) neurons. Hormones such as leptin, prolactin and growth hormone are good examples of cytokines that lead to Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway activation, consequently exerting effects in kisspeptin neurons. Different studies have investigated how specific components of the JAK/STAT signaling pathway affect the functions of kisspeptin cells, but the role of the suppressor of cytokine signaling 3 (SOCS3) in mediating cytokine actions in kisspeptin cells remains unknown. Cre-Loxp technology was used in the present study to ablate Socs3 expression in kisspeptin cells (Kiss1/Socs3-KO). Then, male and female control and Kiss1/Socs3-KO mice were evaluated for sexual maturation, energy homeostasis features, and fertility. It was found that hypothalamic Kiss1 mRNA expression is significantly downregulated in Kiss1/Socs3-KO mice. Despite reduced hypothalamic Kiss1 mRNA content, these mice did not present any sexual maturation or fertility impairments. Additionally, body weight gain, leptin sensitivity and glucose homeostasis were similar to control mice. Interestingly, Kiss1/Socs3-KO mice were partially protected against lipopolysaccharide (LPS)-induced body weight loss. Our results suggest that Socs3 ablation in kisspeptin cells partially prevents the sickness behavior induced by LPS, suggesting that kisspeptin cells can modulate energy metabolism in mice in certain situations.
Collapse
|
23
|
Velasco ER, Florido A, Flores Á, Senabre E, Gomez-Gomez A, Torres A, Roca A, Norrholm S, Newman EL, Das P, Ross RA, Lori A, Pozo OJ, Ressler KJ, Garcia-Esteve LL, Jovanovic T, Andero R. PACAP-PAC1R modulates fear extinction via the ventromedial hypothalamus. Nat Commun 2022; 13:4374. [PMID: 35902577 PMCID: PMC9334354 DOI: 10.1038/s41467-022-31442-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/16/2022] [Indexed: 12/14/2022] Open
Abstract
Exposure to traumatic stress can lead to fear dysregulation, which has been associated with posttraumatic stress disorder (PTSD). Previous work showed that a polymorphism in the PACAP-PAC1R (pituitary adenylate cyclase-activating polypeptide) system is associated with PTSD risk in women, and PACAP (ADCYAP1)-PAC1R (ADCYAP1R1) are highly expressed in the hypothalamus. Here, we show that female mice subjected to acute stress immobilization (IMO) have fear extinction impairments related to Adcyap1 and Adcyap1r1 mRNA upregulation in the hypothalamus, PACAP-c-Fos downregulation in the Medial Amygdala (MeA), and PACAP-FosB/ΔFosB upregulation in the Ventromedial Hypothalamus dorsomedial part (VMHdm). DREADD-mediated inhibition of MeA neurons projecting to the VMHdm during IMO rescues both PACAP upregulation in VMHdm and the fear extinction impairment. We also found that women with the risk genotype of ADCYAP1R1 rs2267735 polymorphism have impaired fear extinction.
Collapse
Affiliation(s)
- E R Velasco
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - A Florido
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Á Flores
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - E Senabre
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, University Pompeu Fabra, Barcelona, Spain
| | - A Gomez-Gomez
- Integrative Pharmacology and Systems Neuroscience Research Group, Neurosciences Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - A Torres
- Perinatal Mental health Unit, Department of Psychiatry and Clinical Psychology, Institute of Neuroscience, Hospital Clínic, IDIBAPS, Barcelona, Spain
- Programme for the Prevention and Treatment of Psychic Effects in Sexually Assaulted Women. Hospital Clínic de Barcelona, Barcelona, Spain
| | - A Roca
- Perinatal Mental health Unit, Department of Psychiatry and Clinical Psychology, Institute of Neuroscience, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - S Norrholm
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University, Detroit, MI, USA
| | - E L Newman
- McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
| | - P Das
- Department of Neuroscience, Albert Einstein College of Medicine, Psychiatry Research Institute of Montefiore and Einstein, New York, NY, USA
| | - R A Ross
- Department of Neuroscience, Albert Einstein College of Medicine, Psychiatry Research Institute of Montefiore and Einstein, New York, NY, USA
| | - A Lori
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
- American Cancer Society, Inc., Atlanta, GA, USA
| | - O J Pozo
- Integrative Pharmacology and Systems Neuroscience Research Group, Neurosciences Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - K J Ressler
- McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
| | - L L Garcia-Esteve
- Perinatal Mental health Unit, Department of Psychiatry and Clinical Psychology, Institute of Neuroscience, Hospital Clínic, IDIBAPS, Barcelona, Spain
- Programme for the Prevention and Treatment of Psychic Effects in Sexually Assaulted Women. Hospital Clínic de Barcelona, Barcelona, Spain
| | - T Jovanovic
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University, Detroit, MI, USA
| | - R Andero
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
- Centro de Investigación Biomédica En Red en Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain.
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.
- ICREA, Barcelona, Spain.
| |
Collapse
|
24
|
Jamieson BB, Piet R. Kisspeptin neuron electrophysiology: Intrinsic properties, hormonal modulation, and regulation of homeostatic circuits. Front Neuroendocrinol 2022; 66:101006. [PMID: 35640722 DOI: 10.1016/j.yfrne.2022.101006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 05/05/2022] [Accepted: 05/19/2022] [Indexed: 11/04/2022]
Abstract
The obligatory role of kisspeptin (KISS1) and its receptor (KISS1R) in regulating the hypothalamic-pituitary-gonadal axis, puberty and fertility was uncovered in 2003. In the few years that followed, an impressive body of work undertaken in many species established that neurons producing kisspeptin orchestrate gonadotropin-releasing hormone (GnRH) neuron activity and subsequent GnRH and gonadotropin hormone secretory patterns, through kisspeptin-KISS1R signaling, and mediate many aspects of gonadal steroid hormone feedback regulation of GnRH neurons. Here, we review knowledge accrued over the past decade, mainly in genetically modified mouse models, of the electrophysiological properties of kisspeptin neurons and their regulation by hormonal feedback. We also discuss recent progress in our understanding of the role of these cells within neuronal circuits that control GnRH neuron activity and GnRH secretion, energy balance and, potentially, other homeostatic and reproductive functions.
Collapse
Affiliation(s)
| | - Richard Piet
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, USA.
| |
Collapse
|
25
|
Sobrino V, Avendaño MS, Perdices-López C, Jimenez-Puyer M, Tena-Sempere M. Kisspeptins and the neuroendocrine control of reproduction: Recent progress and new frontiers in kisspeptin research. Front Neuroendocrinol 2022; 65:100977. [PMID: 34999056 DOI: 10.1016/j.yfrne.2021.100977] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 12/31/2022]
Abstract
In late 2003, a major breakthrough in our understanding of the mechanisms that govern reproduction occurred with the identification of the reproductive roles of kisspeptins, encoded by the Kiss1 gene, and their receptor, Gpr54 (aka, Kiss1R). The discovery of this unsuspected reproductive facet attracted an extraordinary interest and boosted an intense research activity, in human and model species, that, in a relatively short period, established a series of basic concepts on the physiological roles of kisspeptins. Such fundamental knowledge, gathered in these early years of kisspeptin research, set the scene for the more recent in-depth dissection of the intimacies of the neuronal networks involving Kiss1 neurons, their precise mechanisms of regulation and the molecular underpinnings of the function of kisspeptins as pivotal regulators of all key aspects of reproductive function, from puberty onset to pulsatile gonadotropin secretion and the metabolic control of fertility. While no clear temporal boundaries between these two periods can be defined, in this review we will summarize the most prominent advances in kisspeptin research occurred in the last ten years, as a means to provide an up-dated view of the state of the art and potential paths of future progress in this dynamic, and ever growing domain of Neuroendocrinology.
Collapse
Affiliation(s)
- Veronica Sobrino
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Hospital Universitario Reina Sofia, 14004 Cordoba, Spain
| | - Maria Soledad Avendaño
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Hospital Universitario Reina Sofia, 14004 Cordoba, Spain
| | - Cecilia Perdices-López
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Hospital Universitario Reina Sofia, 14004 Cordoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004 Cordoba, Spain
| | - Manuel Jimenez-Puyer
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Hospital Universitario Reina Sofia, 14004 Cordoba, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Hospital Universitario Reina Sofia, 14004 Cordoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004 Cordoba, Spain; Institute of Biomedicine, University of Turku, FIN-20520 Turku, Finland.
| |
Collapse
|
26
|
Mansano NDS, Paradela RS, Bohlen TM, Zanardi IM, Chaves FM, Silveira MA, Tavares MR, Donato J, Frazao R. Vasoactive intestinal peptide exerts an excitatory effect on hypothalamic kisspeptin neurons during estrogen negative feedback. Mol Cell Endocrinol 2022; 542:111532. [PMID: 34915098 DOI: 10.1016/j.mce.2021.111532] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/30/2022]
Abstract
Hypothalamic kisspeptin neurons are the primary modulators of gonadotropin-releasing hormone (GnRH) neurons. It has been shown that circadian rhythms driven by the suprachiasmatic nucleus (SCN) contribute to GnRH secretion. Kisspeptin neurons are potential targets of SCN neurons due to reciprocal connections with the anteroventral periventricular and rostral periventricular nuclei (AVPV/PeN) and the arcuate nucleus of the hypothalamus (ARH). Vasoactive intestinal peptide (VIP), a notable SCN neurotransmitter, modulates GnRH secretion depending on serum estradiol levels, aging or time of the day. Considering that kisspeptin neurons may act as interneurons and mediate VIP's effects on the reproductive axis, we investigated the effects of VIP on hypothalamic kisspeptin neurons in female mice during estrogen negative feedback. Our findings indicate that VIP induces a TTX-independent depolarization of approximately 30% of AVPV/PeN kisspeptin neurons in gonad-intact (diestrus) and ovariectomized (OVX) mice. In the ARH, the percentage of kisspeptin neurons that were depolarized by VIP was even higher (approximately 90%). An intracerebroventricular infusion of VIP leds to an increased percentage of kisspeptin neurons expressing the phosphoSer133 cAMP-response-element-binding protein (pCREB) in the AVPV/PeN. On the other hand, pCREB expression in ARH kisspeptin neurons was similar between saline- and VIP-injected mice. Thus, VIP can recruit different signaling pathways to modulate AVPV/PeN or ARH kisspeptin neurons, resulting in distinct cellular responses. The expression of VIP receptors (VPACR) was upregulated in the AVPV/PeN, but not in the ARH, of OVX mice compared to mice on diestrus and estradiol-primed OVX mice. Our findings indicate that VIP directly influences distinct cellular aspects of the AVPV/PeN and ARH kisspeptin neurons during estrogen negative feedback, possibly to influence pulsatile LH secretion.
Collapse
Affiliation(s)
- Naira da Silva Mansano
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Regina Silva Paradela
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Tabata M Bohlen
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Izabela M Zanardi
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Fernanda Machado Chaves
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Marina Augusto Silveira
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Mariana Rosolen Tavares
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Renata Frazao
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
27
|
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
|
28
|
Franssen D, Svingen T, Lopez Rodriguez D, Van Duursen M, Boberg J, Parent AS. A Putative Adverse Outcome Pathway Network for Disrupted Female Pubertal Onset to Improve Testing and Regulation of Endocrine Disrupting Chemicals. Neuroendocrinology 2022; 112:101-114. [PMID: 33640887 DOI: 10.1159/000515478] [Citation(s) in RCA: 6] [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: 10/20/2020] [Accepted: 02/25/2021] [Indexed: 11/19/2022]
Abstract
The average age for pubertal onset in girls has declined over recent decades. Epidemiological studies in humans and experimental studies in animals suggest a causal role for endocrine disrupting chemicals (EDCs) that are present in our environment. Of concern, current testing and screening regimens are inadequate in identifying EDCs that may affect pubertal maturation, not least because they do not consider early-life exposure. Also, the causal relationship between EDC exposure and pubertal timing is still a matter of debate. To address this issue, we have used current knowledge to elaborate a network of putative adverse outcome pathways (pAOPs) to identify how chemicals can affect pubertal onset. By using the AOP framework, we highlight current gaps in mechanistic understanding that need to be addressed and simultaneously point towards events causative of pubertal disturbance that could be exploited for alternative test methods. We propose 6 pAOPs that could explain the disruption of pubertal timing by interfering with the central hypothalamic trigger of puberty, GnRH neurons, and by so doing highlight specific modes of action that could be targeted for alternative test method development.
Collapse
Affiliation(s)
- Delphine Franssen
- Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Terje Svingen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Majorie Van Duursen
- Department of Environment and Health, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Julie Boberg
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Anne-Simone Parent
- Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Liège, Belgium
- Department of Pediatrics, CHU de Liège, Liège, Belgium
| |
Collapse
|
29
|
Barabás K, Kovács G, Vértes V, Kövesdi E, Faludi P, Udvarácz I, Pham D, Reglődi D, Abraham IM, Nagy Z. Stereology of gonadotropin-releasing hormone and kisspeptin neurons in PACAP gene-deficient female mice. Front Endocrinol (Lausanne) 2022; 13:993228. [PMID: 36387875 PMCID: PMC9640735 DOI: 10.3389/fendo.2022.993228] [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: 07/13/2022] [Accepted: 10/05/2022] [Indexed: 11/13/2022] Open
Abstract
The hypothalamic gonadotropin-releasing hormone (GnRH)-kisspeptin neuronal network regulates fertility in all mammals. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide isolated from the hypothalamus that is involved in the regulation of several releasing hormones and trop hormones. It is well-known that PACAP influences fertility at central and peripheral levels. However, the effects of PACAP on GnRH and kisspeptin neurons are not well understood. The present study investigated the integrity of the estrous cycle in PACAP-knockout (KO) mice. The number and immunoreactivity of GnRH (GnRH-ir) neurons in wild-type (WT) and PACAP KO female mice were determined using immunohistochemistry. In addition, the number of kisspeptin neurons was measured by counting kisspeptin mRNA-positive cells in the rostral periventricular region of the third ventricle (RP3V) and arcuate nucleus (ARC) using the RNAscope technique. Finally, the mRNA and protein expression of estrogen receptor alpha (ERα) was also examined. Our data showed that the number of complete cycles decreased, and the length of each cycle was longer in PACAP KO mice. Furthermore, the PACAP KO mice experienced longer periods of diestrus and spent significantly less time in estrus. There was no difference in GnRH-ir or number of GnRH neurons. In contrast, the number of kisspeptin neurons was decreased in the ARC, but not in the R3PV, in PACAP KO mice compared to WT littermates. Furthermore, ERα mRNA and protein expression was decreased in the ARC, whereas in the R3PV region, ERα mRNA levels were elevated. Our results demonstrate that embryonic deletion of PACAP significantly changes the structure and presumably the function of the GnRH-kisspeptin neuronal network, influencing fertility.
Collapse
Affiliation(s)
- Klaudia Barabás
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- *Correspondence: Klaudia Barabás, ; Gergely Kovács,
| | - Gergely Kovács
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- *Correspondence: Klaudia Barabás, ; Gergely Kovács,
| | - Viola Vértes
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
| | - Erzsébet Kövesdi
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - Péter Faludi
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - Ildikó Udvarácz
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - Dániel Pham
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- Department of Anatomy, Medical School, University of Pécs, Pécs, Hungary
| | - Dóra Reglődi
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- Department of Anatomy, Medical School, University of Pécs, Pécs, Hungary
| | - Istvan M. Abraham
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - Zsuzsanna Nagy
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
30
|
Minnig MA, Park T, Echeveste Sanchez M, Cottone P, Sabino V. Viral-Mediated Knockdown of Nucleus Accumbens Shell PAC1 Receptor Promotes Excessive Alcohol Drinking in Alcohol-Preferring Rats. Front Behav Neurosci 2021; 15:787362. [PMID: 34924973 PMCID: PMC8678417 DOI: 10.3389/fnbeh.2021.787362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/08/2021] [Indexed: 01/04/2023] Open
Abstract
Alcohol use disorder (AUD) is a chronic, relapsing disorder whose genetic and environmental susceptibility components are not fully understood. Neuropeptidergic signaling has been repeatedly implicated in modulating excessive alcohol drinking, especially within sub-regions of the striatum. Here, we investigated the potential involvement of the selective receptor for pituitary adenylate cyclase-activating polypeptide (PACAP), PAC1R, in the nucleus accumbens shell (NAcc Shell) in excessive alcohol drinking in alcohol-preferring rats, an established animal model of the genetic propensity for alcoholism. Scr:sP alcohol-preferring rats were trained to operantly self-administer alcohol and then either an AAV virus short-hairpin RNA (shRNA) targeted to knockdown PAC1R, or an AAV control virus were microinfused into the NAcc Shell. NAcc Shell PAC1R shRNA knockdown virus was confirmed to significantly decrease PAC1R levels in the NAcc Shell. The effects of NAcc Shell PAC1R shRNA knockdown on ethanol self-administration were investigated using a Fixed Ratio (FR) 1 and a Progressive Ratio (PR) schedule of reinforcement. The effect of PAC1R knockdown on self-administration of an alternative reinforcer, saccharin, was also assessed. The results showed that the reduction in PAC1R in the NAcc Shell led to excessive ethanol drinking, increased preference for ethanol, and higher motivation to drink. NAcc Shell PAC1R shRNA knockdown did not comparably increase saccharin self-administration, suggesting selectivity of action. These data suggest that NAcc Shell PAC1R may serves as a "brake" on alcohol drinking, and thereby the loss of function of PAC1R leads to excessive alcohol consumption. Therefore, the PACAP/PAC1R system may represent a novel target for the treatment of AUD.
Collapse
Affiliation(s)
| | | | | | | | - Valentina Sabino
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
31
|
Deciphering the spatial-temporal transcriptional landscape of human hypothalamus development. Cell Stem Cell 2021; 29:328-343.e5. [PMID: 34879244 DOI: 10.1016/j.stem.2021.11.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 09/13/2021] [Accepted: 11/12/2021] [Indexed: 11/24/2022]
Abstract
The hypothalamus comprises various nuclei and neuronal subpopulations that control fundamental homeostasis and behaviors. However, spatiotemporal molecular characterization of hypothalamus development in humans is largely unexplored. Here, we revealed spatiotemporal transcriptome profiles and cell-type characteristics of human hypothalamus development and illustrated the molecular diversity of neural progenitors and the cell-fate decision, which is programmed by a combination of transcription factors. Different neuronal and glial fates are sequentially produced and showed spatial developmental asynchrony. Moreover, human hypothalamic gliogenesis occurs at an earlier stage of gestation and displays distinctive transcription profiles compared with those in mouse. Notably, early oligodendrocyte cells in humans exhibit different gene patterns and interact with neuronal cells to regulate neuronal maturation by Wnt, Hippo, and integrin signals. Overall, our study provides a comprehensive molecular landscape of human hypothalamus development at early- and mid-embryonic stages and a foundation for understanding its spatial and functional complexity.
Collapse
|
32
|
Quaresma PGF, Wasinski F, Mansano NS, Furigo IC, Teixeira PDS, Gusmao DO, Frazao R, Donato J. Leptin Receptor Expression in GABAergic Cells is Not Sufficient to Normalize Metabolism and Reproduction in Mice. Endocrinology 2021; 162:6353267. [PMID: 34402859 DOI: 10.1210/endocr/bqab168] [Citation(s) in RCA: 8] [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: 06/25/2021] [Indexed: 12/12/2022]
Abstract
Previous studies indicate that leptin receptor (LepR) expression in GABAergic neurons is necessary for the biological effects of leptin. However, it is not clear whether LepR expression only in GABAergic neurons is sufficient to prevent the metabolic and neuroendocrine imbalances caused by LepR deficiency. In the present study, we produced mice that express the LepR exclusively in GABAergic cells (LepRVGAT mice) and compared them with wild-type (LepR+/+) and LepR-deficient (LepRNull/Null) mice. Although LepRVGAT mice showed a pronounced reduction in body weight and fat mass, as compared with LepRNull/Null mice, male and female LepRVGAT mice exhibited an obese phenotype relative to LepR+/+ mice. Food intake was normalized in LepRVGAT mice; however, LepRVGAT mice still exhibited lower energy expenditure in both sexes and reduced ambulatory activity in the females, compared with LepR+/+ mice. The acute anorexigenic effect of leptin and hedonic feeding were normalized in LepRVGAT mice despite the hyperleptinemia they present. Although LepRVGAT mice showed improved glucose homeostasis compared with LepRNull/Null mice, both male and female LepRVGAT mice exhibited insulin resistance. In contrast, LepR expression only in GABAergic cells was sufficient to normalize the density of agouti-related peptide (AgRP) and α-MSH immunoreactive fibers in the paraventricular nucleus of the hypothalamus. However, LepRVGAT mice exhibited reproductive dysfunctions, including subfertility in males and alterations in the estrous cycle of females. Taken together, our findings indicate that LepR expression in GABAergic cells, although critical to the physiology of leptin, is insufficient to normalize several metabolic aspects and the reproductive function in mice.
Collapse
Affiliation(s)
- Paula G F Quaresma
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Frederick Wasinski
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Naira S Mansano
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Isadora C Furigo
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Pryscila D S Teixeira
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Daniela O Gusmao
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Renata Frazao
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Jose Donato
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| |
Collapse
|
33
|
Xu S, Zhou L, Guo S, Hu Q, Shi X, Xia C, Zhang H, Ye C, Jia Y, Hu G. Different pituitary action of NK3Ra and NK3Rb in grass carp. Gen Comp Endocrinol 2021; 313:113829. [PMID: 34087185 DOI: 10.1016/j.ygcen.2021.113829] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/03/2021] [Accepted: 05/31/2021] [Indexed: 10/21/2022]
Abstract
In mammals, NK3R is the specific receptor for NKB, which played an important role in reproduction. Recently, two NK3R isoforms, namely NK3Ra and NK3Rb, have been identified in fish. However, little is known about the pituitary actions of the two NK3R isoforms in fish. In this study, both NK3Ra and NK3Rb were isolated from grass carp pituitary. Although their sequence similarity was only 61.6%, the two NK3R isoforms displayed similar ligand selectivity and binding affinity to TAC3 gene products (NKBa, NKBRPa and NKBRPb). In addition, both NK3Ra and NK3Rb displayed similar signaling pathways, including PKA, PKC, MAPK and Ca2+ cascades. Tissue distribution indicated that both NK3Ra and NK3Rb were highly detected in grass carp pituitary. Further study found that NK3Ra was mainly located in pituitary LHβ cells, while NK3Rb was only detected in pituitary SLα cells. Furthermore, NK3Ra and NK3Rb activation could induce LHβ and SLα promoter activity, respectively. These results suggested that the two NK3R isoforms displayed different pituitary actions in fish. Using grass carp pituitary cells as model, we found that PACAP could significantly reduce NK3Ra, but induce NK3Rb mRNA expression coupled with cAMP/PKA and PLC/PKC pathways. Interestingly, PACAP could also significantly inhibit LHβ, but stimulate SLα mRNA expression in grass carp pituitary cells. Furthermore, NK3R antagonist could not only inhibit LHβ mRNA expression, but also block PACAP-induced SLα mRNA expression in grass carp pituitary cells. These results suggested that NK3Ra and NK3Rb could mediate PACAP-reduced LHβ and -induced SLα mRNA expression in grass carp pituitary, respectively.
Collapse
Affiliation(s)
- Shaohua Xu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Lingling Zhou
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Shuming Guo
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Qiongyao Hu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Xuetao Shi
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Chuanhui Xia
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Huiying Zhang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Cheng Ye
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Yongyi Jia
- Agriculture Ministry Key Laboratory of Healthy Freshwater Aquaculture, Key Laboratory of Fish Health and Nutrition of Zhejiang Province, Zhejiang Institute of Freshwater Fisheries, Huzhou 313001, China.
| | - Guangfu Hu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
34
|
Maione L, Bouvattier C, Kaiser UB. Central precocious puberty: Recent advances in understanding the aetiology and in the clinical approach. Clin Endocrinol (Oxf) 2021; 95:542-555. [PMID: 33797780 PMCID: PMC8586890 DOI: 10.1111/cen.14475] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 12/13/2022]
Abstract
Central precocious puberty (CPP) results from early activation of the hypothalamic-pituitary-gonadal (HPG) axis. The current state of knowledge of the complex neural network acting at the level of the hypothalamus and the GnRH neuron to control puberty onset has expanded, particularly in the context of molecular interactions. Along with these advances, the knowledge of pubertal physiology and pathophysiology has also increased. This review focuses on regulatory abnormalities occurring at the hypothalamic level of the HPG axis to cause CPP. The clinical approach to diagnosis of puberty and pubertal disorders is also reviewed, with a particular focus on aetiologies of CPP. The recent identification of mutations in MKRN3 and DLK1 in familial as well sporadic forms of CPP has changed the state of the art of the approach to patients with CPP. Genetic advances have also had important repercussions beyond consideration of puberty alone. Syndromic disorders and central nervous system lesions associated with CPP are also discussed. If untreated, these conditions may lead to adverse physical, psychosocial and medical outcomes.
Collapse
Affiliation(s)
- Luigi Maione
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Rares de l’Hypophyse, Université Paris-Saclay, Paris-Saclay University, Le Kremlin-Bicêtre, France
| | - Claire Bouvattier
- Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Rares de l’Hypophyse, Université Paris-Saclay, Paris-Saclay University, Le Kremlin-Bicêtre, France
| | - Ursula B. Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
35
|
Leon S, Talbi R, McCarthy EA, Ferrari K, Fergani C, Naule L, Choi JH, Carroll RS, Kaiser UB, Aylwin CF, Lomniczi A, Navarro VM. Sex-specific pubertal and metabolic regulation of Kiss1 neurons via Nhlh2. eLife 2021; 10:e69765. [PMID: 34494548 PMCID: PMC8439651 DOI: 10.7554/elife.69765] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 09/03/2021] [Indexed: 12/21/2022] Open
Abstract
Hypothalamic Kiss1 neurons control gonadotropin-releasing hormone release through the secretion of kisspeptin. Kiss1 neurons serve as a nodal center that conveys essential regulatory cues for the attainment and maintenance of reproductive function. Despite this critical role, the mechanisms that control kisspeptin synthesis and release remain largely unknown. Using Drop-Seq data from the arcuate nucleus of adult mice and in situ hybridization, we identified Nescient Helix-Loop-Helix 2 (Nhlh2), a transcription factor of the basic helix-loop-helix family, to be enriched in Kiss1 neurons. JASPAR analysis revealed several binding sites for NHLH2 in the Kiss1 and Tac2 (neurokinin B) 5' regulatory regions. In vitro luciferase assays evidenced a robust stimulatory action of NHLH2 on human KISS1 and TAC3 promoters. The recruitment of NHLH2 to the KISS1 and TAC3 promoters was further confirmed through chromatin immunoprecipitation. In vivo conditional ablation of Nhlh2 from Kiss1 neurons using Kiss1Cre:Nhlh2fl/fl mice induced a male-specific delay in puberty onset, in line with a decrease in arcuate Kiss1 expression. Females retained normal reproductive function albeit with irregular estrous cycles. Further analysis of male Kiss1Cre:Nhlh2fl/fl mice revealed higher susceptibility to metabolic challenges in the release of luteinizing hormone and impaired response to leptin. Overall, in Kiss1 neurons, Nhlh2 contributes to the metabolic regulation of kisspeptin and NKB synthesis and release, with implications for the timing of puberty onset and regulation of fertility in male mice.
Collapse
Affiliation(s)
- Silvia Leon
- Harvard Medical SchoolBostonUnited States
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s HospitalBostonUnited States
| | - Rajae Talbi
- Harvard Medical SchoolBostonUnited States
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s HospitalBostonUnited States
| | - Elizabeth A McCarthy
- Harvard Medical SchoolBostonUnited States
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s HospitalBostonUnited States
| | - Kaitlin Ferrari
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s HospitalBostonUnited States
| | - Chrysanthi Fergani
- Harvard Medical SchoolBostonUnited States
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s HospitalBostonUnited States
| | - Lydie Naule
- Harvard Medical SchoolBostonUnited States
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s HospitalBostonUnited States
| | - Ji Hae Choi
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s HospitalBostonUnited States
| | - Rona S Carroll
- Harvard Medical SchoolBostonUnited States
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s HospitalBostonUnited States
| | - Ursula B Kaiser
- Harvard Medical SchoolBostonUnited States
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s HospitalBostonUnited States
| | - Carlos F Aylwin
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Alejandro Lomniczi
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Víctor M Navarro
- Harvard Medical SchoolBostonUnited States
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s HospitalBostonUnited States
- Harvard Program in NeuroscienceBostonUnited States
| |
Collapse
|
36
|
Bozadjieva-Kramer N, Ross RA, Johnson DQ, Fenselau H, Haggerty DL, Atwood B, Lowell B, Flak JN. The Role of Mediobasal Hypothalamic PACAP in the Control of Body Weight and Metabolism. Endocrinology 2021; 162:6103920. [PMID: 33460433 PMCID: PMC7875177 DOI: 10.1210/endocr/bqab012] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Indexed: 12/26/2022]
Abstract
Body energy homeostasis results from balancing energy intake and energy expenditure. Central nervous system administration of pituitary adenylate cyclase activating polypeptide (PACAP) dramatically alters metabolic function, but the physiologic mechanism of this neuropeptide remains poorly defined. PACAP is expressed in the mediobasal hypothalamus (MBH), a brain area essential for energy balance. Ventromedial hypothalamic nucleus (VMN) neurons contain, by far, the largest and most dense population of PACAP in the medial hypothalamus. This region is involved in coordinating the sympathetic nervous system in response to metabolic cues in order to re-establish energy homeostasis. Additionally, the metabolic cue of leptin signaling in the VMN regulates PACAP expression. We hypothesized that PACAP may play a role in the various effector systems of energy homeostasis, and tested its role by using VMN-directed, but MBH encompassing, adeno-associated virus (AAVCre) injections to ablate Adcyap1 (gene coding for PACAP) in mice (Adcyap1MBHKO mice). Adcyap1MBHKO mice rapidly gained body weight and adiposity, becoming hyperinsulinemic and hyperglycemic. Adcyap1MBHKO mice exhibited decreased oxygen consumption (VO2), without changes in activity. These effects appear to be due at least in part to brown adipose tissue (BAT) dysfunction, and we show that PACAP-expressing cells in the MBH can stimulate BAT thermogenesis. While we observed disruption of glucose clearance during hyperinsulinemic/euglycemic clamp studies in obese Adcyap1MBHKO mice, these parameters were normal prior to the onset of obesity. Thus, MBH PACAP plays important roles in the regulation of metabolic rate and energy balance through multiple effector systems on multiple time scales, which highlight the diverse set of functions for PACAP in overall energy homeostasis.
Collapse
Affiliation(s)
| | - Rachel A Ross
- Albert Einstein College of Medicine, Bronx, NY, USA
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - David Q Johnson
- Indiana Biosciences Research Institute, Diabetes Research Center, Indianapolis, IN, USA
| | - Henning Fenselau
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - David L Haggerty
- Indiana University School of Medicine, Pharmacology and Toxicology, Indianapolis, IN, USA
| | - Brady Atwood
- Indiana University School of Medicine, Pharmacology and Toxicology, Indianapolis, IN, USA
| | - Bradford Lowell
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jonathan N Flak
- Indiana Biosciences Research Institute, Diabetes Research Center, Indianapolis, IN, USA
- Indiana University School of Medicine, Pharmacology and Toxicology, Indianapolis, IN, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Correspondence: Jonathan N. Flak, PhD, Indiana Biosciences Research Institute, 1345 W. 16th Street, Indianapolis, IN 46022, USA.
| |
Collapse
|
37
|
Wasinski F, Chaves FM, Pedroso JA, Mansano NS, Camporez JP, Gusmão DO, List EO, Kopchick JJ, Frazão R, Szawka RE, Donato J. Growth hormone receptor in dopaminergic neurones regulates stress-induced prolactin release in male mice. J Neuroendocrinol 2021; 33:e12957. [PMID: 33769619 PMCID: PMC9670090 DOI: 10.1111/jne.12957] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 02/03/2021] [Accepted: 02/10/2021] [Indexed: 12/15/2022]
Abstract
Arcuate nucleus (ARH) dopaminergic neurones regulate several biological functions, including prolactin secretion and metabolism. These cells are responsive to growth hormone (GH), although it is still unknown whether GH action on ARH dopaminergic neurones is required to regulate different physiological aspects. Mice carrying specific deletion of GH receptor (GHR) in tyrosine hydroxylase (TH)- or dopamine transporter (DAT)-expressing cells were produced. We investigated possible changes in energy balance, glucose homeostasis, fertility, pup survival and restraint stress-induced prolactin release. GHR deletion in DAT- or TH-expressing cells did not cause changes in food intake, energy expenditure, ambulatory activity, nutrient oxidation, glucose tolerance, insulin sensitivity and counter-regulatory response to hypoglycaemia in male and female mice. In addition, GHR deletion in dopaminergic cells caused no gross effects on reproduction and pup survival. However, restraint stress-induced prolactin release was significantly impaired in DAT- and TH-specific GHR knockout male mice, as well as in pegvisomant-treated wild-type males, whereas an intact response was observed in females. Patch clamp recordings were performed in ARH DAT neurones and, in contrast to prolactin, GH did not cause acute changes in the electrical activity of DAT neurones. Furthermore, TH phosphorylation at Ser40 in ARH neurones and median eminence axonal terminals was not altered in DAT-specific GHR knockout male mice during restraint stress. In conclusion, GH action in dopaminergic neurones is required for stress-induced prolactin release in male mice, suggesting the existence of sex differences in the capacity of GHR signalling to affect prolactin secretion. The mechanism behind this regulation still needs to be identified.
Collapse
Affiliation(s)
- Frederick Wasinski
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo, Brazil
| | - Fernanda M. Chaves
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo, Brazil
| | - João A.B. Pedroso
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo, Brazil
| | - Naira S. Mansano
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Anatomia, Sao Paulo, Brazil
| | - João Paulo Camporez
- Universidade de Sao Paulo, Faculdade de Medicina de Ribeirao Preto, Departamento de Fisiologia, Ribeirao Preto, Brazil
| | - Daniela O. Gusmão
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Edward O. List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - John J. Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Renata Frazão
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Anatomia, Sao Paulo, Brazil
| | - Raphael E. Szawka
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jose Donato
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo, Brazil
| |
Collapse
|
38
|
Abstract
A healthy nutritional state is required for all aspects of reproduction and is signaled by the adipokine leptin. Leptin acts in a relatively narrow concentration range: too much or too little will compromise fertility. The leptin signal timing is important to prepubertal development in both sexes. In the brain, leptin acts on ventral premammillary neurons which signal kisspeptin (Kiss1) neurons to stimulate gonadotropin releasing hormone (GnRH) neurons. Suppression of Kiss1 neurons occurs when agouti-related peptide neurons are activated by reduced leptin, because leptin normally suppresses these orexigenic neurons. In the pituitary, leptin stimulates production of GnRH receptors (GnRHRs) and follicle-stimulating hormone at midcycle, by activating pathways that derepress actions of the messenger ribonucleic acid translational regulatory protein Musashi. In females, rising estrogen stimulates a rise in serum leptin, which peaks at midcycle, synchronizing with nocturnal luteinizing hormone pulses. The normal range of serum leptin levels (10-20 ng/mL) along with gonadotropins and growth factors promote ovarian granulosa and theca cell functions and oocyte maturation. In males, the prepubertal rise in leptin promotes testicular development. However, a decline in leptin levels in prepubertal boys reflects inhibition of leptin secretion by rising androgens. In adult males, leptin levels are 10% to 50% of those in females, and high leptin inhibits testicular function. The obesity epidemic has elucidated leptin resistance pathways, with too much leptin in either sex leading to infertility. Under conditions of balanced nutrition, however, the secretion of leptin is timed and regulated within a narrow level range that optimizes its trophic effects.
Collapse
Affiliation(s)
- Gwen V Childs
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Correspondence: Gwen V. Childs, PhD, University of Arkansas for Medical Sciences, Little Rock, AR, USA. E-mail:
| | - Angela K Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
39
|
Shi Y, Stornetta DS, Reklow RJ, Sahu A, Wabara Y, Nguyen A, Li K, Zhang Y, Perez-Reyes E, Ross RA, Lowell BB, Stornetta RL, Funk GD, Guyenet PG, Bayliss DA. A brainstem peptide system activated at birth protects postnatal breathing. Nature 2021; 589:426-430. [PMID: 33268898 PMCID: PMC7855323 DOI: 10.1038/s41586-020-2991-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 09/29/2020] [Indexed: 01/29/2023]
Abstract
Among numerous challenges encountered at the beginning of extrauterine life, the most celebrated is the first breath that initiates a life-sustaining motor activity1. The neural systems that regulate breathing are fragile early in development, and it is not clear how they adjust to support breathing at birth. Here we identify a neuropeptide system that becomes activated immediately after birth and supports breathing. Mice that lack PACAP selectively in neurons of the retrotrapezoid nucleus (RTN) displayed increased apnoeas and blunted CO2-stimulated breathing; re-expression of PACAP in RTN neurons corrected these breathing deficits. Deletion of the PACAP receptor PAC1 from the pre-Bötzinger complex-an RTN target region responsible for generating the respiratory rhythm-phenocopied the breathing deficits observed after RTN deletion of PACAP, and suppressed PACAP-evoked respiratory stimulation in the pre-Bötzinger complex. Notably, a postnatal burst of PACAP expression occurred in RTN neurons precisely at the time of birth, coinciding with exposure to the external environment. Neonatal mice with deletion of PACAP in RTN neurons displayed increased apnoeas that were further exacerbated by changes in ambient temperature. Our findings demonstrate that well-timed PACAP expression by RTN neurons provides an important supplementary respiratory drive immediately after birth and reveal key molecular components of a peptidergic neural circuit that supports breathing at a particularly vulnerable period in life.
Collapse
Affiliation(s)
- Yingtang Shi
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel S. Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Robert J. Reklow
- Department of Physiology, Women & Children’s Health Research Institute, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Alisha Sahu
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Yvonne Wabara
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Ashley Nguyen
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Keyong Li
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Yong Zhang
- Department of Physiology, Women & Children’s Health Research Institute, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Edward Perez-Reyes
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Rachel A. Ross
- Beth Israel Deaconess Medical Center, Harvard University, Boston, MA, USA,McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
| | - Bradford B. Lowell
- Beth Israel Deaconess Medical Center, Harvard University, Boston, MA, USA
| | - Ruth L. Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Gregory D. Funk
- Department of Physiology, Women & Children’s Health Research Institute, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Patrice G. Guyenet
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Douglas A. Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
40
|
Winters SJ, Moore JP. PACAP: A regulator of mammalian reproductive function. Mol Cell Endocrinol 2020; 518:110912. [PMID: 32561449 PMCID: PMC7606562 DOI: 10.1016/j.mce.2020.110912] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/14/2020] [Accepted: 06/06/2020] [Indexed: 12/19/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is an ancestral molecule that was isolated from sheep hypothalamic extracts based on its action to stimulate cAMP production by pituitary cell cultures. PACAP is one of a number of ligands that coordinate with GnRH to control reproduction. While initially viewed as a hypothalamic releasing factor, PACAP and its receptors are widely distributed, and there is growing evidence that PACAP functions as a paracrine/autocrine regulator in the CNS, pituitary, gonads and placenta, among other tissues. This review will summarize current knowledge concerning the expression and function of PACAP in the hypothalamic-pituitary-gonadal axis with special emphasis on its role in pituitary function in the fetus and newborn.
Collapse
Affiliation(s)
- Stephen J Winters
- Division of Endocrinology, Metabolism and Diabetes, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| | - Joseph P Moore
- Division of Endocrinology, Metabolism and Diabetes, University of Louisville School of Medicine, Louisville, KY, 40202, USA; Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| |
Collapse
|
41
|
Mickelsen LE, Flynn WF, Springer K, Wilson L, Beltrami EJ, Bolisetty M, Robson P, Jackson AC. Cellular taxonomy and spatial organization of the murine ventral posterior hypothalamus. eLife 2020; 9:58901. [PMID: 33119507 PMCID: PMC7595735 DOI: 10.7554/elife.58901] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/21/2020] [Indexed: 01/02/2023] Open
Abstract
The ventral posterior hypothalamus (VPH) is an anatomically complex brain region implicated in arousal, reproduction, energy balance, and memory processing. However, neuronal cell type diversity within the VPH is poorly understood, an impediment to deconstructing the roles of distinct VPH circuits in physiology and behavior. To address this question, we employed a droplet-based single-cell RNA sequencing (scRNA-seq) approach to systematically classify molecularly distinct cell populations in the mouse VPH. Analysis of >16,000 single cells revealed 20 neuronal and 18 non-neuronal cell populations, defined by suites of discriminatory markers. We validated differentially expressed genes in selected neuronal populations through fluorescence in situ hybridization (FISH). Focusing on the mammillary bodies (MB), we discovered transcriptionally-distinct clusters that exhibit neuroanatomical parcellation within MB subdivisions and topographic projections to the thalamus. This single-cell transcriptomic atlas of VPH cell types provides a resource for interrogating the circuit-level mechanisms underlying the diverse functions of VPH circuits.
Collapse
Affiliation(s)
- Laura E Mickelsen
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, United States.,Connecticut Institute for the Brain and Cognitive Sciences, Storrs, United States
| | - William F Flynn
- The Jackson Laboratory for Genomic Medicine, Farmington, United States
| | - Kristen Springer
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, United States
| | - Lydia Wilson
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, United States
| | - Eric J Beltrami
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, United States
| | - Mohan Bolisetty
- The Jackson Laboratory for Genomic Medicine, Farmington, United States
| | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, Farmington, United States.,Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, United States.,Institute for Systems Genomics, University of Connecticut, Farmington, United States
| | - Alexander C Jackson
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, United States.,Connecticut Institute for the Brain and Cognitive Sciences, Storrs, United States.,Institute for Systems Genomics, University of Connecticut, Farmington, United States
| |
Collapse
|
42
|
Hypothalamic and Cell-Specific Transcriptomes Unravel a Dynamic Neuropil Remodeling in Leptin-Induced and Typical Pubertal Transition in Female Mice. iScience 2020; 23:101563. [PMID: 33083731 PMCID: PMC7522126 DOI: 10.1016/j.isci.2020.101563] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/16/2020] [Accepted: 09/10/2020] [Indexed: 01/01/2023] Open
Abstract
Epidemiological and genome-wide association studies (GWAS) have shown high correlation between childhood obesity and advance in puberty. Early age at menarche is associated with a series of morbidities, including breast cancer, cardiovascular diseases, type 2 diabetes, and obesity. The adipocyte hormone leptin signals the amount of fat stores to the neuroendocrine reproductive axis via direct actions in the brain. Using mouse genetics, we and others have identified the hypothalamic ventral premammillary nucleus (PMv) and the agouti-related protein (AgRP) neurons in the arcuate nucleus (Arc) as primary targets of leptin action in pubertal maturation. However, the molecular mechanisms underlying leptin's effects remain unknown. Here we assessed changes in the PMv and Arc transcriptional program during leptin-stimulated and typical pubertal development using overlapping analysis of bulk RNA sequecing, TRAP sequencing, and the published database. Our findings demonstrate that dynamic somatodendritic remodeling and extracellular space organization underlie leptin-induced and typical pubertal maturation in female mice. MBH DEGs between lean and Lepob mice are highly represented in development Short-term leptin to Lepob mice alters MBH DEGs associated with reproduction PMv/Arc LepRb DEGs between lean and Lepob mice are abundant in extracellular space DEGs in developing PMv/Arc are conspicuous in extracellular and neuropil remodeling
Collapse
|
43
|
Roberts SA, Abreu AP, Navarro VM, Liang JN, Maguire CA, Kim HK, Carroll RS, Kaiser UB. The Peripubertal Decline in Makorin Ring Finger Protein 3 Expression is Independent of Leptin Action. J Endocr Soc 2020; 4:bvaa059. [PMID: 32587933 PMCID: PMC7304661 DOI: 10.1210/jendso/bvaa059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/19/2020] [Indexed: 01/04/2023] Open
Abstract
A critical body weight is necessary for pubertal development, an effect mediated in part by leptin. The potential regulation by leptin of Makorin Ring Finger Protein 3 (MKRN3), in which loss-of-function mutations are the most common genetic cause of central precocious puberty, has not been previously explored. In mice, expression of Mkrn3 in the hypothalamic arcuate nucleus is high early in life and declines before the onset of puberty. Therefore, we aimed to explore if leptin contributes to the decrease in hypothalamic Mkrn3 mRNA levels observed in mice during pubertal development. We first used a leptin-deficient (ob/ob) mouse model. Mkrn3 mRNA levels in the mediobasal hypothalamus (MBH), which includes the arcuate nucleus, and in the preoptic area (POA), both showed a significant decrease with age from postnatal day (PND) 12 to PND30 in ob/ob mice in both males and females, similar to that observed in wild-type mice. To further explore the effects of leptin on Mkrn3 expression, we exposed prepubertal wild-type mice to high levels of leptin from age PND9-12, which did not result in any significant difference in Mkrn3 expression levels in either the MBH or POA. In summary, regulation of Mkrn3 expression by leptin was not observed in either the MBH or the POA, 2 hypothalamic sites important for pubertal maturation. These data suggest that the decline in Mkrn3 at the onset of puberty may occur independently of leptin and support our hypothesis that MKRN3 is a bona fide controller of puberty initiation.
Collapse
Affiliation(s)
- Stephanie A Roberts
- Division of Endocrinology, Boston Children’s Hospital, Boston, Massachusetts
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Ana Paula Abreu
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Victor M Navarro
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Joy N Liang
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Caroline A Maguire
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Han Kyeol Kim
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Rona S Carroll
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
44
|
Circulating PACAP peptide and PAC1R genotype as possible transdiagnostic biomarkers for anxiety disorders in women: a preliminary study. Neuropsychopharmacology 2020; 45:1125-1133. [PMID: 31910434 PMCID: PMC7235237 DOI: 10.1038/s41386-020-0604-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/30/2019] [Accepted: 12/30/2019] [Indexed: 01/04/2023]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP, gene Adcyap1) is a neuropeptide and hormone thought to play a critical role in stress response (Stroth et al., Ann NY Acad Sci 1220:49-59, 2011; Hashimoto et al., Curr Pharm Des 17:985-989, 2011). Research in humans implicates PACAP as a useful biomarker for the severity of psychiatric symptoms in response to psychological stressors, and work in rodent models suggests that PACAP manipulation exerts downstream effects on peripheral hormones and behaviors linked to the stress response, providing a potential therapeutic target. Prior work has also suggested a potential sex difference in PACAP effects due to differential estrogen regulation of this pathway. Therefore, we examined serum PACAP and associated PAC1R genotype in a cohort of males and females with a primary diagnosis of generalized anxiety disorder (GAD) and nonpsychiatric controls. We found that, while circulating hormone levels were not associated with a GAD diagnosis overall (p = 0.19, g = 0.25), PACAP may be associated with GAD in females (p = 0.04, g = 0.33). Additionally, among patients with GAD, the risk genotype identified in the PTSD literature (rs2267735, CC genotype) was associated with higher somatic anxiety symptom severity in females but lower somatic anxiety symptom severity in males (-3.27, 95%CI [-5.76, -0.77], adjusted p = 0.03). Taken together, the associations between the risk genotype, circulating PACAP, and somatic anxiety severity were stronger among females than males. These results indicate a potential underlying biological etiology for sex differences in stress-related anxiety disorders that warrants further study.
Collapse
|
45
|
Cangiano B, Swee DS, Quinton R, Bonomi M. Genetics of congenital hypogonadotropic hypogonadism: peculiarities and phenotype of an oligogenic disease. Hum Genet 2020; 140:77-111. [PMID: 32200437 DOI: 10.1007/s00439-020-02147-1] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 03/04/2020] [Indexed: 12/30/2022]
Abstract
A genetic basis of congenital isolated hypogonadotropic hypogonadism (CHH) can be defined in almost 50% of cases, albeit not necessarily the complete genetic basis. Next-generation sequencing (NGS) techniques have led to the discovery of a great number of loci, each of which has illuminated our understanding of human gonadotropin-releasing hormone (GnRH) neurons, either in respect of their embryonic development or their neuroendocrine regulation as the "pilot light" of human reproduction. However, because each new gene linked to CHH only seems to underpin another small percentage of total patient cases, we are still far from achieving a comprehensive understanding of the genetic basis of CHH. Patients have generally not benefited from advances in genetics in respect of novel therapies. In most cases, even genetic counselling is limited by issues of apparent variability in expressivity and penetrance that are likely underpinned by oligogenicity in respect of known and unknown genes. Robust genotype-phenotype relationships can generally only be established for individuals who are homozygous, hemizygous or compound heterozygotes for the same gene of variant alleles that are predicted to be deleterious. While certain genes are purely associated with normosmic CHH (nCHH) some purely with the anosmic form (Kallmann syndrome-KS), other genes can be associated with both nCHH and KS-sometimes even within the same kindred. Even though the anticipated genetic overlap between CHH and constitutional delay in growth and puberty (CDGP) has not materialised, previously unanticipated genetic relationships have emerged, comprising conditions of combined (or multiple) pituitary hormone deficiency (CPHD), hypothalamic amenorrhea (HA) and CHARGE syndrome. In this review, we report the current evidence in relation to phenotype and genetic peculiarities regarding 60 genes whose loss-of-function variants can disrupt the central regulation of reproduction at many levels: impairing GnRH neurons migration, differentiation or activation; disrupting neuroendocrine control of GnRH secretion; preventing GnRH neuron migration or function and/or gonadotropin secretion and action.
Collapse
Affiliation(s)
- Biagio Cangiano
- Department of Clinical Sciences and Community Health, University of Milan, 20100, Milan, Italy.,Department of Endocrine and Metabolic Diseases and Laboratory of Endocrine and Metabolic Research, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, 20149, Milan, Italy
| | - Du Soon Swee
- Department of Endocrinology, Singapore General Hospital, Singapore, Singapore
| | - Richard Quinton
- Endocrine Unit, Royal Victoria Infirmary, Department of Endocrinology, Diabetes and Metabolism, Newcastle-Upon-Tyne Hospitals, Newcastle-Upon-Tyne, NE1 4LP, UK. .,Translational and Clinical Research Institute, University of Newcastle-Upon-Tyne, Newcastle-Upon-Tyne, UK.
| | - Marco Bonomi
- Department of Clinical Sciences and Community Health, University of Milan, 20100, Milan, Italy. .,Department of Endocrine and Metabolic Diseases and Laboratory of Endocrine and Metabolic Research, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, 20149, Milan, Italy.
| |
Collapse
|
46
|
Köves K, Szabó E, Kántor O, Heinzlmann A, Szabó F, Csáki Á. Current State of Understanding of the Role of PACAP in the Hypothalamo-Hypophyseal Gonadotropin Functions of Mammals. Front Endocrinol (Lausanne) 2020; 11:88. [PMID: 32210912 PMCID: PMC7067695 DOI: 10.3389/fendo.2020.00088] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 02/11/2020] [Indexed: 01/25/2023] Open
Abstract
PACAP was discovered 30 years ago in Dr. Akira Arimura's laboratory. In the past three decades since then, it has become evident that this peptide plays numerous crucial roles in mammalian organisms. The most important functions of PACAP are the following: 1. neurotransmitter, 2. neuromodulator, 3. hypophysiotropic hormone, 4. neuroprotector. This paper reviews the accumulated data regarding the distribution of PACAP and its receptors in the mammalian hypothalamus and pituitary gland, the role of PACAP in the gonadotropin hormone secretion of females and males. The review also summarizes the interaction between PACAP, GnRH, and sex steroids as well as hypothalamic peptides including kisspeptin. The possible role of PACAP in reproductive functions through the biological clock is also discussed. Finally, the significance of PACAP in the hypothalamo-hypophysial system is considered and the facts missing, that would help better understand the function of PACAP in this system, are also highlighted.
Collapse
Affiliation(s)
- Katalin Köves
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Enikő Szabó
- Department of Conservative Dentistry, Faculty of Dentistry, Semmelweis University, Budapest, Hungary
| | - Orsolya Kántor
- Department of Molecular Embryology, Medical Faculty, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | - Andrea Heinzlmann
- Department of Anatomy and Histology, University of Veterinary Sciences, Budapest, Hungary
| | - Flóra Szabó
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA, United States
| | - Ágnes Csáki
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
47
|
Rønnekleiv OK, Qiu J, Kelly MJ. Arcuate Kisspeptin Neurons Coordinate Reproductive Activities with Metabolism. Semin Reprod Med 2019; 37:131-140. [PMID: 31869841 DOI: 10.1055/s-0039-3400251] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Hypothalamic control of fertility is the quintessential homeostatic function. However, fertility is metabolically demanding; so, there must be coordination between energy states and reproductive functions. Because gonadotropin-releasing hormone (GnRH) neurons are devoid of many of the critical metabolic hormone receptors for sensing nutrient levels, it has long been recognized that the sensing of energy stores had to be done by neurons presynaptic to GnRH neurons. Some of the obvious players have been the anorexigenic proopiomelanocortin (POMC) and orexigenic neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurons, both of which are in close apposition to the median eminence, a circumventricular organ. Indeed, POMC and NPY/AgRP neurons are inversely regulated by glucose and metabolic hormones including insulin and leptin. However, their synaptic connections with GnRH neurons are sparse and/or GnRH neurons are lacking the postsynaptic receptors to mediate the appropriate physiological response. Kisspeptin neurons were discovered in the early part of this century and subsequently shown to project to and control GnRH neuronal excitability. In fact, more recently the arcuate kisspeptin neurons have been identified as the command neurons driving pulsatile release of GnRH. Subsequently, it was shown that arcuate kisspeptin neurons express not only steroid hormone receptors but also metabolic hormone receptors such that similar to POMC neurons, they are excited by insulin and leptin. Therefore, based on the premise that arcuate kisspeptin neurons are the key neurons coordinating energy states with reproduction, we will review not only how these vital neurons control pulsatile GnRH release but how they control energy homeostasis through their synaptic connections with POMC and NPY/AgRP neurons and ultimately how E2 can regulate their excitability.
Collapse
Affiliation(s)
- Oline K Rønnekleiv
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon.,Division of Neuroscience, National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon
| | - Jian Qiu
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon
| | - Martin J Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon.,Division of Neuroscience, National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon
| |
Collapse
|
48
|
Meggyes M, Lajko A, Fulop BD, Reglodi D, Szereday L. Phenotypic characterization of testicular immune cells expressing immune checkpoint molecules in wild-type and pituitary adenylate cyclase-activating polypeptide-deficient mice. Am J Reprod Immunol 2019; 83:e13212. [PMID: 31758623 DOI: 10.1111/aji.13212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/17/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022] Open
Abstract
PROBLEM Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide having several regulatory functions in the nervous system and in peripheral organs including those of the reproductive system. PACAP-deficient male mice have several morphological, biochemical, behavioral defects and show disturbed signaling in spermatogenesis affecting fertility in PACAP KO mice. Reproductive functions such as fertility, mating, and maternal behaviors have been widely investigated, but no immune analyses are available regarding the testicular immune-privileged environment in male PACAP-deficient mice. METHOD OF STUDY We performed detailed immunophenotyping of testicular immune cells and investigated the expression of TIM-3 and PD-1 Immune checkpoint molecules of immune cells together with the detection of galectin-9 and perforin. We investigated the percentage of numerous immune cell populations in the testis of wild-type and PACAP-deficient mice. RESULTS We demonstrated a significant increase in the frequency of testicular CD8+ T cells together with the decrease in Treg cell number obtained from PACAP KO mice compared with wild-type mice. Investigating Immune checkpoint receptors, only PD-1 showed a significantly decreased expression in CD8+ T cells in PACAP KO mice compared with wild-type suggesting an impaired PD-1/PD-L1 pathway. Regarding TIM-3 expression, we did not find any significant difference between the investigated groups. CONCLUSION We hypothesize that these local changes may result in an immune activation with disturbed testicular immunoregulation in PACAP KO mice; however, determining the exact function requires further investigations. Our data further support the view that besides a systemic immune tolerance, localized active immunosuppression is involved in the regulation of testicular immune privilege.
Collapse
Affiliation(s)
- Matyas Meggyes
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, Pecs, Hungary.,Janos Szentagothai Research Center, Pecs, Hungary
| | - Adrienn Lajko
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, Pecs, Hungary
| | - Balazs Daniel Fulop
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, Medical School, University of Pecs, Pecs, Hungary
| | - Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, Medical School, University of Pecs, Pecs, Hungary
| | - Laszlo Szereday
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, Pecs, Hungary.,Janos Szentagothai Research Center, Pecs, Hungary
| |
Collapse
|
49
|
Moore AM, Coolen LM, Lehman MN. Kisspeptin/Neurokinin B/Dynorphin (KNDy) cells as integrators of diverse internal and external cues: evidence from viral-based monosynaptic tract-tracing in mice. Sci Rep 2019; 9:14768. [PMID: 31611573 PMCID: PMC6791851 DOI: 10.1038/s41598-019-51201-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/25/2019] [Indexed: 02/01/2023] Open
Abstract
Neurons in the hypothalamic arcuate nucleus (ARC) that co-express kisspeptin, neurokinin B and dynorphin (KNDy cells) are essential for mammalian reproduction as key regulators of gonadotropin-releasing hormone (GnRH) secretion. Although multiple endogenous and exogenous signals act indirectly via KNDy neurons to regulate GnRH, the identity of upstream neurons that provide synaptic input to this subpopulation is unclear. We used rabies-mediated tract-tracing in transgenic Kiss1-Cre mice combined with whole-brain optical clearing and multiple-label immunofluorescence to create a comprehensive and quantitative brain-wide map of neurons providing monosynaptic input to KNDy cells, as well as identify the estrogen receptor content and peptidergic phenotype of afferents. Over 90% of monosynaptic input to KNDy neurons originated from hypothalamic nuclei in both male and female mice. The greatest input arose from non-KNDy ARC neurons, including proopiomelanocortin-expressing cells. Significant female-dominant sex differences in afferent input were detected from estrogen-sensitive hypothalamic nuclei critical for reproductive endocrine function and sexual behavior in mice, indicating KNDy cells may provide a unique site for the coordination of sex-specific behavior and gonadotropin release. These data provide key insight into the structural framework underlying the ability of KNDy neurons to integrate endogenous and environmental signals important for the regulation of reproductive function.
Collapse
Affiliation(s)
- Aleisha M Moore
- Brain Health Research Institute and Dept. of Biological Sciences, Kent State University, Kent, OH, USA.
| | - Lique M Coolen
- Brain Health Research Institute and Dept. of Biological Sciences, Kent State University, Kent, OH, USA
| | - Michael N Lehman
- Brain Health Research Institute and Dept. of Biological Sciences, Kent State University, Kent, OH, USA
| |
Collapse
|
50
|
Bohlen TM, Zampieri TT, Furigo IC, Teixeira PDS, List EO, Kopchick JJ, Donato J, Frazao R. Central growth hormone signaling is not required for the timing of puberty. J Endocrinol 2019; 243:JOE-19-0242.R1. [PMID: 31470413 PMCID: PMC6994354 DOI: 10.1530/joe-19-0242] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/30/2019] [Indexed: 12/11/2022]
Abstract
Growth hormone (GH) is a key factor in the regulation of body growth, as well as a variety of other cellular and metabolic processes. Neurons expressing kisspeptin and leptin receptors (LepR) have been shown to modulate the hypothalamic-pituitary-gonadal (HPG) axis and are considered GH-responsive. The presence of functional GH receptors (GHR) in these neural populations suggests that GH may regulate the HPG axis via a central mechanism. However, there have been no studies evaluating whether or not GH-induced intracellular signaling in the brain plays a role in the timing of puberty or mediates the ovulatory cycle. Towards the goal of understanding the influence of GH on the central nervous system as a mediator of reproductive functions, GHR ablation was induced in kisspeptin and LepR expressing cells or in the entire brain. The results demonstrated that GH signaling in specific neural populations can potentially modulate the hypothalamic expression of genes related to the reproductive system or indirectly contribute to the progression of puberty. GH action in kisspeptin cells or in the entire brain was not required for sexual maturation. On the other hand, GHR ablation in LepR cells delayed puberty progression, reduced serum leptin levels, decreased body weight gain and compromised the ovulatory cycle in some individuals, while the lack of GH effects in the entire brain prompted shorter estrous cycles. These findings suggest that GH can modulate brain components of the HPG axis, although central GH signaling is not required for the timing of puberty.
Collapse
Affiliation(s)
- Tabata M Bohlen
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP – Brazil
| | - Thais T Zampieri
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP – Brazil
| | - Isadora C. Furigo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP – Brazil
| | - Pryscila DS Teixeira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP – Brazil
| | - Edward O. List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701 – USA
| | - John J. Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701 – USA
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP – Brazil
| | - Renata Frazao
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP – Brazil
| |
Collapse
|