1
|
Cui X, Chen L, Tao B, Zhang X, Song Y, Chen J, Duan M, Li W, Chen K, Pei Y, Hu X, Feng K, Luo D, Luo H, Qiao Z, Zhou F, Zhu Z, Trudeau VL, Hu W. Olfactory GnRH3 crypt sensory neurons transduce sex pheromone signals to induce male courtship behavior in zebrafish. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-025-2917-5. [PMID: 40347216 DOI: 10.1007/s11427-025-2917-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 03/25/2025] [Indexed: 05/12/2025]
Abstract
Olfactory activation of neuroendocrine pathways plays vital roles in many organisms for reproduction and survival. The importance of gonadotropin-releasing hormone (GnRH) neurons for reproduction is well-established but little is known about whether they can directly sense and transmit sex pheromone signals. We have uncovered the migration path and distribution pattern of a new GnRH neuronal population that fulfills this role. GnRH3 neurons arise from the region located beneath olfactory placode, undergo bidirectional migration along the olfactory nerve, and cell bodies lie within the olfactory epithelium, olfactory bulb and hypothalamus. These olfactory epithelial GnRH3 neurons express ora4, the olfactory receptor that detects pheromones. GnRH3-OB neurons with olfactory epithelial GnRH3 neurons ablation failed to respond to the waterborne post-ovulatory sex pheromone prostaglandin F2α (PGF2α). GnRH3 neurons in gnrh3-/- mutants have a reduced basal firing rate leading to abnormal responses to PGF2α. Male gnrh3-/- zebrafish exhibit deficiencies in courtship behavior and a decreased capacity to compete and spawn with females. These findings indicate that GnRH3-OE neurons function as crypt sensory neurons transducing sex pheromone-encoded information critical to reproductive success.
Collapse
Affiliation(s)
- Xuefan Cui
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lu Chen
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Binbin Tao
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
| | - Xiya Zhang
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanlong Song
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
| | - Ji Chen
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
| | - Ming Duan
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
| | - Weiwei Li
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kuangxin Chen
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
| | - Yang Pei
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuerui Hu
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
| | - Ke Feng
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
| | - Daji Luo
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
| | - Hongrui Luo
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
| | - Zhixian Qiao
- Analytical and Testing Center, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Fang Zhou
- Analytical and Testing Center, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Zuoyan Zhu
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China.
| | - Vance L Trudeau
- Department of Biology, University of Ottawa, Ottawa, K1N 6N5, Canada.
| | - Wei Hu
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
2
|
Carta I, Arora T, Lutzu S, Podda G, Vera Ortega GN, Rudolph S, Autry AE. Sex-specific hypothalamic neural projection activity drives caregiving in mice. Nat Commun 2025; 16:4116. [PMID: 40316517 PMCID: PMC12048593 DOI: 10.1038/s41467-025-59352-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 04/18/2025] [Indexed: 05/04/2025] Open
Abstract
Infant care is critical for survival and healthy development. In laboratory mice, unmated males and females display infant-directed behavior ranging from neglect and aggression to alloparental care. Previous research suggests that excitatory neurons in the perifornical area of the hypothalamus (PeFA) mediate pup-directed aggression. Because medial preoptic area galanin-expressing (MPOAGal) neurons are indispensable for caregiving, we hypothesized that inhibitory MPOAGal projections to PeFA prevent pup-directed aggression. We found that MPOAGal→PeFA projection activity increased during pup approach in both sexes, and alloparental females showed increased projection activity during pup grooming compared to males. Anatomical differences did not explain this disparity in activity between sexes. Optogenetic inhibition of MPOAGal→PeFA projections reduced pup grooming in alloparental females but did not affect male caregiving, while projection stimulation reduced infant-directed aggression in males. Altogether, we show that this projection holds greater significance for female caregiving due to its heightened activity during natural behavior toward pups.
Collapse
Affiliation(s)
- Ilaria Carta
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tushar Arora
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Stefano Lutzu
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Giovanni Podda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Stephanie Rudolph
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anita E Autry
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
3
|
Cavalcante JC, da Silva FG, Sáenz de Miera C, Elias CF. The ventral premammillary nucleus at the interface of environmental cues and social behaviors. Front Neurosci 2025; 19:1589156. [PMID: 40276575 PMCID: PMC12018337 DOI: 10.3389/fnins.2025.1589156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 03/28/2025] [Indexed: 04/26/2025] Open
Abstract
The survival of species heavily depends on social behaviors, which in turn rely on the ability to recognize conspecifics within an appropriate environmental context. These behaviors are regulated by the hypothalamus, which processes signals from both the external environment (such as food availability, photoperiod, and chemical cues from other animals) and the internal state (including sex, estrous cycle stage, nutritional status, and levels of stress). Understanding the brain circuits responsible for specific behaviors in experimental animals is a complex task given the intricate interactions between these factors and the diverse behavioral strategies employed by different species. In this review, we will critically evaluate recent studies focused on the ventral premammillary nucleus (PMv) and discuss findings that reveal the PMv as a key, yet sometimes overlooked, node in integrating external and internal environmental cues. We will examine its structural components, internal connectivity, humoral influences, and associated functions, demonstrating the PMv role in the neural regulation of neuroendocrine responses and social behaviors. While much of the existing research centers on rats and mice as model organisms, we will highlight relevant species differences and include a dedicated section for findings in other species.
Collapse
Affiliation(s)
- Judney Cley Cavalcante
- Laboratory of Neuroanatomy, Department of Morphology, Center of Biosciences, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Fabiano Gomes da Silva
- Laboratory of Neuroanatomy, Department of Morphology, Center of Biosciences, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Cristina Sáenz de Miera
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Carol Fuzeti Elias
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
4
|
Cotellessa L, Sobrino V, Silva MSB, Delit M, Maitre H, Caron E, Ternier G, da Silva Lima N, Lhomme T, Giton F, Sorrentino A, Carraresi L, Di Nardo G, Nogueiras R, Tena-Sempere M, Prevot V, Giacobini P. Preventing and correcting polycystic ovary syndrome by targeting anti-Müllerian hormone signaling in minipuberty and adulthood in mice. Cell Metab 2025:S1550-4131(25)00116-0. [PMID: 40220763 DOI: 10.1016/j.cmet.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 02/03/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
Polycystic ovary syndrome (PCOS), the most common endocrinopathy in women, causes significant reproductive and metabolic comorbidities, with no current cure. Gestational androgen and anti-Müllerian hormone (AMH) excess are linked to PCOS, and prenatal aberrant exposure to these hormones induces PCOS-like traits in animal models. However, whether the AMH effects on PCOS programming could extend to early postnatal life remains unknown. Clinical observations showed higher AMH levels during minipuberty in infants of mothers with PCOS, but whether this contributes to PCOS development is uncertain. Here, we show that exposure to high AMH levels during minipuberty in mice causes PCOS-like reproductive and metabolic defects in both sexes. A neutralizing antibody targeting AMH receptor 2 (AMHR2) prevented these defects when administered during minipuberty and alleviated symptoms when given in adulthood. These findings highlight the causal role of elevated AMH in PCOS and suggest AMHR2-targeting therapy as a potential preventive or curative approach.
Collapse
Affiliation(s)
- Ludovica Cotellessa
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Veronica Sobrino
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC) and Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
| | - Mauro S B Silva
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Maxime Delit
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Hélène Maitre
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Emilie Caron
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Gaëtan Ternier
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Natalia da Silva Lima
- CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Tori Lhomme
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Frank Giton
- IMRB - Inserm U955, Faculté de Santé, Créteil, France
| | - Andrea Sorrentino
- DIVAL Toscana Srl, Sesto Fiorentino, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laura Carraresi
- DIVAL Toscana Srl, Sesto Fiorentino, Florence, Italy; Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Giovanna Di Nardo
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| | - Ruben Nogueiras
- CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Manuel Tena-Sempere
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC) and Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
| | - Vincent Prevot
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Paolo Giacobini
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France.
| |
Collapse
|
5
|
Mikulovic S, Lenschow C. Neural control of sex differences in affiliative and prosocial behaviors. Neurosci Biobehav Rev 2025; 171:106039. [PMID: 39914700 DOI: 10.1016/j.neubiorev.2025.106039] [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: 06/12/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 03/01/2025]
Abstract
Social interactions are vital for various taxa and species. Prosocial and affiliative dynamics within a group and between individuals are not only pleasurable and rewarding, but also appear to actively contribute to well-being, cognitive performance, and disease prevention. Moreover, disturbances in acting or being prosocial can represent a major burden for an individual and their affective partners. These disruptions are evident across a spectrum of neuropsychiatric conditions, including depression and autism spectrum disorders. Importantly, interactive patterns of prosocial and affiliative behavior can vary with sex. The fact that genders are differentially affected by neuropsychiatric disorders associated with social impairment underscores the high importance of this research in uncovering the underlying neural correlates and mechanisms. This review focuses on elucidating sex-related differences in prosocial and affiliative behaviors and their potential association with sexually different neural correlates. Specifically, we aim to shed light on the complex interplay between sex, behavior, and neurobiology in affiliative and prosocial interaction patterns.
Collapse
Affiliation(s)
- Sanja Mikulovic
- Leibniz Institute for Neurobiology, Brennecke Straße, Magdeburg, Germany.
| | - Constanze Lenschow
- Otto-von-Guericke University Magdeburg, Institute of Biology (House 91), Leipziger Straße 44, Magdeburg 39120, Germany.
| |
Collapse
|
6
|
Kaplan HS, Logeman BL, Zhang K, Yawitz TA, Santiago C, Sohail N, Talay M, Seo C, Naumenko S, Ho Sui SJ, Ginty DD, Ren B, Dulac C. Sensory input, sex and function shape hypothalamic cell type development. Nature 2025:10.1038/s41586-025-08603-0. [PMID: 40044853 DOI: 10.1038/s41586-025-08603-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 12/12/2024] [Indexed: 03/12/2025]
Abstract
Mammalian behaviour and physiology undergo major changes in early life. Young animals rely on conspecifics to meet their needs and start showing nutritional independence and sex-specific social interactions at weaning and puberty, respectively. How neuronal populations regulating homeostatic functions and social behaviours develop during these transitions remains unclear. We used paired transcriptomic and chromatin accessibility profiling to examine the developmental trajectories of neuronal populations in the hypothalamic preoptic region, where cell types with key roles in physiological and behavioural control have been identified1-6. These data show a marked diversity of developmental trajectories shaped by the sex of the animal, and the location and behavioural or physiological function of the corresponding cell types. We identify key stages of preoptic development, including early diversification, perinatal emergence of sex differences, postnatal maturation and refinement of signalling networks, and nonlinear transcriptional changes accelerating at the time of weaning and puberty. We assessed preoptic development in various sensory mutants and find a major role for vomeronasal sensing in the timing of preoptic cell type maturation. These results provide new insights into the development of neurons controlling homeostatic functions and social behaviours and lay ground for examining the dynamics of these functions in early life.
Collapse
Affiliation(s)
- Harris S Kaplan
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Brandon L Logeman
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Kai Zhang
- Department of Cellular and Molecular Medicine, Center for Epigenomics, University of California, San Diego School of Medicine, La Jolla, CA, USA
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
| | - Tate A Yawitz
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Celine Santiago
- Department of Neurobiology, Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, USA
| | - Noor Sohail
- Department of Biostatistics, Harvard Chan School of Public Health, Boston, MA, USA
| | - Mustafa Talay
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Changwoo Seo
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Serhiy Naumenko
- Department of Biostatistics, Harvard Chan School of Public Health, Boston, MA, USA
- Newborn Screening Ontario, Ottawa, Ontario, Canada
| | - Shannan J Ho Sui
- Department of Biostatistics, Harvard Chan School of Public Health, Boston, MA, USA
| | - David D Ginty
- Department of Neurobiology, Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, USA
| | - Bing Ren
- Department of Cellular and Molecular Medicine, Center for Epigenomics, University of California, San Diego School of Medicine, La Jolla, CA, USA
| | - Catherine Dulac
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
7
|
Kacimi L, Prevot V. GnRH and Cognition. Endocrinology 2025; 166:bqaf033. [PMID: 39996304 DOI: 10.1210/endocr/bqaf033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/06/2025] [Accepted: 02/23/2025] [Indexed: 02/26/2025]
Abstract
GnRH is traditionally recognized as the central regulator of reproduction through its pulsatile secretion, which governs the hypothalamic-pituitary-gonadal axis. However, recent evidence has highlighted its broader role in brain development and function, including in cognitive and higher intellectual processes. GnRH production follows distinct phases, from its early activation during minipuberty-the first postnatal activation of GnRH neurons during the infantile period-, its reactivation and stabilization starting at puberty, and its eventual decline with age and the loss of gonadal steroid feedback. This evolution depends on the establishment, maturation and activation of GnRH neurons, a complex process regulated by the cellular and molecular environment of these neurons, including multiple neuronal and glial types as well as a minipubertal "switch" in gene expression, the perturbation of which may have long-term or delayed consequences for both reproductive and cognitive function. The cognitive role of GnRH may be related to its recently revealed involvement in maintaining myelination and synaptic plasticity, whereas disruptions in its finely tuned rhythmic secretion, either age-related or pathological, are associated with cognitive decline and neurodegenerative disorders. Restoring physiological GnRH levels and pulsatility can reverse age-related cognitive decline and improve sensory functions even in adulthood, suggesting a mobilization of the "cognitive reserve" in both animal models and human patients. This review highlights recent advances in our understanding of the GnRH system and the therapeutic potential of pulsatile GnRH therapy to mitigate age-related cognitive decline and neurodegenerative processes.
Collapse
Affiliation(s)
- Loïc Kacimi
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, FHU 1000 days for health, EGID, DistALZ, UMR_S112, Lille 59000, France
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, FHU 1000 days for health, EGID, DistALZ, UMR_S112, Lille 59000, France
| |
Collapse
|
8
|
Yeo SH, Han SY, Herbison AE. Shifting GnRH Neuron Ensembles Underlie Successive Preovulatory Luteinizing Hormone Surges. J Neurosci 2025; 45:e1383242024. [PMID: 39505408 PMCID: PMC11735651 DOI: 10.1523/jneurosci.1383-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/30/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024] Open
Abstract
The gonadotropin-releasing hormone (GnRH) neurons operate as a neuronal ensemble exhibiting coordinated activity once every reproductive cycle to generate the preovulatory GnRH surge. Using GCaMP fiber photometry at the GnRH neuron distal dendrons to measure the output of this widely scattered population in female mice, we find that the onset, amplitude, and profile of GnRH neuron surge activity exhibits substantial variability from cycle to cycle both between and within individual mice. This was also evident when measuring successive proestrous luteinizing hormone surges. Studies combining short (c-Fos and c-Jun) and long (genetic robust activity marking) term indices of immediate early gene activation revealed that, while ∼50% of GnRH neurons were activated at the time of each surge, only half of these neurons had been active during the previous proestrous surge. These observations reveal marked inter- and intra-individual variability in the GnRH surge mechanism. Remarkably, different subpopulations of overlapping GnRH neurons are recruited to the ensemble each estrous cycle to generate the GnRH surge. While engendering variability in the surge mechanism itself, this likely provides substantial robustness to a key event underlying mammalian reproduction.
Collapse
Affiliation(s)
- Shel-Hwa Yeo
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | - Su Young Han
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | - Allan E Herbison
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| |
Collapse
|
9
|
Han SY, Yeo SH, Kim JC, Zhou Z, Herbison AE. Multi-dimensional oscillatory activity of mouse GnRH neurons in vivo. eLife 2025; 13:RP100856. [PMID: 39773874 PMCID: PMC11709428 DOI: 10.7554/elife.100856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
The gonadotropin-releasing hormone (GnRH) neurons represent the key output cells of the neural network controlling mammalian fertility. We used GCaMP fiber photometry to record the population activity of the GnRH neuron distal projections in the ventral arcuate nucleus where they merge before entering the median eminence to release GnRH into the portal vasculature. Recordings in freely behaving intact male and female mice revealed abrupt ~8 min duration increases in activity that correlated perfectly with the appearance of a subsequent pulse of luteinizing hormone (LH). The GnRH neuron dendrons also exhibited a low level of unchanging clustered, rapidly fluctuating baseline activity in males and throughout the estrous cycle in females. In female mice, a gradual increase in basal activity that exhibited ~80 min oscillations began in the afternoon of proestrus and lasted for 12 hr. This was associated with the onset of the LH surge that ended several hours before the fall in the GCaMP signal. Abrupt 8 min duration episodes of GCaMP activity continued to occur on top of the rising surge baseline before ceasing in estrus. These observations provide the first description of GnRH neuron activity in freely behaving animals. They demonstrate that three distinct patterns of oscillatory activity occur in GnRH neurons. These are comprised of low-level rapid baseline activity, abrupt 8 min duration oscillations that drive pulsatile gonadotropin secretion, and, in females, a gradual and very prolonged oscillating increase in activity responsible for the preovulatory LH surge.
Collapse
Affiliation(s)
- Su Young Han
- Department of Physiology, Development and Neuroscience, Downing site, University of CambridgeCambridgeUnited Kingdom
| | - Shel-Hwa Yeo
- Department of Physiology, Development and Neuroscience, Downing site, University of CambridgeCambridgeUnited Kingdom
| | - Jae-Chang Kim
- Zurich Center for Neuroeconomics, Department of Economics, University of ZurichZurichSwitzerland
| | - Ziyue Zhou
- Department of Physiology, Development and Neuroscience, Downing site, University of CambridgeCambridgeUnited Kingdom
| | - Allan E Herbison
- Department of Physiology, Development and Neuroscience, Downing site, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
10
|
Elango K, Kekäläinen J. Putting Nose into Reproduction: Influence of Nasal and Reproductive Odourant Signaling on Male Reproduction. Mol Reprod Dev 2025; 92:e70010. [PMID: 39834068 DOI: 10.1002/mrd.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
Odourant receptors (ORs) are not restricted only to the nose, but also occur in many other organs and tissues, including the reproductive system. In fact, ORs are the most heavily expressed in testis than in any other extra-nasal tissue. Accumulating evidence suggests that olfactory and reproductive systems are both structurally and functionally linked and that these interconnections can influence various aspects of reproduction. In this article, we first review our current understanding of these interconnections and then collate accumulated evidence on the presence of ORs in the male reproductive system and sperm cells. We then investigate the potential role of female reproductive tract odourants in sperm chemotaxis and selection. Finally, since the existing evidence especially for sperm odor sensing capability and its physiological function are controversial, we also review potential reasons for the controversy and propose some ways to resolve the debate. Collectively, we conclude that reproductive odourant signaling may play an important, although currently largely unclear role in many key processes directly related to male fertility. However, since we lack holistic understanding of the functional significance of ORs and odor sensing pathways of the male reproductive system, more empirical research is warranted.
Collapse
Affiliation(s)
- Kamaraj Elango
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| | - Jukka Kekäläinen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| |
Collapse
|
11
|
Papes F, Nakahara TS, Camargo AP. Behavioral Assays in the Study of Olfaction. Methods Mol Biol 2025; 2915:213-314. [PMID: 40249495 DOI: 10.1007/978-1-0716-4466-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Olfaction is a fundamental sense in most animal species. In mammals, the olfactory system comprises several subpopulations of sensory neurons located throughout the nasal cavity, which detect a variety of chemical stimuli, including odorants, intraspecies, and interspecies chemical communication cues. Some of these compounds are important for regulating innate or learned behaviors and endocrine changes in response to other animals in the environment. With a particular focus on laboratory rodent species, this chapter provides a comprehensive description of the most important behavioral assays used for studying the olfactory system and is meant to be a practical guide for those who investigate olfaction-mediated behaviors or who have an interest in deciphering the molecular, cellular, or neural mechanisms through which the sense of smell controls the generation of adaptive behavioral outputs.
Collapse
Affiliation(s)
- Fabio Papes
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, SP, Brazil.
| | - Thiago S Nakahara
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Antonio P Camargo
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
12
|
Li CY, Bowers JM, Alexander TA, Behrens KA, Jackson P, Amini CJ, Juntti SA. A pheromone receptor in cichlid fish mediates attraction to females but inhibits male parental care. Curr Biol 2024; 34:3866-3880.e7. [PMID: 39094572 PMCID: PMC11387146 DOI: 10.1016/j.cub.2024.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/24/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
Reproductive behaviors differ across species, but the mechanisms that control variation in mating and parental care systems remain unclear. In many animal species, pheromones guide mating and parental care. However, it is not well understood how vertebrate pheromone signaling evolution can lead to new reproductive behavior strategies. In fishes, prostaglandin F2α (PGF2α) drives mating and reproductive pheromone signaling in fertile females, but this pheromonal activity appears restricted to specific lineages, and it remains unknown how a female fertility pheromone is sensed for most fish species. Here, we utilize single-cell transcriptomics and CRISPR gene editing in a cichlid fish model to identify and test the roles of key genes involved in olfactory sensing of reproductive cues. We find that a pheromone receptor, Or113a, detects fertile cichlid females and thereby promotes male attraction and mating behavior, sensing a ligand other than PGF2α. Furthermore, while cichlid fishes exhibit extensive parental care, for most species, care is provided solely by females. We find that males initiate mouthbrooding parental care if they have disrupted signaling in ciliated sensory neurons due to cnga2b mutation or if or113a is inactivated. Together, these results show that distinct mechanisms of pheromonal signaling drive reproductive behaviors across taxa. Additionally, these findings indicate that a single pheromone receptor has gained a novel role in behavior regulation, driving avoidance of paternal care among haplochromine cichlid fishes. Lastly, a sexually dimorphic, evolutionarily derived parental behavior is controlled by central circuits present in both sexes, while olfactory signals gate this behavior in a sex-specific manner.
Collapse
Affiliation(s)
- Cheng-Yu Li
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Jessica M Bowers
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | | | - Kristen A Behrens
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Peter Jackson
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Cyrus J Amini
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Scott A Juntti
- Department of Biology, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
13
|
Decoster L, Trova S, Zucca S, Bulk J, Gouveia A, Ternier G, Lhomme T, Legrand A, Gallet S, Boehm U, Wyatt A, Wahl V, Wartenberg P, Hrabovszky E, Rácz G, Luzzati F, Nato G, Fogli M, Peretto P, Schriever SC, Bernecker M, Pfluger PT, Steculorum SM, Bovetti S, Rasika S, Prevot V, Silva MSB, Giacobini P. A GnRH neuronal population in the olfactory bulb translates socially relevant odors into reproductive behavior in male mice. Nat Neurosci 2024; 27:1758-1773. [PMID: 39095587 DOI: 10.1038/s41593-024-01724-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
Hypothalamic gonadotropin-releasing hormone (GnRH) neurons regulate fertility and integrate hormonal status with environmental cues to ensure reproductive success. Here we show that GnRH neurons in the olfactory bulb (GnRHOB) of adult mice can mediate social recognition. Specifically, we show that GnRHOB neurons extend neurites into the vomeronasal organ and olfactory epithelium and project to the median eminence. GnRHOB neurons in males express vomeronasal and olfactory receptors, are activated by female odors and mediate gonadotropin release in response to female urine. Male preference for female odors required the presence and activation of GnRHOB neurons, was impaired after genetic inhibition or ablation of these cells and relied on GnRH signaling in the posterodorsal medial amygdala. GnRH receptor expression in amygdala kisspeptin neurons appear to be required for GnRHOB neurons' actions on male mounting behavior. Taken together, these results establish GnRHOB neurons as regulating fertility, sex recognition and mating in male mice.
Collapse
Affiliation(s)
- Laurine Decoster
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Sara Trova
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
- Centro CMP3VdA, Istituto Italiano di Tecnologia (IIT), Aosta, Italy
| | - Stefano Zucca
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Janice Bulk
- Max Planck Institute for Metabolism Research, Max Planck Research Group Neurocircuit Wiring and Function, Cologne, Germany
| | - Ayden Gouveia
- Max Planck Institute for Metabolism Research, Max Planck Research Group Neurocircuit Wiring and Function, Cologne, Germany
| | - Gaetan Ternier
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Tori Lhomme
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Amandine Legrand
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Sarah Gallet
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Amanda Wyatt
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Vanessa Wahl
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Philipp Wartenberg
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Hun-Ren Institute of Experimental Medicine, Budapest, Hungary
| | - Gergely Rácz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Federico Luzzati
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Giulia Nato
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Marco Fogli
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Paolo Peretto
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Sonja C Schriever
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Research Unit Neurobiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
| | - Miriam Bernecker
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Research Unit Neurobiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- Division of Neurobiology of Diabetes, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Paul T Pfluger
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Research Unit Neurobiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- Division of Neurobiology of Diabetes, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Sophie M Steculorum
- Max Planck Institute for Metabolism Research, Max Planck Research Group Neurocircuit Wiring and Function, Cologne, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Serena Bovetti
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Sowmyalakshmi Rasika
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Mauro S B Silva
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Paolo Giacobini
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France.
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France.
| |
Collapse
|
14
|
Ow MC, Nishiguchi MA, Dar AR, Butcher RA, Hall SE. RNAi-dependent expression of sperm genes in ADL chemosensory neurons is required for olfactory responses in Caenorhabditis elegans. Front Mol Biosci 2024; 11:1396587. [PMID: 39055986 PMCID: PMC11269235 DOI: 10.3389/fmolb.2024.1396587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/11/2024] [Indexed: 07/28/2024] Open
Abstract
Environmental conditions experienced early in the life of an animal can result in gene expression changes later in its life history. We have previously shown that C. elegans animals that experienced the developmentally arrested and stress resistant dauer stage (postdauers) retain a cellular memory of early-life stress that manifests during adulthood as genome-wide changes in gene expression, chromatin states, and altered life history traits. One consequence of developmental reprogramming in C. elegans postdauer adults is the downregulation of osm-9 TRPV channel gene expression in the ADL chemosensory neurons resulting in reduced avoidance to a pheromone component, ascr#3. This altered response to ascr#3 requires the principal effector of the somatic nuclear RNAi pathway, the Argonaute (AGO) NRDE-3. To investigate the role of the somatic nuclear RNAi pathway in regulating the developmental reprogramming of ADL due to early-life stress, we profiled the mRNA transcriptome of control and postdauer ADL in wild-type and nrde-3 mutant adults. We found 711 differentially expressed (DE) genes between control and postdauer ADL neurons, 90% of which are dependent upon NRDE-3. Additionally, we identified a conserved sequence that is enriched in the upstream regulatory sequences of the NRDE-3-dependent differentially expressed genes. Surprisingly, 214 of the ADL DE genes are considered "germline-expressed", including 21 genes encoding the Major Sperm Proteins and two genes encoding the sperm-specific PP1 phosphatases, GSP-3 and GSP-4. Loss of function mutations in gsp-3 resulted in both aberrant avoidance and attraction behaviors. We also show that an AGO pseudogene, Y49F6A.1 (wago-11), is expressed in ADL and is required for ascr#3 avoidance. Overall, our results suggest that small RNAs and reproductive genes program the ADL mRNA transcriptome during their developmental history and highlight a nexus between neuronal and reproductive networks in calibrating animal neuroplasticity.
Collapse
Affiliation(s)
- Maria C. Ow
- Biology Department, Syracuse University, Syracuse, NY, United States
| | | | - Abdul Rouf Dar
- Department of Chemistry, University of Florida, Gainesville, FL, United States
| | - Rebecca A. Butcher
- Department of Chemistry, University of Florida, Gainesville, FL, United States
| | - Sarah E. Hall
- Biology Department, Syracuse University, Syracuse, NY, United States
| |
Collapse
|
15
|
Qin P, Pan Z, Zhang W, Wang R, Li X, Lu J, Xu S, Gong X, Ye J, Yan X, Liu Y, Li Y, Zhang Y, Fang F. Integrative proteomic and transcriptomic analysis in the female goat ovary to explore the onset of puberty. J Proteomics 2024; 301:105183. [PMID: 38688390 DOI: 10.1016/j.jprot.2024.105183] [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: 12/10/2023] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
Puberty is considered a prerequisite for affecting reproductive performance and productivity. Little was known about molecular changes in pubertal goat ovaries. Therefore, we measured and performed a correlation analysis of the mRNA and proteins changes in the pre-pubertal and pubertal goat ovaries. The results showed that only six differentially expressed genes and differentially abundant proteins out of 18,139 genes and 7550 proteins quantified had significant correlations. CNTN2 and THBS1, discovered in the mRNA-mRNA interaction network, probably participated in pubertal and reproductive regulation by influencing GnRH receptor signals, follicular development, and ovulation. The predicted core transcription factors may either promote or inhibit the expression of reproductive genes and act synergistically to maintain normal reproductive function in animals. The interaction between PKM and TIMP3 with other proteins may impact animal puberty through energy metabolism and ovarian hormone secretion. Pathway enrichment analyses revealed that the co-associated key pathways between ovarian genes and proteins at puberty included calcium signalling pathway and olfactory transduction. These pathways were associated with gonadotropin-releasing hormone synthesis and secretion, signal transmission, and cell proliferation. In summary, these results enriched the potential molecules and signalling pathways that affect puberty and provided new insights for regulating and promoting the onset of puberty. SIGNIFICANCE: This study conducted the first transcriptomic and proteomic correlation analysis of pre-pubertal and pubertal goat ovaries and identified six significantly correlated molecules at both the gene and protein levels. Meanwhile, we were drawn to several molecules and signalling pathways that may play a regulatory role in the onset of puberty and reproduction by influencing reproductive-related gene expression, GnRH receptor signals, energy metabolism, ovarian hormone secretion, follicular development, and ovulation. This information contributed to identify potential biomarkers in pubertal goat ovaries, which was vital for predicting the onset of puberty and improving livestock performance.
Collapse
Affiliation(s)
- Ping Qin
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Zhihao Pan
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Wei Zhang
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Rui Wang
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xiaoqian Li
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Juntai Lu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Shuangshuang Xu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xinbao Gong
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Jing Ye
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xu Yan
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ya Liu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yunsheng Li
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yunhai Zhang
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Fugui Fang
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China.
| |
Collapse
|
16
|
Cerbantez-Bueno V, Viñuela-Berni V, Muñoz-Mayorga DE, Morales T, Corona R. Prolactin promotes the recruitment of main olfactory bulb cells and enhances the behavioral exploration toward a socio-sexual stimulus in female mice. Horm Behav 2024; 162:105527. [PMID: 38492348 DOI: 10.1016/j.yhbeh.2024.105527] [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: 09/01/2023] [Revised: 11/30/2023] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
Olfactory communication is triggered by pheromones that profoundly influence neuroendocrine responses to drive social interactions. Two principal olfactory systems process pheromones: the main and the vomeronasal or accessory system. Prolactin receptors are expressed in both systems suggesting a participation in the processing of olfactory information. We previously reported that prolactin participates in the sexual and olfactory bulb maturation of females. Therefore, we explored the expression of prolactin receptors within the olfactory bulb during sexual maturation and the direct responses of prolactin upon pheromonal exposure. Additionally, we assessed the behavioral response of adult females exposed to male sawdust after prolactin administration and the consequent activation of main and accessory olfactory bulb and their first central relays, the piriform cortex and the medial amygdala. Last, we investigated the intracellular pathway activated by prolactin within the olfactory bulb. Here, prolactin receptor expression remained constant during all maturation stages within the main olfactory bulb but decreased in adulthood in the accessory olfactory bulb. Behaviorally, females that received prolactin actively explored the male stimulus. An increased cFos activation in the amygdala and in the glomerular cells of the whole olfactory bulb was observed, but an augmented response in the mitral cells was only found within the main olfactory bulb after prolactin administration and the exposure to male stimulus. Interestingly, the ERK pathway was upregulated in the main olfactory bulb after exposure to a male stimulus. Overall, our results suggest that, in female mice, prolactin participates in the processing of chemosignals and behavioral responses by activating the main olfactory system and diminishing the classical vomeronasal response to pheromones.
Collapse
Affiliation(s)
- Viridiana Cerbantez-Bueno
- Laboratorio de Neuroanatomía Funcional y Neuroendocrinología, Instituto de Neurobiología (INB), Universidad Nacional Autónoma de México (UNAM), Juriquilla, Querétaro, Mexico
| | - Verónica Viñuela-Berni
- Laboratorio de Neuroanatomía Funcional y Neuroendocrinología, Instituto de Neurobiología (INB), Universidad Nacional Autónoma de México (UNAM), Juriquilla, Querétaro, Mexico
| | - Daniel Eduardo Muñoz-Mayorga
- Laboratorio de Neuroanatomía Funcional y Neuroendocrinología, Instituto de Neurobiología (INB), Universidad Nacional Autónoma de México (UNAM), Juriquilla, Querétaro, Mexico
| | - Teresa Morales
- Laboratorio de Neuroanatomía Funcional y Neuroendocrinología, Instituto de Neurobiología (INB), Universidad Nacional Autónoma de México (UNAM), Juriquilla, Querétaro, Mexico
| | - Rebeca Corona
- Laboratorio de Neuroanatomía Funcional y Neuroendocrinología, Instituto de Neurobiología (INB), Universidad Nacional Autónoma de México (UNAM), Juriquilla, Querétaro, Mexico.
| |
Collapse
|
17
|
Zhang Y, Luo W, Heinricher MM, Ryabinin AE. CFA-treated mice induce hyperalgesia in healthy mice via an olfactory mechanism. Eur J Pain 2024; 28:578-598. [PMID: 37985943 PMCID: PMC10947942 DOI: 10.1002/ejp.2201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 10/04/2023] [Accepted: 10/24/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Social interactions with subjects experiencing pain can increase nociceptive sensitivity in observers, even without direct physical contact. In previous experiments, extended indirect exposure to soiled bedding from mice with alcohol withdrawal-related hyperalgesia enhanced nociception in their conspecifics. This finding suggested that olfactory cues could be sufficient for nociceptive hypersensitivity in otherwise untreated animals (also known as "bystanders"). AIM The current study addressed this possibility using an inflammation-based hyperalgesia model and long- and short-term exposure paradigms in C57BL/6J mice. MATERIALS & METHOD Adult male and female mice received intraplantar injection of complete Freund's adjuvant (CFA) and were used as stimulus animals to otherwise naïve same-sex bystander mice (BS). Another group of untreated mice (OLF) was simultaneously exposed to the bedding of the stimulus mice. RESULTS In the long-term, 15-day exposure paradigm, the presence of CFA mice or their bedding resulted in reduced von Frey threshold but not Hargreaves paw withdrawal latency in BS or OLF mice. In the short-term paradigm, 1-hr interaction with CFA conspecifics or 1-hr exposure to their bedding induced mechanical hypersensitivity in BS and OLF mice lasting for 3 hrs. Chemical ablation of the main olfactory epithelium prevented bedding-induced and stimulus mice-induced mechanical hypersensitivity. Gas chromatography-mass spectrometry (GC-MS) analysis of the volatile compounds in the bedding of experimental mice revealed that CFA-treated mice released an increased number of compounds indicative of disease states. DISCUSSION AND CONCLUSION These results demonstrate that CFA-induced inflammatory pain can modulate nociception in bystander mice via an olfactory mechanism involving dynamic changes in volatile compounds detectable in the rodent bedding. SIGNIFICANCE Social context can influence nociceptive sensitivity. Recent studies suggested involvement of olfaction in this influence. In agreement with this idea, the present study shows that the presence of mice with inflammatory pain produces nociceptive hypersensitivity in nearby conspecifics. This enhanced nociception occurs via olfactory cues present in the mouse bedding. Analysis of the bedding from mice with inflammatory pain identifies a number of compounds indicative of disease states. These findings demonstrate the importance of olfactory system in influencing pain states.
Collapse
Affiliation(s)
- Yangmiao Zhang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
| | - Wentai Luo
- Department of Chemistry, Portland State University, Portland, OR 97207
| | - Mary M. Heinricher
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR 97239
| | - Andrey E. Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
| |
Collapse
|
18
|
Kaplan HS, Logeman BL, Zhang K, Santiago C, Sohail N, Naumenko S, Ho Sui SJ, Ginty DD, Ren B, Dulac C. Sensory Input, Sex, and Function Shape Hypothalamic Cell Type Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.23.576835. [PMID: 38328205 PMCID: PMC10849564 DOI: 10.1101/2024.01.23.576835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Mammalian behavior and physiology undergo dramatic changes in early life. Young animals rely on conspecifics to meet their homeostatic needs, until weaning and puberty initiate nutritional independence and sex-specific social interactions, respectively. How neuronal populations regulating homeostatic functions and social behaviors develop and mature during these transitions remains unclear. We used paired transcriptomic and chromatin accessibility profiling to examine the developmental trajectories of neuronal populations in the hypothalamic preoptic region, where cell types with key roles in physiological and behavioral control have been identified1-6. These data reveal a remarkable diversity of developmental trajectories shaped by the sex of the animal, and the location and behavioral or physiological function of the corresponding cell types. We identify key stages of preoptic development, including the perinatal emergence of sex differences, postnatal maturation and subsequent refinement of signaling networks, and nonlinear transcriptional changes accelerating at the time of weaning and puberty. We assessed preoptic development in various sensory mutants and find a major role for vomeronasal sensing in the timing of preoptic cell type maturation. These results provide novel insights into the development of neurons controlling homeostatic functions and social behaviors and lay ground for examining the dynamics of these functions in early life.
Collapse
Affiliation(s)
- Harris S. Kaplan
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Brandon L. Logeman
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Kai Zhang
- Department of Cellular and Molecular Medicine, Center for Epigenomics, University of California, San Diego School of Medicine, La Jolla, CA 92093, USA
- Current address: Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
| | - Celine Santiago
- Department of Neurobiology, Harvard Medical School, Howard Hughes Medical Institute, 220 Longwood Ave, Boston, MA, 02115, USA
| | - Noor Sohail
- Department of Biostatistics, Harvard Chan School of Public Health, Boston, MA, USA
| | - Serhiy Naumenko
- Department of Biostatistics, Harvard Chan School of Public Health, Boston, MA, USA
- Newborn Screening Ontario, Ottawa, ON, Canada
| | - Shannan J. Ho Sui
- Department of Biostatistics, Harvard Chan School of Public Health, Boston, MA, USA
| | - David D. Ginty
- Department of Neurobiology, Harvard Medical School, Howard Hughes Medical Institute, 220 Longwood Ave, Boston, MA, 02115, USA
| | - Bing Ren
- Department of Cellular and Molecular Medicine, Center for Epigenomics, University of California, San Diego School of Medicine, La Jolla, CA 92093, USA
| | - Catherine Dulac
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| |
Collapse
|
19
|
Menon R, Neumann ID. Detection, processing and reinforcement of social cues: regulation by the oxytocin system. Nat Rev Neurosci 2023; 24:761-777. [PMID: 37891399 DOI: 10.1038/s41583-023-00759-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 10/29/2023]
Abstract
Many social behaviours are evolutionarily conserved and are essential for the healthy development of an individual. The neuropeptide oxytocin (OXT) is crucial for the fine-tuned regulation of social interactions in mammals. The advent and application of state-of-the-art methodological approaches that allow the activity of neuronal circuits involving OXT to be monitored and functionally manipulated in laboratory mammals have deepened our understanding of the roles of OXT in these behaviours. In this Review, we discuss how OXT promotes the sensory detection and evaluation of social cues, the subsequent approach and display of social behaviour, and the rewarding consequences of social interactions in selected reproductive and non-reproductive social behaviours. Social stressors - such as social isolation, exposure to social defeat or social trauma, and partner loss - are often paralleled by maladaptations of the OXT system, and restoring OXT system functioning can reinstate socio-emotional allostasis. Thus, the OXT system acts as a dynamic mediator of appropriate behavioural adaptations to environmental challenges by enhancing and reinforcing social salience and buffering social stress.
Collapse
Affiliation(s)
- Rohit Menon
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - Inga D Neumann
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
20
|
Mishra S, Dabaja M, Akhlaq A, Pereira B, Marbach K, Rovcanin M, Chandra R, Caballero A, Fernandes de Abreu D, Ch'ng Q, Alcedo J. Specific sensory neurons and insulin-like peptides modulate food type-dependent oogenesis and fertilization in Caenorhabditis elegans. eLife 2023; 12:e83224. [PMID: 37975568 PMCID: PMC10665013 DOI: 10.7554/elife.83224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/13/2023] [Indexed: 11/19/2023] Open
Abstract
An animal's responses to environmental cues are critical for its reproductive program. Thus, a mechanism that allows the animal to sense and adjust to its environment should make for a more efficient reproductive physiology. Here, we demonstrate that in Caenorhabditis elegans specific sensory neurons influence onset of oogenesis through insulin signaling in response to food-derived cues. The chemosensory neurons ASJ modulate oogenesis onset through the insulin-like peptide (ILP) INS-6. In contrast, other sensory neurons, the olfactory neurons AWA, regulate food type-dependent differences in C. elegans fertilization rates, but not onset of oogenesis. AWA modulates fertilization rates at least partly in parallel to insulin receptor signaling, since the insulin receptor DAF-2 regulates fertilization independently of food type, which requires ILPs other than INS-6. Together our findings suggest that optimal reproduction requires the integration of diverse food-derived inputs through multiple neuronal signals acting on the C. elegans germline.
Collapse
Affiliation(s)
- Shashwat Mishra
- Department of Biological Sciences, Wayne State UniversityDetroitUnited States
| | - Mohamed Dabaja
- Department of Biological Sciences, Wayne State UniversityDetroitUnited States
| | - Asra Akhlaq
- Department of Biological Sciences, Wayne State UniversityDetroitUnited States
| | - Bianca Pereira
- Department of Biological Sciences, Wayne State UniversityDetroitUnited States
| | - Kelsey Marbach
- Department of Biological Sciences, Wayne State UniversityDetroitUnited States
| | - Mediha Rovcanin
- Department of Biological Sciences, Wayne State UniversityDetroitUnited States
| | - Rashmi Chandra
- Department of Biological Sciences, Wayne State UniversityDetroitUnited States
| | - Antonio Caballero
- Centre for Developmental Neurobiology, King’s College LondonLondonUnited Kingdom
| | | | - QueeLim Ch'ng
- Centre for Developmental Neurobiology, King’s College LondonLondonUnited Kingdom
| | - Joy Alcedo
- Department of Biological Sciences, Wayne State UniversityDetroitUnited States
| |
Collapse
|
21
|
Silva MSB, Decoster L, Delpouve G, Lhomme T, Ternier G, Prevot V, Giacobini P. Overactivation of GnRH neurons is sufficient to trigger polycystic ovary syndrome-like traits in female mice. EBioMedicine 2023; 97:104850. [PMID: 37898094 PMCID: PMC10630624 DOI: 10.1016/j.ebiom.2023.104850] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/18/2023] [Accepted: 10/12/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is the most common endocrine disorder leading to anovulatory infertility. Abnormalities in the central neuroendocrine system governed by gonadotropin-releasing hormone (GnRH) neurons might be related to ovarian dysfunction in PCOS, although the link in this disordered brain-to-ovary communication remains unclear. Here, we manipulated GnRH neurons using chemogenetics in adult female mice to unveil whether chronic overaction of these neurons would trigger PCOS-like hormonal and reproductive impairments. METHODS We used adult Gnrh1cre female mice to selectively target and express the designer receptors exclusively activated by designer drugs (DREADD)-based chemogenetic tool hM3D(Gq) in hypophysiotropic GnRH neurons. Chronic chemogenetic activation protocol was carried out with clozapine N-oxide (CNO) i.p. injections every 48 h over a month. We evaluated the reproductive and hormonal profile before, during, and two months after chemogenetic manipulations. FINDINGS We discovered that the overactivation of GnRH neurons was sufficient to disrupt reproductive cycles, promote hyperandrogenism, and induce ovarian dysfunction. These PCOS features were detected with a long-lasting neuroendocrine dysfunction through abnormally high luteinizing hormone (LH) pulse secretion. Additionally, the GnRH-R blockade prevented the establishment of long-term neuroendocrine dysfunction and androgen excess in these animals. INTERPRETATION Taken together, our results show that hyperactivity of hypothalamic GnRH neurons is a major driver of reproductive and hormonal impairments in PCOS and suggest that antagonizing the aberrant GnRH signaling could be an efficient therapeutic venue for the treatment of PCOS. FUNDING European Research Council (ERC) under the European Union's Horizon 2020 research and innovation program (grant agreement n◦ 725149).
Collapse
Affiliation(s)
- Mauro S B Silva
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France; Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Laurine Decoster
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France; Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Gaspard Delpouve
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France; Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Tori Lhomme
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France; Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Gaetan Ternier
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France; Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France; Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Paolo Giacobini
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France; Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France.
| |
Collapse
|
22
|
Bayless DW, Davis CHO, Yang R, Wei Y, de Andrade Carvalho VM, Knoedler JR, Yang T, Livingston O, Lomvardas A, Martins GJ, Vicente AM, Ding JB, Luo L, Shah NM. A neural circuit for male sexual behavior and reward. Cell 2023; 186:3862-3881.e28. [PMID: 37572660 PMCID: PMC10615179 DOI: 10.1016/j.cell.2023.07.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/22/2023] [Accepted: 07/12/2023] [Indexed: 08/14/2023]
Abstract
Male sexual behavior is innate and rewarding. Despite its centrality to reproduction, a molecularly specified neural circuit governing innate male sexual behavior and reward remains to be characterized. We have discovered a developmentally wired neural circuit necessary and sufficient for male mating. This circuit connects chemosensory input to BNSTprTac1 neurons, which innervate POATacr1 neurons that project to centers regulating motor output and reward. Epistasis studies demonstrate that BNSTprTac1 neurons are upstream of POATacr1 neurons, and BNSTprTac1-released substance P following mate recognition potentiates activation of POATacr1 neurons through Tacr1 to initiate mating. Experimental activation of POATacr1 neurons triggers mating, even in sexually satiated males, and it is rewarding, eliciting dopamine release and self-stimulation of these cells. Together, we have uncovered a neural circuit that governs the key aspects of innate male sexual behavior: motor displays, drive, and reward.
Collapse
Affiliation(s)
- Daniel W Bayless
- Departments of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Chung-Ha O Davis
- Stanford Neurosciences Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Renzhi Yang
- Departments of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Yichao Wei
- Departments of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | | | - Joseph R Knoedler
- Departments of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Taehong Yang
- Departments of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Oscar Livingston
- Departments of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Akira Lomvardas
- Departments of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | | | - Ana Mafalda Vicente
- Allen Institute for Neural Dynamics, Seattle, WA 98109; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027
| | - Jun B Ding
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Departments of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Liqun Luo
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Department of Neurobiology, Stanford University, Stanford, CA 94305, USA
| | - Nirao M Shah
- Departments of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Department of Neurobiology, Stanford University, Stanford, CA 94305, USA; Department of Obstetrics and Gynecology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
23
|
Hoyos Sanchez MC, Bayat T, Gee RRF, Fon Tacer K. Hormonal Imbalances in Prader-Willi and Schaaf-Yang Syndromes Imply the Evolution of Specific Regulation of Hypothalamic Neuroendocrine Function in Mammals. Int J Mol Sci 2023; 24:13109. [PMID: 37685915 PMCID: PMC10487939 DOI: 10.3390/ijms241713109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
The hypothalamus regulates fundamental aspects of physiological homeostasis and behavior, including stress response, reproduction, growth, sleep, and feeding, several of which are affected in patients with Prader-Willi (PWS) and Schaaf-Yang syndrome (SYS). PWS is caused by paternal deletion, maternal uniparental disomy, or imprinting defects that lead to loss of expression of a maternally imprinted region of chromosome 15 encompassing non-coding RNAs and five protein-coding genes; SYS patients have a mutation in one of them, MAGEL2. Throughout life, PWS and SYS patients suffer from musculoskeletal deficiencies, intellectual disabilities, and hormonal abnormalities, which lead to compulsive behaviors like hyperphagia and temper outbursts. Management of PWS and SYS is mostly symptomatic and cures for these debilitating disorders do not exist, highlighting a clear, unmet medical need. Research over several decades into the molecular and cellular roles of PWS genes has uncovered that several impinge on the neuroendocrine system. In this review, we will discuss the expression and molecular functions of PWS genes, connecting them with hormonal imbalances in patients and animal models. Besides the observed hormonal imbalances, we will describe the recent findings about how the loss of individual genes, particularly MAGEL2, affects the molecular mechanisms of hormone secretion. These results suggest that MAGEL2 evolved as a mammalian-specific regulator of hypothalamic neuroendocrine function.
Collapse
Affiliation(s)
- Maria Camila Hoyos Sanchez
- School of Veterinary Medicine, Texas Tech University, 7671 Evans Dr., Amarillo, TX 79106, USA
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, TX 79106, USA
| | - Tara Bayat
- School of Veterinary Medicine, Texas Tech University, 7671 Evans Dr., Amarillo, TX 79106, USA
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, TX 79106, USA
| | - Rebecca R. Florke Gee
- School of Veterinary Medicine, Texas Tech University, 7671 Evans Dr., Amarillo, TX 79106, USA
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, TX 79106, USA
| | - Klementina Fon Tacer
- School of Veterinary Medicine, Texas Tech University, 7671 Evans Dr., Amarillo, TX 79106, USA
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, TX 79106, USA
| |
Collapse
|
24
|
Abstract
Our ability to understand the function of the nervous system is dependent upon defining the connections of its constituent neurons. Development of methods to define connections within neural networks has always been a growth industry in the neurosciences. Transneuronal spread of neurotropic viruses currently represents the best means of defining synaptic connections within neural networks. The method exploits the ability of viruses to invade neurons, replicate, and spread through the intimate synaptic connections that enable communication among neurons. Since the method was first introduced in the 1970s, it has benefited from an increased understanding of the virus life cycle, the function of viral genomes, and the ability to manipulate the viral genome in support of directional spread of virus and the expression of transgenes. In this article, we review these advances in viral tracing technology and the ways in which they may be applied for functional dissection of neural networks. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Retrograde infection of CNS circuits by peripheral injection of virus Basic Protocol 2: Transneuronal analysis by intracerebral injection Alternate Protocol 1: Transneuronal analysis with multiple recombinant strains Alternate Protocol 2: Conditional replication and spread of PRV Alternate Protocol 3: Conditional reporters of PRV infection and spread Alternate Protocol 4: Reporters of neural activity in polysynaptic circuits Support Protocol 1: Growing and titering a PRV viral stock Support Protocol 2: Immunohistochemical processing and detection Support Protocol 3: Dual-immunofluorescence localization.
Collapse
Affiliation(s)
- Esteban A Engel
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
- Current address: Spark Therapeutics, Philadelphia, PA, 19104
| | - J Patrick Card
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lynn W Enquist
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| |
Collapse
|
25
|
López M, Fernández-Real JM, Tomarev SI. Obesity wars: may the smell be with you. Am J Physiol Endocrinol Metab 2023; 324:E569-E576. [PMID: 37166265 PMCID: PMC10259866 DOI: 10.1152/ajpendo.00040.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/12/2023]
Abstract
Classically, the regulation of energy balance has been based on central and peripheral mechanisms sensing energy, nutrients, metabolites, and hormonal cues. Several cellular mechanisms at central level, such as hypothalamic AMP-activated protein kinase (AMPK), integrate this information to elicit counterregulatory responses that control feeding, energy expenditure, and glucose homeostasis, among other processes. Recent data have added more complexity to the homeostatic regulation of metabolism by introducing, for example, the key role of "traditional" senses and sensorial information in this complicated network. In this regard, current evidence is showing that olfaction plays a key and bidirectional role in energy homeostasis. Although nutritional status dynamically and profoundly impacts olfactory sensitivity, the sense of smell is involved in food appreciation and selection, as well as in brown adipose tissue (BAT) thermogenesis and substrate utilization, with some newly described actors, such as olfactomedin 2 (OLFM2), likely playing a major role. Thus, olfactory inputs are contributing to the regulation of both sides of the energy balance equation, namely, feeding and energy expenditure (EE), as well as whole body metabolism. Here, we will review the current knowledge and advances about the role of olfaction in the regulation of energy homeostasis.
Collapse
Affiliation(s)
- Miguel López
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain
| | - José Manuel Fernández-Real
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain
- Service of Diabetes, Endocrinology and Nutrition (UDEN), Institut d'Investigació Biomédica de Girona (IDIBGI), Department of Medical Sciences, University of Girona, Girona, Spain
| | - Stanislav I Tomarev
- Section of Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
26
|
Stopková R, Matějková T, Dodoková A, Talacko P, Zacek P, Sedlacek R, Piálek J, Stopka P. Variation in mouse chemical signals is genetically controlled and environmentally modulated. Sci Rep 2023; 13:8573. [PMID: 37237091 DOI: 10.1038/s41598-023-35450-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
In most mammals and particularly in mice, chemical communication relies on the detection of ethologically relevant fitness-related cues from other individuals. In mice, urine is the primary source of these signals, so we employed proteomics and metabolomics to identify key components of chemical signalling. We show that there is a correspondence between urinary volatiles and proteins in the representation of genetic background, sex and environment in two house mouse subspecies Mus musculus musculus and M. m. domesticus. We found that environment has a strong influence upon proteomic and metabolomic variation and that volatile mixtures better represent males while females have surprisingly more sex-biased proteins. Using machine learning and combined-omics techniques, we identified mixtures of metabolites and proteins that are associated with biological features.
Collapse
Affiliation(s)
- Romana Stopková
- Department of Zoology, Faculty of Science, BIOCEV, Charles University, Vestec, Prague, Czech Republic
| | - Tereza Matějková
- Department of Zoology, Faculty of Science, BIOCEV, Charles University, Vestec, Prague, Czech Republic
| | - Alica Dodoková
- Department of Zoology, Faculty of Science, BIOCEV, Charles University, Vestec, Prague, Czech Republic
| | - Pavel Talacko
- Department of Zoology, Faculty of Science, BIOCEV, Charles University, Vestec, Prague, Czech Republic
| | - Petr Zacek
- Department of Zoology, Faculty of Science, BIOCEV, Charles University, Vestec, Prague, Czech Republic
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Jaroslav Piálek
- Research Facility Studenec, Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic
| | - Pavel Stopka
- Department of Zoology, Faculty of Science, BIOCEV, Charles University, Vestec, Prague, Czech Republic.
| |
Collapse
|
27
|
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
|
28
|
Carta I, Autry AE. Hush little baby, don't you cry: How aversion to infant distress calls drives caregiving. Neuron 2023; 111:917-919. [PMID: 37023712 DOI: 10.1016/j.neuron.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 04/08/2023]
Abstract
Historically associated with aversion, the lateral habenula has a poorly characterized role in parenting. In this issue of Neuron, Lecca and colleagues1 show that these seemingly opposing roles converge in a subnucleus where aversion to pup cries may drive motivation for caregiving.
Collapse
Affiliation(s)
- Ilaria Carta
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anita E Autry
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
29
|
Yang T, Bayless DW, Wei Y, Landayan D, Marcelo IM, Wang Y, DeNardo LA, Luo L, Druckmann S, Shah NM. Hypothalamic neurons that mirror aggression. Cell 2023; 186:1195-1211.e19. [PMID: 36796363 PMCID: PMC10081867 DOI: 10.1016/j.cell.2023.01.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/13/2022] [Accepted: 01/17/2023] [Indexed: 02/17/2023]
Abstract
Social interactions require awareness and understanding of the behavior of others. Mirror neurons, cells representing an action by self and others, have been proposed to be integral to the cognitive substrates that enable such awareness and understanding. Mirror neurons of the primate neocortex represent skilled motor tasks, but it is unclear if they are critical for the actions they embody, enable social behaviors, or exist in non-cortical regions. We demonstrate that the activity of individual VMHvlPR neurons in the mouse hypothalamus represents aggression performed by self and others. We used a genetically encoded mirror-TRAP strategy to functionally interrogate these aggression-mirroring neurons. We find that their activity is essential for fighting and that forced activation of these cells triggers aggressive displays by mice, even toward their mirror image. Together, we have discovered a mirroring center in an evolutionarily ancient region that provides a subcortical cognitive substrate essential for a social behavior.
Collapse
Affiliation(s)
- Taehong Yang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Daniel W Bayless
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Yichao Wei
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Dan Landayan
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Ivo M Marcelo
- Champalimaud Neuroscience Program, Champalimaud Center for the Unknown, 1400-038 Lisbon, Portugal; Department of Psychiatry, Erasmus MC University Medical Center, 3015 GD Rotterdam, the Netherlands
| | - Yangpeng Wang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Laura A DeNardo
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Liqun Luo
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Shaul Druckmann
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Department of Neurobiology, Stanford University, Stanford, CA 94305, USA
| | - Nirao M Shah
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Department of Neurobiology, Stanford University, Stanford, CA 94305, USA; Department of Obstetrics and Gynecology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
30
|
Petzold A, van den Munkhof HE, Figge-Schlensok R, Korotkova T. Complementary lateral hypothalamic populations resist hunger pressure to balance nutritional and social needs. Cell Metab 2023; 35:456-471.e6. [PMID: 36827985 PMCID: PMC10028225 DOI: 10.1016/j.cmet.2023.02.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 05/03/2022] [Accepted: 02/08/2023] [Indexed: 02/25/2023]
Abstract
Animals continuously weigh hunger and thirst against competing needs, such as social contact and mating, according to state and opportunity. Yet neuronal mechanisms of sensing and ranking nutritional needs remain poorly understood. Here, combining calcium imaging in freely behaving mice, optogenetics, and chemogenetics, we show that two neuronal populations of the lateral hypothalamus (LH) guide increasingly hungry animals through behavioral choices between nutritional and social rewards. While increased food consumption was marked by increasing inhibition of a leptin receptor-expressing (LepRLH) subpopulation at a fast timescale, LepRLH neurons limited feeding or drinking and promoted social interaction despite hunger or thirst. Conversely, neurotensin-expressing LH neurons preferentially encoded water despite hunger pressure and promoted water seeking, while relegating social needs. Thus, hunger and thirst gate both LH populations in a complementary manner to enable the flexible fulfillment of multiple essential needs.
Collapse
Affiliation(s)
- Anne Petzold
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne and University Clinic Cologne, Cologne 50931, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Hanna Elin van den Munkhof
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne and University Clinic Cologne, Cologne 50931, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Rebecca Figge-Schlensok
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne and University Clinic Cologne, Cologne 50931, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Tatiana Korotkova
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne and University Clinic Cologne, Cologne 50931, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne 50931, Germany.
| |
Collapse
|
31
|
Abaffy T, Lu HY, Matsunami H. Sex steroid hormone synthesis, metabolism, and the effects on the mammalian olfactory system. Cell Tissue Res 2023; 391:19-42. [PMID: 36401093 PMCID: PMC9676892 DOI: 10.1007/s00441-022-03707-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 11/03/2022] [Indexed: 11/21/2022]
Abstract
Sex steroid hormones influence olfactory-mediated social behaviors, and it is generally hypothesized that these effects result from circulating hormones and/or neurosteroids synthesized in the brain. However, it is unclear whether sex steroid hormones are synthesized in the olfactory epithelium or the olfactory bulb, and if they can modulate the activity of the olfactory sensory neurons. Here, we review important discoveries related to the metabolism of sex steroids in the mouse olfactory epithelium and olfactory bulb, along with potential areas of future research. We summarize current knowledge regarding the expression, neuroanatomical distribution, and biological activity of the steroidogenic enzymes, sex steroid receptors, and proteins that are important to the metabolism of these hormones and reflect on their potential to influence early olfactory processing. We also review evidence related to the effects of sex steroid hormones on the development and activity of olfactory sensory neurons. By better understanding how these hormones are metabolized and how they act both at the periphery and olfactory bulb level, we can better appreciate the complexity of the olfactory system and discover potential similarities and differences in early olfactory processing between sexes.
Collapse
Affiliation(s)
- Tatjana Abaffy
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC 27710 USA
| | - Hsiu-Yi Lu
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC 27710 USA
| | - Hiroaki Matsunami
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC 27710 USA
| |
Collapse
|
32
|
Liu X, Porteous R, Herbison AE. Robust GABAergic Regulation of the GnRH Neuron Distal Dendron. Endocrinology 2022; 164:6862923. [PMID: 36458869 PMCID: PMC9749702 DOI: 10.1210/endocr/bqac194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 12/04/2022]
Abstract
The amino acid transmitter γ-aminobutyric acid (GABA) is suspected to play an important role in regulating the activity of the gonadotropin-releasing hormone (GnRH) neurons controlling fertility. Rodent GnRH neurons have a novel dendritic compartment termed the "distal dendron" through which action potentials pass to the axon terminals and where inputs from the kisspeptin pulse generator drive pulsatile GnRH secretion. Combining Gnrh1-Cre mice with the Cre-dependent calcium sensor GCaMP6 and confocal imaging of acute brain slices, we examined whether GABA regulated intracellular calcium concentrations ([Ca2+]) in the GnRH neuron distal dendron. Short puffs of GABA on the dendron evoked either a monophasic sustained suppression of [Ca2+] or a biphasic acute elevation in [Ca2+] followed by the sustained suppression. Application of muscimol to the dendron replicated the acute elevation in [Ca2+] while baclofen generated the sustained suppression. Robust GABAB receptor-mediated inhibition was observed in 80% to 100% of dendrons recorded from females across the estrous cycle and from approximately 70% of dendrons in males. In contrast, the GABAA receptor-mediated excitation was rare in males and varied across the estrous cycle, being most prominent at proestrus. The activation of GABAB receptors potently suppressed the stimulatory effect of kisspeptin on the dendron. These observations demonstrate that the great majority of GnRH neuron distal dendrons are regulated by GABAergic inputs in a sex- and estrous cycle-dependent manner, with robust GABAB receptor-mediated inhibition being the primary mode of signaling. This provides a new, kisspeptin-independent, pathway for the regulation of pulsatile and surge modes of GnRH secretion in the rodent.
Collapse
Affiliation(s)
- Xinhuai Liu
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin 9054, New Zealand
| | - Robert Porteous
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin 9054, New Zealand
| | - Allan E Herbison
- Correspondence: Allan E. Herbison, PhD, Department of Physiology, Development and Neuroscience, Downing Site, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
33
|
Ingram RJ, Leverton LK, Daniels VC, Li J, Christian-Hinman CA. Increased GABA transmission to GnRH neurons after intrahippocampal kainic acid injection in mice is sex-specific and associated with estrous cycle disruption. Neurobiol Dis 2022; 172:105822. [PMID: 35868435 PMCID: PMC9455811 DOI: 10.1016/j.nbd.2022.105822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/29/2022] [Accepted: 07/16/2022] [Indexed: 11/25/2022] Open
Abstract
Patients with epilepsy develop reproductive endocrine comorbidities at a rate higher than that of the general population. Clinical studies have identified disrupted luteinizing hormone (LH) release patterns in patients of both sexes, suggesting potential epilepsy-associated changes in hypothalamic gonadotropin-releasing hormone (GnRH) neuron function. In previous work, we found that GnRH neuron firing is increased in diestrous females and males in the intrahippocampal kainic acid (IHKA) mouse model of temporal lobe epilepsy. Notably, GABAA receptor activation is depolarizing in adult GnRH neurons. Therefore, here we tested the hypothesis that increased GnRH neuron firing in IHKA mice is associated with increased GABAergic drive to GnRH neurons. When ionotropic glutamate receptors (iGluRs) were blocked to isolate GABAergic postsynaptic currents (PSCs), no differences in PSC frequency were seen between GnRH neurons from control and IHKA diestrous females. In the absence of iGluR blockade, however, GABA PSC frequency was increased in GnRH neurons from IHKA females with disrupted estrous cycles, but not saline-injected controls nor IHKA females without estrous cycle disruption. GABA PSC amplitude was also increased in IHKA females with disrupted estrous cycles. These findings suggest the presence of an iGluR-dependent increase in feed-forward GABAergic transmission to GnRH neurons specific to IHKA females with comorbid cycle disruption. In males, GABA PSC frequency and amplitude were unchanged but PSC duration was reduced. Together, these findings suggest that increased GABA transmission helps drive elevated firing in IHKA females on diestrus and indicate the presence of a sex-specific hypothalamic mechanism underlying reproductive endocrine dysfunction in IHKA mice.
Collapse
Affiliation(s)
- Robbie J Ingram
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America
| | - Leanna K Leverton
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America
| | - Victoria C Daniels
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America
| | - Jiang Li
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America
| | - Catherine A Christian-Hinman
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America; Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America.
| |
Collapse
|
34
|
Manfredi-Lozano M, Leysen V, Adamo M, Paiva I, Rovera R, Pignat JM, Timzoura FE, Candlish M, Eddarkaoui S, Malone SA, Silva MSB, Trova S, Imbernon M, Decoster L, Cotellessa L, Tena-Sempere M, Claret M, Paoloni-Giacobino A, Plassard D, Paccou E, Vionnet N, Acierno J, Maceski AM, Lutti A, Pfrieger F, Rasika S, Santoni F, Boehm U, Ciofi P, Buée L, Haddjeri N, Boutillier AL, Kuhle J, Messina A, Draganski B, Giacobini P, Pitteloud N, Prevot V. GnRH replacement rescues cognition in Down syndrome. Science 2022; 377:eabq4515. [PMID: 36048943 PMCID: PMC7613827 DOI: 10.1126/science.abq4515] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
At the present time, no viable treatment exists for cognitive and olfactory deficits in Down syndrome (DS). We show in a DS model (Ts65Dn mice) that these progressive nonreproductive neurological symptoms closely parallel a postpubertal decrease in hypothalamic as well as extrahypothalamic expression of a master molecule that controls reproduction-gonadotropin-releasing hormone (GnRH)-and appear related to an imbalance in a microRNA-gene network known to regulate GnRH neuron maturation together with altered hippocampal synaptic transmission. Epigenetic, cellular, chemogenetic, and pharmacological interventions that restore physiological GnRH levels abolish olfactory and cognitive defects in Ts65Dn mice, whereas pulsatile GnRH therapy improves cognition and brain connectivity in adult DS patients. GnRH thus plays a crucial role in olfaction and cognition, and pulsatile GnRH therapy holds promise to improve cognitive deficits in DS.
Collapse
Affiliation(s)
- Maria Manfredi-Lozano
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Valerie Leysen
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Michela Adamo
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Isabel Paiva
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Université de Strasbourg-CNRS, Strasbourg, France
| | - Renaud Rovera
- Univ. Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron 69500, France
| | - Jean-Michel Pignat
- Department of Clinical Neurosciences, Neurorehabilitation Unit, University Hospital CHUV, Lausanne, Switzerland
| | - Fatima Ezzahra Timzoura
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Michael Candlish
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, 66421, Homburg, Germany
| | - Sabiha Eddarkaoui
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
| | - Samuel A. Malone
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Mauro S. B. Silva
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Sara Trova
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Monica Imbernon
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Laurine Decoster
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Ludovica Cotellessa
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Manuel Tena-Sempere
- Univ. Cordoba, IMIBC/HURS, CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain
| | - Ariane Paoloni-Giacobino
- Department of Genetic Medicine, University Hospitals of Geneva, 4 rue Gabrielle-Perret-Gentil, 1211, Genève 14, Switzerland
| | - Damien Plassard
- CNRS UMR 7104, INSERM U1258, GenomEast Platform, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Emmanuelle Paccou
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Nathalie Vionnet
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - James Acierno
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Aleksandra Maleska Maceski
- Neurologic Clinic and Polyclinic, MS Centre and Research Centre for Clinical Neuroimmunology and Neuroscience Basel; University Hospital Basel, University of Basel, Basel Switzerland
| | - Antoine Lutti
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Frank Pfrieger
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, 67000 Strasbourg, France
| | - S. Rasika
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Federico Santoni
- Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, 66421, Homburg, Germany
| | - Philippe Ciofi
- Univ. Bordeaux, Inserm, U1215, Neurocentre Magendie, Bordeaux, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
| | - Nasser Haddjeri
- Univ. Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron 69500, France
| | - Anne-Laurence Boutillier
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Université de Strasbourg-CNRS, Strasbourg, France
| | - Jens Kuhle
- Neurologic Clinic and Polyclinic, MS Centre and Research Centre for Clinical Neuroimmunology and Neuroscience Basel; University Hospital Basel, University of Basel, Basel Switzerland
| | - Andrea Messina
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Bogdan Draganski
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Switzerland
- Neurology Department, Max-Planck-Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Nelly Pitteloud
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| |
Collapse
|
35
|
Roa J, Ruiz-Cruz M, Ruiz-Pino F, Onieva R, Vazquez MJ, Sanchez-Tapia MJ, Ruiz-Rodriguez JM, Sobrino V, Barroso A, Heras V, Velasco I, Perdices-Lopez C, Ohlsson C, Avendaño MS, Prevot V, Poutanen M, Pinilla L, Gaytan F, Tena-Sempere M. Dicer ablation in Kiss1 neurons impairs puberty and fertility preferentially in female mice. Nat Commun 2022; 13:4663. [PMID: 35945211 PMCID: PMC9363423 DOI: 10.1038/s41467-022-32347-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/26/2022] [Indexed: 12/16/2022] Open
Abstract
Kiss1 neurons, producing kisspeptins, are essential for puberty and fertility, but their molecular regulatory mechanisms remain unfolded. Here, we report that congenital ablation of the microRNA-synthesizing enzyme, Dicer, in Kiss1 cells, causes late-onset hypogonadotropic hypogonadism in both sexes, but is compatible with pubertal initiation and preserved Kiss1 neuronal populations at the infantile/juvenile period. Yet, failure to complete puberty and attain fertility is observed only in females. Kiss1-specific ablation of Dicer evokes disparate changes of Kiss1-cell numbers and Kiss1/kisspeptin expression between hypothalamic subpopulations during the pubertal-transition, with a predominant decline in arcuate-nucleus Kiss1 levels, linked to enhanced expression of its repressors, Mkrn3, Cbx7 and Eap1. Our data unveil that miRNA-biosynthesis in Kiss1 neurons is essential for pubertal completion and fertility, especially in females, but dispensable for initial reproductive maturation and neuronal survival in both sexes. Our results disclose a predominant miRNA-mediated inhibitory program of repressive signals that is key for precise regulation of Kiss1 expression and, thereby, reproductive function.
Collapse
Affiliation(s)
- Juan Roa
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain. .,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain. .,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain. .,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain.
| | - Miguel Ruiz-Cruz
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
| | - Francisco Ruiz-Pino
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
| | - Rocio Onieva
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
| | - Maria J Vazquez
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
| | - Maria J Sanchez-Tapia
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
| | - Jose M Ruiz-Rodriguez
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
| | - Veronica Sobrino
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
| | - Alexia Barroso
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
| | - Violeta Heras
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
| | - Inmaculada Velasco
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
| | - Cecilia Perdices-Lopez
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Maria Soledad Avendaño
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, 59000, Lille, France
| | - Matti Poutanen
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530, Gothenburg, Sweden.,Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, and Turku Center for Disease Modeling, University of Turku, 20520, Turku, Finland
| | - Leonor Pinilla
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
| | - Francisco Gaytan
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba, 14004, Córdoba, Spain. .,Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain. .,Hospital Universitario Reina Sofia, 14004, Córdoba, Spain. .,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain. .,Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, and Turku Center for Disease Modeling, University of Turku, 20520, Turku, Finland.
| |
Collapse
|
36
|
Li J, Christian-Hinman CA. Epilepsy-associated increase in gonadotropin-releasing hormone neuron firing in diestrous female mice is independent of chronic seizure burden severity. Epilepsy Res 2022; 184:106948. [PMID: 35690025 PMCID: PMC10416707 DOI: 10.1016/j.eplepsyres.2022.106948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/04/2022] [Accepted: 05/16/2022] [Indexed: 12/21/2022]
Abstract
Reproductive endocrine disorders are common comorbidities of temporal lobe epilepsy (TLE). Our previous studies using the intrahippocampal kainic acid (IHKA) mouse model of TLE demonstrated that many females show prolonged estrous cycles and hypothalamic gonadotropin-releasing hormone (GnRH) neurons exhibit elevated firing during diestrus. However, it is unknown whether the degree of change in GnRH neuron activity is dependent on epilepsy severity. Here, we used 24/7 in vivo electroencephalography (EEG) and in vitro electrophysiological recordings in acute brain slices to assess GnRH neuron firing in relation to chronic seizure burden in diestrous female mice at two months after IHKA injection. We found that percentage of time in seizure activity in the 24 h prior to slice preparation is an accurate proxy of overall seizure burden. Firing rates of GnRH neurons from EEG-recorded IHKA mice were increased in comparison to controls, but no relationships were found between GnRH neuron firing and seizure burden measured in vivo. The independence of GnRH neuron firing rate in relation to seizure burden was unaffected by GnRH neuron soma location or estrous cycle length. Furthermore, GnRH neuron firing rates were not yet different from control values when measured 1 month after injection, when epileptogenesis is already complete in IHKA mice. These findings indicate that the severity of epilepsy and the degree of downstream disruption to GnRH neuron activity are independent, suggesting that susceptibility to reproductive endocrine comorbidities is driven by other risk factors.
Collapse
Affiliation(s)
- Jiang Li
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Catherine A Christian-Hinman
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
37
|
Guzmán-Ruiz MA, Jiménez A, Cárdenas-Rivera A, Guerrero-Vargas NN, Organista-Juárez D, Guevara-Guzmán R. Regulation of Metabolic Health by an "Olfactory-Hypothalamic Axis" and Its Possible Implications for the Development of Therapeutic Approaches for Obesity and T2D. Cell Mol Neurobiol 2022; 42:1727-1743. [PMID: 33813677 PMCID: PMC11421737 DOI: 10.1007/s10571-021-01080-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 03/12/2021] [Indexed: 12/12/2022]
Abstract
The olfactory system is responsible for the reception, integration and interpretation of odors. However, in the last years, it has been discovered that the olfactory perception of food can rapidly modulate the activity of hypothalamic neurons involved in the regulation of energy balance. Conversely, the hormonal signals derived from changes in the metabolic status of the body can also change the sensitivity of the olfactory system, suggesting that the bidirectional relationship established between the olfactory and the hypothalamic systems is key for the maintenance of metabolic homeostasis. In the first part of this review, we describe the possible mechanisms and anatomical pathways involved in the modulation of energy balance regulated by the olfactory system. Hence, we propose a model to explain its implication in the maintenance of the metabolic homeostasis of the organism. In the second part, we discuss how the olfactory system could be involved in the development of metabolic diseases such as obesity and type two diabetes and, finally, we propose the use of intranasal therapies aimed to regulate and improve the activity of the olfactory system that in turn will be able to control the neuronal activity of hypothalamic centers to prevent or ameliorate metabolic diseases.
Collapse
Affiliation(s)
- Mara Alaide Guzmán-Ruiz
- Laboratorio Sensorial, Departamento de Fisiología, Facultad de Medicina, Edificio A, 4º piso, Universidad Nacional Autónoma de México (UNAM), 04510, Ciudad de México, México.
| | - Adriana Jiménez
- Laboratorio Sensorial, Departamento de Fisiología, Facultad de Medicina, Edificio A, 4º piso, Universidad Nacional Autónoma de México (UNAM), 04510, Ciudad de México, México
| | - Alfredo Cárdenas-Rivera
- Centro de Investigación en Bioingeniería, Universidad de Ingeniería y Tecnología, Lima, Perú
| | - Natalí N Guerrero-Vargas
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, México
| | - Diana Organista-Juárez
- Laboratorio Sensorial, Departamento de Fisiología, Facultad de Medicina, Edificio A, 4º piso, Universidad Nacional Autónoma de México (UNAM), 04510, Ciudad de México, México
| | - Rosalinda Guevara-Guzmán
- Laboratorio Sensorial, Departamento de Fisiología, Facultad de Medicina, Edificio A, 4º piso, Universidad Nacional Autónoma de México (UNAM), 04510, Ciudad de México, México.
| |
Collapse
|
38
|
The role of ciliopathy-associated type 3 adenylyl cyclase in infanticidal behavior in virgin adult male mice. iScience 2022; 25:104534. [PMID: 35754726 PMCID: PMC9218507 DOI: 10.1016/j.isci.2022.104534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/11/2022] [Accepted: 06/01/2022] [Indexed: 12/04/2022] Open
Abstract
Virgin adult male mice often display killing of alien newborns, defined as infanticide, and this behavior is dependent on olfactory signaling. Olfactory perception is achieved by the main olfactory system (MOS) or vomeronasal system (VNS). Although it has been established that the VNS is crucial for infanticide in male mice, the role of the MOS in infanticide remains unknown. Herein, by producing lesions via ZnSO4 perfusion and N-methyl-D-aspartic acid stereotactic injection, we demonstrated that the main olfactory epithelium (MOE), anterior olfactory nucleus (AON), or ventromedial hypothalamus (VMH) is crucial for infanticide in adult males. By using CRISPR-Cas9 coupled with adeno-associated viruses to induce specific knockdown of type 3 adenylyl cyclase (AC3) in these tissues, we further demonstrated that AC3, a ciliopathy-associated protein, in the MOE and the expression of related proteins in the AON or VMH are necessary for infanticidal behavior in virgin adult male mice. MOE lesions and knockdown of AC3 in the MOE result in abnormal infanticidal behavior The infanticidal behavior of male mice is impaired by lesioning of the AON or VMH AC3 knockdown in the AON or VMH affects the infanticidal behavior of male mice
Collapse
|
39
|
Qin P, Ye J, Gong X, Yan X, Lin M, Lin T, Liu T, Li H, Wang X, Zhu Y, Li X, Liu Y, Li Y, Ling Y, Zhang X, Fang F. Quantitative proteomics analysis to assess protein expression levels in the ovaries of pubescent goats. BMC Genomics 2022; 23:507. [PMID: 35831802 PMCID: PMC9281040 DOI: 10.1186/s12864-022-08699-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/13/2022] [Indexed: 11/30/2022] Open
Abstract
Background Changes in the abundance of ovarian proteins play a key role in the regulation of reproduction. However, to date, no studies have investigated such changes in pubescent goats. Herein we applied isobaric tags for relative and absolute quantitation (iTRAQ) and liquid chromatography–tandem mass spectrometry to analyze the expression levels of ovarian proteins in pre-pubertal (n = 3) and pubertal (n = 3) goats. Results Overall, 7,550 proteins were recognized; 301 (176 up- and 125 downregulated) were identified as differentially abundant proteins (DAPs). Five DAPs were randomly selected for expression level validation by Western blotting; the results of Western blotting and iTRAQ analysis were consistent. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis indicated that DAPs were enriched in olfactory transduction, glutathione metabolism, and calcium signaling pathways. Besides, gene ontology functional enrichment analysis revealed that several DAPs enriched in biological processes were associated with cellular process, biological regulation, metabolic process, and response to stimulus. Protein–protein interaction network showed that proteins interacting with CDK1, HSPA1A, and UCK2 were the most abundant. Conclusions We identified 301 DAPs, which were enriched in olfactory transduction, glutathione metabolism, and calcium signaling pathways, suggesting the involvement of these processes in the onset of puberty. Further studies are warranted to more comprehensively explore the function of the identified DAPs and aforementioned signaling pathways to gain novel, deeper insights into the mechanisms underlying the onset of puberty. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08699-y.
Collapse
Affiliation(s)
- Ping Qin
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.,Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Jing Ye
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.,Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Xinbao Gong
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.,Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Xu Yan
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.,Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Maosen Lin
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.,Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Tao Lin
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.,Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Tong Liu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.,Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Hailing Li
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.,Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Xiujuan Wang
- Animal Husbandry Development Center, Huoqiu Animal Health Supervision Institute, Huoqiu County, Auditorium Road, Luan, 237400, Anhui, China
| | - Yanyun Zhu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Xiaoqian Li
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Ya Liu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.,Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Yunsheng Li
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.,Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Yinghui Ling
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Xiaorong Zhang
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.,Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Fugui Fang
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China. .,Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China.
| |
Collapse
|
40
|
Bai X, Fu L, Jin N, Liu X, Chen L, Shan Y, Zhang N, Wang P. Rescue of obesity-induced infertility in female mice by silencing AgRP neurons. Biochem Biophys Res Commun 2022; 623:32-38. [PMID: 35870259 DOI: 10.1016/j.bbrc.2022.07.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/08/2022] [Indexed: 11/18/2022]
Abstract
Obesity impacts multiple sites of the hypothalamus-pituitary gland-ovary axis (HPO axis) and has become a leading cause of female infertility. However, the critical hypothalamic neurons that participate in the development of obesity-induced infertility have not been well defined yet. Previous studies suggested that metabolic-sensing agouti-related peptide-expressing (AgRP) neurons in the arcuate nucleus (ARC) are hyperactive in diet-induced obesity (DIO) mice. We hypothesize that these neurons may convey metabolic dysfunction onto the HPO axis and contribute to obesity-induced infertility's pathophysiological process. To determine if AgRP neurons in obesity play a necessary role in the development of reproductive impairment in obesity, we used the chemogenetic method to normalize the neuronal activity of AgRP neurons in DIO female mice and test if their fertility can be restored. Our results indicated that chemogenetic inhibition of AgRP neurons could fully rescue the reproductive performance of DIO female mice, as manifested by recovered sex hormonal levels, ovulation, and fecundity. Moreover, we assayed serum AgRP levels in normal-weight and obese women and found elevated AgRP levels in obese subjects, suggesting the correlation between obesity and AgRP neuronal hyperactivity. Our results indicated that AgRP neurons constitute a central node connecting metabolism and reproduction, and dysfunctions of these neurons play a crucial role in reproductive impairment induced by metabolic abnormalities.
Collapse
Affiliation(s)
- Xueyan Bai
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, PR China
| | - Lei Fu
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, PR China
| | - Naiqian Jin
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, PR China
| | - Xiaoyan Liu
- Reproductive Medicine Center, Yantai Yuhuangding Hospital, Shandong, PR China
| | - Lili Chen
- Reproductive Medicine Center, Yantai Yuhuangding Hospital, Shandong, PR China
| | - Yinghua Shan
- Reproductive Medicine Center, Yantai Yuhuangding Hospital, Shandong, PR China
| | - Ning Zhang
- Reproductive Medicine Center, Yantai Yuhuangding Hospital, Shandong, PR China.
| | - Peng Wang
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, PR China.
| |
Collapse
|
41
|
Stincic TL, Kelly MJ. Estrogenic regulation of reproduction and energy homeostasis by a triumvirate of hypothalamic arcuate neurons. J Neuroendocrinol 2022; 34:e13145. [PMID: 35581942 DOI: 10.1111/jne.13145] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/31/2022] [Accepted: 04/15/2022] [Indexed: 11/29/2022]
Abstract
Pregnancy is energetically demanding and therefore, by necessity, reproduction and energy balance are inextricably linked. With insufficient or excessive energy stores a female is liable to suffer complications during pregnancy or produce unhealthy offspring. Gonadotropin-releasing hormone neurons are responsible for initiating both the pulsatile and subsequent surge release of luteinizing hormone to control ovulation. Meticulous work has identified two hypothalamic populations of kisspeptin (Kiss1) neurons that are critical for this pattern of release. The involvement of the hypothalamus is unsurprising because its quintessential function is to couple the endocrine and nervous systems, coordinating energy balance and reproduction. Estrogens, more specifically 17β-estradiol (E2 ), orchestrate the activity of a triumvirate of hypothalamic neurons within the arcuate nucleus (ARH) that govern the physiological underpinnings of these behavioral dynamics. Arising from a common progenitor pool, these cells differentiate into ARH kisspeptin, pro-opiomelanocortin (POMC), and agouti related peptide/neuropeptide Y (AgRP) neurons. Although the excitability of all these subpopulations is subject to genomic and rapid estrogenic regulation, Kiss1 neurons are the most sensitive, reflecting their integral function in female fertility. Based on the premise that E2 coordinates autonomic functions around reproduction, we review recent findings on how Kiss1 neurons interact with gonadotropin-releasing hormone, AgRP and POMC neurons, as well as how the rapid membrane-initiated and intracellular signaling cascades activated by E2 in these neurons are critical for control of homeostatic functions supporting reproduction. In particular, we highlight how Kiss1 and POMC neurons conspire to inhibit AgRP neurons and diminish food motivation in service of reproductive success.
Collapse
Affiliation(s)
- Todd L Stincic
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
| | - Martin J Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| |
Collapse
|
42
|
Campbell RE, Coolen LM, Hoffman GE, Hrabovszky E. Highlights of neuroanatomical discoveries of the mammalian gonadotropin-releasing hormone system. J Neuroendocrinol 2022; 34:e13115. [PMID: 35502534 PMCID: PMC9232911 DOI: 10.1111/jne.13115] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/09/2022] [Accepted: 03/01/2022] [Indexed: 11/17/2022]
Abstract
The anatomy and morphology of gonadotropin-releasing hormone (GnRH) neurons makes them both a joy and a challenge to investigate. They are a highly unique population of neurons given their developmental migration into the brain from the olfactory placode, their relatively small number, their largely scattered distribution within the rostral forebrain, and, in some species, their highly varied individual anatomical characteristics. These unique features have posed technological hurdles to overcome and promoted fertile ground for the establishment and use of creative approaches. Historical and more contemporary discoveries defining GnRH neuron anatomy remain critical in shaping and challenging our views of GnRH neuron function in the regulation of reproductive function. We begin this review with a historical overview of anatomical discoveries and developing methodologies that have shaped our understanding of the reproductive axis. We then highlight significant discoveries across specific groups of mammalian species to address some of the important comparative aspects of GnRH neuroanatomy. Lastly, we touch on unresolved questions and opportunities for future neuroanatomical research on this fascinating and important population of neurons.
Collapse
Affiliation(s)
- Rebecca E. Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| | - Lique M. Coolen
- Department of Biological SciencesKent State UniversityKentOhioUSA
| | | | - Erik Hrabovszky
- Laboratory of Reproductive NeurobiologyInstitute of Experimental MedicineBudapestHungary
| |
Collapse
|
43
|
Hypothalamic kisspeptin and kisspeptin receptors: Species variation in reproduction and reproductive behaviours. Front Neuroendocrinol 2022; 64:100951. [PMID: 34757093 DOI: 10.1016/j.yfrne.2021.100951] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/22/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023]
Abstract
Kisspeptin, encoded by the KISS1 gene, was first discovered as a potential metastasis suppressor gene. The prepro-kisspeptin precursor is cleaved into shorter mature bioactive peptides of varying sizes that bind to the G protein-coupled receptor GPR54 (=KISS1R). Over the last two decades, multiple types of Kiss and KissR genes have been discovered in mammalian and non-mammalian vertebrate species, but they are remarkably absent in birds. Kiss neuronal populations are distributed mainly in the hypothalamus. The KissRs are widely distributed in the brain, including the hypothalamic and non-hypothalamic regions, such as the hippocampus, amygdala, and habenula. The role of KISS1-KISS1R in humans and Kiss1-Kiss1R in rodents is associated with puberty, gonadal maturation, and the reproductive axis. However, recent gene deletion studies in zebrafish and medaka have provided controversial results, suggesting that the reproductive role of kiss is dispensable. This review highlights the evolutionary history, localisation, and significance of Kiss-KissR in reproduction and reproductive behaviours in mammalian and non-mammalian vertebrates.
Collapse
|
44
|
GnRH neurons recruit astrocytes in infancy to facilitate network integration and sexual maturation. Nat Neurosci 2021; 24:1660-1672. [PMID: 34795451 DOI: 10.1038/s41593-021-00960-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 10/08/2021] [Indexed: 12/25/2022]
Abstract
Neurons that produce gonadotropin-releasing hormone (GnRH), which control fertility, complete their nose-to-brain migration by birth. However, their function depends on integration within a complex neuroglial network during postnatal development. Here, we show that rodent GnRH neurons use a prostaglandin D2 receptor DP1 signaling mechanism during infancy to recruit newborn astrocytes that 'escort' them into adulthood, and that the impairment of postnatal hypothalamic gliogenesis markedly alters sexual maturation by preventing this recruitment, a process mimicked by the endocrine disruptor bisphenol A. Inhibition of DP1 signaling in the infantile preoptic region, where GnRH cell bodies reside, disrupts the correct wiring and firing of GnRH neurons, alters minipuberty or the first activation of the hypothalamic-pituitary-gonadal axis during infancy, and delays the timely acquisition of reproductive capacity. These findings uncover a previously unknown neuron-to-neural-progenitor communication pathway and demonstrate that postnatal astrogenesis is a basic component of a complex set of mechanisms used by the neuroendocrine brain to control sexual maturation.
Collapse
|
45
|
Mishor E, Amir D, Weiss T, Honigstein D, Weissbrod A, Livne E, Gorodisky L, Karagach S, Ravia A, Snitz K, Karawani D, Zirler R, Weissgross R, Soroka T, Endevelt-Shapira Y, Agron S, Rozenkrantz L, Reshef N, Furman-Haran E, Breer H, Strotmann J, Uebi T, Ozaki M, Sobel N. Sniffing the human body volatile hexadecanal blocks aggression in men but triggers aggression in women. SCIENCE ADVANCES 2021; 7:eabg1530. [PMID: 34797713 PMCID: PMC8604408 DOI: 10.1126/sciadv.abg1530] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 09/30/2021] [Indexed: 05/29/2023]
Abstract
In terrestrial mammals, body volatiles can effectively trigger or block conspecific aggression. Here, we tested whether hexadecanal (HEX), a human body volatile implicated as a mammalian-wide social chemosignal, affects human aggression. Using validated behavioral paradigms, we observed a marked dissociation: Sniffing HEX blocked aggression in men but triggered aggression in women. Next, using functional brain imaging, we uncovered a pattern of brain activity mirroring behavior: In both men and women, HEX increased activity in the left angular gyrus, an area implicated in perception of social cues. HEX then modulated functional connectivity between the angular gyrus and a brain network implicated in social appraisal (temporal pole) and aggressive execution (amygdala and orbitofrontal cortex) in a sex-dependent manner consistent with behavior: increasing connectivity in men but decreasing connectivity in women. These findings implicate sex-specific social chemosignaling at the mechanistic heart of human aggressive behavior.
Collapse
Affiliation(s)
- Eva Mishor
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Daniel Amir
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Weiss
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Danielle Honigstein
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Aharon Weissbrod
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Ethan Livne
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Lior Gorodisky
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Shiri Karagach
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Aharon Ravia
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Kobi Snitz
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Diyala Karawani
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Rotem Zirler
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Reut Weissgross
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Timna Soroka
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Yaara Endevelt-Shapira
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Shani Agron
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Liron Rozenkrantz
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Netta Reshef
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Edna Furman-Haran
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
| | - Heinz Breer
- Institute of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Joerg Strotmann
- Institute of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Tatsuya Uebi
- Department of Biology, Graduate School of Science, Kobe University, Kobe, Japan
| | - Mamiko Ozaki
- Department of Biology, Graduate School of Science, Kobe University, Kobe, Japan
| | - Noam Sobel
- Azrieli National Center for Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
46
|
Pardasani M, Marathe SD, Purnapatre MM, Dalvi U, Abraham NM. Multimodal learning of pheromone locations. FASEB J 2021; 35:e21836. [PMID: 34407246 PMCID: PMC7611819 DOI: 10.1096/fj.202100167r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/28/2021] [Accepted: 07/20/2021] [Indexed: 11/11/2022]
Abstract
Memorizing pheromonal locations is critical for many mammalian species as it involves finding mates and avoiding competitors. In rodents, pheromonal information is perceived by the main and accessory olfactory systems. However, the role of somatosensation in context-dependent learning and memorizing of pheromone locations remains unexplored. We addressed this problem by training female mice on a multimodal task to locate pheromones by sampling volatiles emanating from male urine through the orifices of varying dimensions or shapes that are sensed by their vibrissae. In this novel pheromone location assay, female mice’ preference toward male urine scent decayed over time when they were permitted to explore pheromones vs neutral stimuli, water. On training them for the associations involving olfactory and whisker systems, it was established that they were able to memorize the location of opposite sex pheromones, when tested 15 days later. This memory was not formed either when the somatosensory inputs through whisker pad were blocked or when the pheromonal cues were replaced with that of same sex. The association between olfactory and somatosensory systems was further confirmed by the enhanced expression of the activity-regulated cytoskeleton protein. Furthermore, the activation of main olfactory bulb circuitry by pheromone volatiles did not cause any modulation in learning and memorizing non-pheromonal volatiles. Our study thus provides the evidence for associations formed between different sensory modalities facilitating the long-term memory formation relevant to social and reproductive behaviors.
Collapse
Affiliation(s)
- Meenakshi Pardasani
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, India
| | - Shruti D Marathe
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, India
| | - Maitreyee Mandar Purnapatre
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, India.,Institute of Bioinformatics & Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Urvashi Dalvi
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, India.,Institute of Bioinformatics & Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Nixon M Abraham
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, India
| |
Collapse
|
47
|
Paternal Finasteride Treatment Can Influence the Testicular Transcriptome Profile of Male Offspring-Preliminary Study. Curr Issues Mol Biol 2021; 43:868-886. [PMID: 34449557 PMCID: PMC8929076 DOI: 10.3390/cimb43020062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Hormone-dependent events that occur throughout spermatogenesis during postnatal testis maturation are significant for adult male fertility. Any disturbances in the T/DHT ratio in male progeny born from females fertilized by finasteride-treated male rats (F0:Fin) can result in the impairment of testicular physiology. The goal of this work was to profile the testicular transcriptome in the male filial generation (F1:Fin) from paternal F0:Fin rats. (2) Methods: The subject material for the study were testis from immature and mature male rats born from females fertilized by finasteride-treated rats. Testicular tissues from the offspring were used in microarray analyses. (3) Results: The top 10 genes having the highest and lowest fold change values were mainly those that encoded odoriferous (Olfr: 31, 331, 365, 633, 774, 814, 890, 935, 1109, 1112, 1173, 1251, 1259, 1253, 1383) and vomeronasal (Vmn1r: 50, 103, 210, 211; Vmn2r: 3, 23, 99) receptors and RIKEN cDNA 5430402E10, also known as odorant-binding protein. (4) Conclusions: Finasteride treatment of male adult rats may cause changes in the testicular transcriptome of their male offspring, leading to a defective function of spermatozoa in response to odorant-like signals, which are recently more and more often noticed as significant players in male fertility.
Collapse
|
48
|
Riddell P, Paris MCJ, Joonè CJ, Pageat P, Paris DBBP. Appeasing Pheromones for the Management of Stress and Aggression during Conservation of Wild Canids: Could the Solution Be Right under Our Nose? Animals (Basel) 2021; 11:ani11061574. [PMID: 34072227 PMCID: PMC8230031 DOI: 10.3390/ani11061574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Many canid species are declining globally. It is important to conserve these species that often serve as important predators within ecosystems. Continued human expansion and the resulting habitat fragmentation necessitate conservation interventions, such as translocation, artificial pack formation, and captive breeding programs. However, chronic stress often occurs during these actions, and can result in aggression, and the physiological suppression of immunity and reproduction. Limited options are currently available for stress and aggression management in wild canids. Pheromones provide a promising natural alternative for stress management; an appeasing pheromone has been identified for multiple domestic species and may reduce stress and aggression behaviours. Many pheromones are species-specific, and the appeasing pheromone has been found to have slight compositional changes across species. In this review, the benefits of a dog appeasing pheromone and the need to investigate species-specific derivatives to produce more pronounced and beneficial behavioural and physiological modulation in target species as a conservation tool are examined. Abstract Thirty-six species of canid exist globally, two are classified as critically endangered, three as endangered, and five as near threatened. Human expansion and the coinciding habitat fragmentation necessitate conservation interventions to mitigate concurrent population deterioration. The current conservation management of wild canids includes animal translocation and artificial pack formation. These actions often cause chronic stress, leading to increased aggression and the suppression of the immune and reproductive systems. Castration and pharmaceutical treatments are currently used to reduce stress and aggression in domestic and captive canids. The undesirable side effects make such treatments inadvisable during conservation management of wild canids. Pheromones are naturally occurring chemical messages that modulate behaviour between conspecifics; as such, they offer a natural alternative for behaviour modification. Animals are able to distinguish between pheromones of closely related species through small compositional differences but are more likely to have greater responses to pheromones from individuals of the same species. Appeasing pheromones have been found to reduce stress- and aggression-related behaviours in domestic species, including dogs. Preliminary evidence suggests that dog appeasing pheromones (DAP) may be effective in wild canids. However, the identification and testing of species-specific derivatives could produce more pronounced and beneficial behavioural and physiological changes in target species. In turn, this could provide a valuable tool to improve the conservation management of many endangered wild canids.
Collapse
Affiliation(s)
- Pia Riddell
- Gamete and Embryology (GAME) Laboratory, College of Public Health, Medical and Veterinary Sciences, James Cook University, James Cook Drive, Townsville, QLD 4811, Australia;
- Institute for Breeding Rare and Endangered African Mammals (IBREAM), 9 Ainslie Place, Edinburgh EH3 6AT SCT, UK;
- Centre for Tropical Environmental and Sustainability Science, James Cook University, James Cook Drive, Townsville, QLD 4811, Australia
| | - Monique C. J. Paris
- Institute for Breeding Rare and Endangered African Mammals (IBREAM), 9 Ainslie Place, Edinburgh EH3 6AT SCT, UK;
- Mammal Research Institute, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria 0028, South Africa
| | - Carolynne J. Joonè
- Discipline of Veterinary Science, College of Public Health, Medical and Veterinary Sciences, James Cook University, Solander Drive, Townsville, QLD 4811, Australia;
| | - Patrick Pageat
- Institut de Recherche en Sémiochemie et Ethologie Appliquée, 84400 Apt, France;
| | - Damien B. B. P. Paris
- Gamete and Embryology (GAME) Laboratory, College of Public Health, Medical and Veterinary Sciences, James Cook University, James Cook Drive, Townsville, QLD 4811, Australia;
- Institute for Breeding Rare and Endangered African Mammals (IBREAM), 9 Ainslie Place, Edinburgh EH3 6AT SCT, UK;
- Centre for Tropical Environmental and Sustainability Science, James Cook University, James Cook Drive, Townsville, QLD 4811, Australia
- Correspondence: ; Tel.: +61-7-4781-6006
| |
Collapse
|
49
|
Tahir MS, Porto-Neto LR, Gondro C, Shittu OB, Wockner K, Tan AWL, Smith HR, Gouveia GC, Kour J, Fortes MRS. Meta-Analysis of Heifer Traits Identified Reproductive Pathways in Bos indicus Cattle. Genes (Basel) 2021; 12:768. [PMID: 34069992 PMCID: PMC8157873 DOI: 10.3390/genes12050768] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Fertility traits measured early in life define the reproductive potential of heifers. Knowledge of genetics and biology can help devise genomic selection methods to improve heifer fertility. In this study, we used ~2400 Brahman cattle to perform GWAS and multi-trait meta-analysis to determine genomic regions associated with heifer fertility. Heifer traits measured were pregnancy at first mating opportunity (PREG1, a binary trait), first conception score (FCS, score 1 to 3) and rebreeding score (REB, score 1 to 3.5). The heritability estimates were 0.17 (0.03) for PREG1, 0.11 (0.05) for FCS and 0.28 (0.05) for REB. The three traits were highly genetically correlated (0.75-0.83) as expected. Meta-analysis was performed using SNP effects estimated for each of the three traits, adjusted for standard error. We identified 1359 significant SNPs (p-value < 9.9 × 10-6 at FDR < 0.0001) in the multi-trait meta-analysis. Genomic regions of 0.5 Mb around each significant SNP from the meta-analysis were annotated to create a list of 2560 positional candidate genes. The most significant SNP was in the vicinity of a genomic region on chromosome 8, encompassing the genes SLC44A1, FSD1L, FKTN, TAL2 and TMEM38B. The genomic region in humans that contains homologs of these genes is associated with age at puberty in girls. Top significant SNPs pointed to additional fertility-related genes, again within a 0.5 Mb region, including ESR2, ITPR1, GNG2, RGS9BP, ANKRD27, TDRD12, GRM1, MTHFD1, PTGDR and NTNG1. Functional pathway enrichment analysis resulted in many positional candidate genes relating to known fertility pathways, including GnRH signaling, estrogen signaling, progesterone mediated oocyte maturation, cAMP signaling, calcium signaling, glutamatergic signaling, focal adhesion, PI3K-AKT signaling and ovarian steroidogenesis pathway. The comparison of results from this study with previous transcriptomics and proteomics studies on puberty of the same cattle breed (Brahman) but in a different population identified 392 genes in common from which some genes-BRAF, GABRA2, GABR1B, GAD1, FSHR, CNGA3, PDE10A, SNAP25, ESR2, GRIA2, ORAI1, EGFR, CHRNA5, VDAC2, ACVR2B, ORAI3, CYP11A1, GRIN2A, ATP2B3, CAMK2A, PLA2G, CAMK2D and MAPK3-are also part of the above-mentioned pathways. The biological functions of the positional candidate genes and their annotation to known pathways allowed integrating the results into a bigger picture of molecular mechanisms related to puberty in the hypothalamus-pituitary-ovarian axis. A reasonable number of genes, common between previous puberty studies and this study on early reproductive traits, corroborates the proposed molecular mechanisms. This study identified the polymorphism associated with early reproductive traits, and candidate genes that provided a visualization of the proposed mechanisms, coordinating the hypothalamic, pituitary, and ovarian functions for reproductive performance in Brahman cattle.
Collapse
Affiliation(s)
- Muhammad S. Tahir
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Laercio R. Porto-Neto
- Commonwealth Scientific and Industrial Research Organization, Brisbane, QLD 4072, Australia;
| | - Cedric Gondro
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA;
| | - Olasege B. Shittu
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Kimberley Wockner
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Andre W. L. Tan
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Hugo R. Smith
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Gabriela C. Gouveia
- Animal Science Department, Veterinary School, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Jagish Kour
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Marina R. S. Fortes
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| |
Collapse
|
50
|
Qiu Q, Wu Y, Ma L, Xu W, Hills M, Ramalingam V, Yu CR. Acquisition of innate odor preference depends on spontaneous and experiential activities during critical period. eLife 2021; 10:e60546. [PMID: 33769278 PMCID: PMC8032394 DOI: 10.7554/elife.60546] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 03/24/2021] [Indexed: 01/15/2023] Open
Abstract
Animals possess an inborn ability to recognize certain odors to avoid predators, seek food, and find mates. Innate odor preference is thought to be genetically hardwired. Here we report that acquisition of innate odor recognition requires spontaneous neural activity and is influenced by sensory experience during early postnatal development. Genetic silencing of mouse olfactory sensory neurons during the critical period has little impact on odor sensitivity, discrimination, and recognition later in life. However, it abolishes innate odor preference and alters the patterns of activation in brain centers. Exposure to innately recognized odors during the critical period abolishes the associated valence in adulthood in an odor-specific manner. The changes are associated with broadened projection of olfactory sensory neurons and expression of axon guidance molecules. Thus, a delicate balance of neural activity is needed during the critical period in establishing innate odor preference and convergent axon input is required to encode innate odor valence.
Collapse
Affiliation(s)
- Qiang Qiu
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Yunming Wu
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Limei Ma
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Wenjing Xu
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Max Hills
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Vivekanandan Ramalingam
- Stowers Institute for Medical ResearchKansas CityUnited States
- Interdisciplinary Graduate Program in Biomedical Sciences, University of Kansas Medical CenterKansas CityUnited States
| | - C Ron Yu
- Stowers Institute for Medical ResearchKansas CityUnited States
- Interdisciplinary Graduate Program in Biomedical Sciences, University of Kansas Medical CenterKansas CityUnited States
- Department of Anatomy and Cell Biology, University of Kansas Medical CenterKansas CityUnited States
| |
Collapse
|