51
|
Mussa BM, Srivastava A, Verberne AJM. COVID-19 and Neurological Impairment: Hypothalamic Circuits and Beyond. Viruses 2021; 13:v13030498. [PMID: 33802995 PMCID: PMC8002703 DOI: 10.3390/v13030498] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/15/2021] [Accepted: 02/26/2021] [Indexed: 12/23/2022] Open
Abstract
In December 2019, a novel coronavirus known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in Wuhan, the capital of Hubei, China. The virus infection, coronavirus disease 2019 (COVID-19), represents a global concern, as almost all countries around the world are affected. Clinical reports have confirmed several neurological manifestations in COVID-19 patients such as headaches, vomiting, and nausea, indicating the involvement of the central nervous system (CNS) and peripheral nervous system (PNS). Neuroinvasion of coronaviruses is not a new phenomenon, as it has been demonstrated by previous autopsies of severe acute respiratory syndrome coronavirus (SARS-CoV) patients who experienced similar neurologic symptoms. The hypothalamus is a complex structure that is composed of many nuclei and diverse neuronal cell groups. It is characterized by intricate intrahypothalamic circuits that orchestrate a finely tuned communication within the CNS and with the PNS. Hypothalamic circuits are critical for maintaining homeostatic challenges including immune responses to viral infections. The present article reviews the possible routes and mechanisms of neuroinvasion of SARS-CoV-2, with a specific focus on the role of the hypothalamic circuits in mediating the neurological symptoms noted during COVID-19 infection.
Collapse
Affiliation(s)
- Bashair M. Mussa
- Basic Medical Science Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Correspondence: ; Tel.: +971-65057220
| | - Ankita Srivastava
- Sharjah Institute for Medical Research and College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates;
| | - Anthony J. M. Verberne
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg 3084, Australia;
| |
Collapse
|
52
|
Qiu Q, Wu Y, Ma L, Yu CR. Encoding innately recognized odors via a generalized population code. Curr Biol 2021; 31:1813-1825.e4. [PMID: 33651991 PMCID: PMC8119320 DOI: 10.1016/j.cub.2021.01.094] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 12/25/2020] [Accepted: 01/27/2021] [Indexed: 01/19/2023]
Abstract
Odors carrying intrinsic values often trigger instinctive aversive or attractive responses. It is not known how innate valence is encoded. An intuitive model suggests that the information is conveyed through specific channels in hardwired circuits along the olfactory pathway, insulated from influences of other odors, to trigger innate responses. Here, we show that in mice, mixing innately aversive or attractive odors with a neutral odor and, surprisingly, mixing two odors with the same valence, abolish the innate behavioral responses. Recordings from the olfactory bulb indicate that odors are not masked at the level of peripheral activation and glomeruli independently encode components in the mixture. In contrast, crosstalk among the mitral and tufted (M/T) cells changes their patterns of activity such that those elicited by the mixtures can no longer be linearly decoded as separate components. The changes in behavioral and M/T cell responses are associated with reduced activation of brain areas linked to odor preferences. Thus, crosstalk among odor channels at the earliest processing stage in the olfactory pathway leads to re-coding of odor identity to abolish valence associated with the odors. These results are inconsistent with insulated labeled lines and support a model of a common mechanism of odor recognition for both innate and learned valence associations.
Collapse
Affiliation(s)
- Qiang Qiu
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Yunming Wu
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Limei Ma
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - C Ron Yu
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA.
| |
Collapse
|
53
|
Yip SH, Liu X, Hessler S, Cheong I, Porteous R, Herbison AE. Indirect Suppression of Pulsatile LH Secretion by CRH Neurons in the Female Mouse. Endocrinology 2021; 162:bqaa237. [PMID: 33543235 DOI: 10.1210/endocr/bqaa237] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Indexed: 01/01/2023]
Abstract
Acute stress is a potent suppressor of pulsatile luteinizing hormone (LH) secretion, but the mechanisms through which corticotrophin-releasing hormone (CRH) neurons inhibit gonadotropin-releasing hormone (GnRH) release remain unclear. The activation of paraventricular nucleus (PVN) CRH neurons with Cre-dependent hM3Dq in Crh-Cre female mice resulted in the robust suppression of pulsatile LH secretion. Channelrhodopsin (ChR2)-assisted circuit mapping revealed that PVN CRH neuron projections existed around kisspeptin neurons in the arcuate nucleus (ARN) although many more fibers made close appositions with GnRH neuron distal dendrons in the ventral ARN. Acutely prepared brain slice electrophysiology experiments in GnRH- green fluorescent protein (GFP) mice showed a dose-dependent (30 and 300 nM CRH) activation of firing in ~20% of GnRH neurons in both intact diestrus and ovariectomized mice with inhibitory effects being uncommon (<8%). Confocal GCaMP6 imaging of GnRH neuron distal dendrons in acute para-horizontal brain slices from GnRH-Cre mice injected with Cre-dependent GCaMP6s adeno-associated viruses demonstrated no effects of 30 to 300 nM CRH on GnRH neuron dendron calcium concentrations. Electrophysiological recordings of ARN kisspeptin neurons in Crh-Cre,Kiss1-GFP mice revealed no effects of 30 -300 nM CRH on basal or neurokinin B-stimulated firing rate. Similarly, the optogenetic activation (2-20 Hz) of CRH nerve terminals in the ARN of Crh-Cre,Kiss1-GFP mice injected with Cre-dependent ChR2 had no effect on kisspeptin neuron firing. Together, these studies demonstrate that PVN CRH neurons potently suppress LH pulsatility but do not exert direct inhibitory control over GnRH neurons, at their cell body or dendron, or the ARN kisspeptin neuron pulse generator in the female mouse.
Collapse
Affiliation(s)
- Siew Hoong Yip
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Xinhuai Liu
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Sabine Hessler
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Isaiah Cheong
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Robert Porteous
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| |
Collapse
|
54
|
Dela Cruz C, Horton CA, Sanders KN, Andersen ND, Tsai PS. Conditional Fgfr1 Deletion in GnRH Neurons Leads to Minor Disruptions in the Reproductive Axis of Male and Female Mice. Front Endocrinol (Lausanne) 2021; 11:588459. [PMID: 33679600 PMCID: PMC7933197 DOI: 10.3389/fendo.2020.588459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/21/2020] [Indexed: 11/13/2022] Open
Abstract
In humans and mice, inactivating mutations in fibroblast growth factor receptor 1 (Fgfr1) lead to gonadotropin-releasing hormone (GnRH) deficiency and a host of downstream reproductive disorders. It was unclear if Fgfr1 signaling directly upon GnRH neurons critically drove the establishment of a functional GnRH system. To answer this question, we generated a mouse model with a conditional deletion of Fgfr1 in GnRH neurons using the Cre/loxP approach. These mice, called Fgfr1cKO mice, were examined along with control mice for their pubertal onset and a host of reproductive axis functions. Our results showed that Fgfr1cKO mice harbored no detectable defects in the GnRH system and pubertal onset, suffered only subtle changes in the pituitary function, but exhibited significantly disrupted testicular and ovarian morphology at 25 days of age, indicating impaired gametogenesis at a young age. However, these disruptions were transient and became undetectable in older mice. Our results suggest that Fgfr1 signaling directly on GnRH neurons supports, to some extent, the reproductive axis function in the period leading to the early phase of puberty, but is not critically required for pubertal onset or reproductive maintenance in sexually mature animals.
Collapse
Affiliation(s)
| | | | | | | | - Pei-San Tsai
- Department of Integrative Physiology, University of Colorado, Boulder, CO, United States
| |
Collapse
|
55
|
The decisive role of subordination in social hierarchy in weanling mice and young children. iScience 2021; 24:102073. [PMID: 33604524 PMCID: PMC7873650 DOI: 10.1016/j.isci.2021.102073] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/10/2020] [Accepted: 01/12/2021] [Indexed: 11/22/2022] Open
Abstract
Social hierarchy plays important roles in maintaining social structures. Despite similarity in concept, frameworks of human hierarchy have seldom been investigated in parallel with other animals. Moreover, the importance of subordination in hierarchical formation has been largely underestimated in previous research. Here we established, compared, and investigated hierarchy in children and weanling mice. Temperament assessments suggested that children who are less persistent, low emotional intensity, and withdrew easily were more likely to be subordinate in competitive scenarios independent of task characteristics and interaction experiences. The tube test further showed that conflicts between mice were not resolved by winner approach but by loser withdrawal, which was mainly determined by intrinsic subordinate status regardless of opponents. Our study presents evolutionary conserved hierarchical relationships in young and a critical role of the intrinsic subordinate characteristics in hierarchical determination. These findings provide a new perspective on social interactions with potential implications for preschool education. Similar social hierarchy can be established in weanling mice and young children Social rankings in children are largely influenced by intrinsic characteristics Conflicts in mouse tube test are not resolved by winner but by loser withdrawal Subordinate withdrawals are determined by intrinsic status regardless of opponents
Collapse
|
56
|
Liu X, Yeo SH, McQuillan HJ, Herde MK, Hessler S, Cheong I, Porteous R, Herbison AE. Highly redundant neuropeptide volume co-transmission underlying episodic activation of the GnRH neuron dendron. eLife 2021; 10:62455. [PMID: 33464205 PMCID: PMC7847305 DOI: 10.7554/elife.62455] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/15/2021] [Indexed: 12/16/2022] Open
Abstract
The necessity and functional significance of neurotransmitter co-transmission remains unclear. The glutamatergic 'KNDy' neurons co-express kisspeptin, neurokinin B (NKB), and dynorphin and exhibit a highly stereotyped synchronized behavior that reads out to the gonadotropin-releasing hormone (GnRH) neuron dendrons to drive episodic hormone secretion. Using expansion microscopy, we show that KNDy neurons make abundant close, non-synaptic appositions with the GnRH neuron dendron. Electrophysiology and confocal GCaMP6 imaging demonstrated that, despite all three neuropeptides being released from KNDy terminals, only kisspeptin was able to activate the GnRH neuron dendron. Mice with a selective deletion of kisspeptin from KNDy neurons failed to exhibit pulsatile hormone secretion but maintained synchronized episodic KNDy neuron behavior that is thought to depend on recurrent NKB and dynorphin transmission. This indicates that KNDy neurons drive episodic hormone secretion through highly redundant neuropeptide co-transmission orchestrated by differential post-synaptic neuropeptide receptor expression at the GnRH neuron dendron and KNDy neuron.
Collapse
Affiliation(s)
- Xinhuai Liu
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Shel-Hwa Yeo
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand.,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - H James McQuillan
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Michel K Herde
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Sabine Hessler
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Isaiah Cheong
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Robert Porteous
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand.,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
57
|
Tirindelli R. Coding of pheromones by vomeronasal receptors. Cell Tissue Res 2021; 383:367-386. [PMID: 33433690 DOI: 10.1007/s00441-020-03376-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/02/2020] [Indexed: 01/11/2023]
Abstract
Communication between individuals is critical for species survival, reproduction, and expansion. Most terrestrial species, with the exception of humans who predominantly use vision and phonation to create their social network, rely on the detection and decoding of olfactory signals, which are widely known as pheromones. These chemosensory cues originate from bodily fluids, causing attractive or avoidance behaviors in subjects of the same species. Intraspecific pheromone signaling is then crucial to identify sex, social ranking, individuality, and health status, thus establishing hierarchies and finalizing the most efficient reproductive strategies. Indeed, all these features require fine tuning of the olfactory systems to detect molecules containing this information. To cope with this complexity of signals, tetrapods have developed dedicated olfactory subsystems that refer to distinct peripheral sensory detectors, called the main olfactory and the vomeronasal organ, and two minor structures, namely the septal organ of Masera and the Grueneberg ganglion. Among these, the vomeronasal organ plays the most remarkable role in pheromone coding by mediating several behavioral outcomes that are critical for species conservation and amplification. In rodents, this organ is organized into two segregated neuronal subsets that express different receptor families. To some extent, this dichotomic organization is preserved in higher projection areas of the central nervous system, suggesting, at first glance, distinct functions for these two neuronal pathways. Here, I will specifically focus on this issue and discuss the role of vomeronasal receptors in mediating important innate behavioral effects through the recognition of pheromones and other biological chemosignals.
Collapse
Affiliation(s)
- Roberto Tirindelli
- Department of Medicine and Surgery, University of Parma, Via Volturno, 39, 43125, Parma, Italy.
| |
Collapse
|
58
|
Rosenfield RL, Cooke DW, Radovick S. Puberty in the Female and Its Disorders. SPERLING PEDIATRIC ENDOCRINOLOGY 2021:528-626. [DOI: 10.1016/b978-0-323-62520-3.00016-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
59
|
Chen T, Chen C, Wu H, Chen X, Xie R, Wang F, Sun H, Chen L. Overexpression of p53 accelerates puberty in high-fat diet-fed mice through Lin28/let-7 system. Exp Biol Med (Maywood) 2021; 246:66-71. [PMID: 32996351 PMCID: PMC7797992 DOI: 10.1177/1535370220961320] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 09/02/2020] [Indexed: 01/22/2023] Open
Abstract
IMPACT STATEMENT High-fat intake and subsequent obesity are associated with premature onset of puberty, but the exact neuroendocrine mechanisms are still unclear. The transcriptional factor p53 has been predicted to be a central hub of the gene networks controlling the pubertal onset. Besides, p53 also plays crucial roles in metabolism. Here, we explored p53 in the hypothalami of mice fed a high-fat diet (HFD), which showed an up-regulated expression. Besides, we also revealed that overexpressed p53 may accelerate hypothalamo-pituitary-gonadal (HPG) axis activation partially through the c-Myc/Lin28/let-7 system. These results can deepen our understanding of the interaction between metabolic regulation and puberty onset control, and may shed light on the neuroendocrine mechanisms of obesity-related central precocious puberty.
Collapse
Affiliation(s)
- Ting Chen
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Jiangsu 215000, China
| | - Cailong Chen
- Office of Human Resource, Children's Hospital of Soochow University, Jiangsu 215000, China
| | - Haiying Wu
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Jiangsu 215000, China
| | - Xiuli Chen
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Jiangsu 215000, China
| | - Rongrong Xie
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Jiangsu 215000, China
| | - Fengyun Wang
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Jiangsu 215000, China
| | - Hui Sun
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Jiangsu 215000, China
| | - Linqi Chen
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Jiangsu 215000, China
| |
Collapse
|
60
|
Ye Y, Lu Z, Zhou W. Pheromone effects on the human hypothalamus in relation to sexual orientation and gender. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:293-306. [PMID: 34266600 DOI: 10.1016/b978-0-12-819973-2.00021-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pheromones are chemicals that serve communicational purposes within a species. In most terrestrial mammals, pheromones are detected by either the olfactory epithelium or the vomeronasal organ and processed by various downstream structures including the medial amygdala and the hypothalamus to regulate motivated behaviors and endocrine responses. The search for human pheromones began in the 1970s. Whereas bioactive ligands are yet to be identified, there has been accumulating evidence that human body odors exert a range of pheromone-like effects on the recipients, including triggering innate behavioral responses, modulating endocrine levels, signaling social information, and affecting mood and cognition. In parallel, results from recent brain imaging studies suggest that body odors evoke distinct neural responses from those observed with common nonsocial odors. Two endogenous steroids androsta-4,16,- dien-3-one and estra-1,3,5(10),16-tetraen-3-ol are considered by some as candidates for human sex pheromones. The two substances produce sexually dimorphic effects on human perception, mood, and physiological arousal. Moreover, they reportedly elicit different hypothalamic response patterns in manners contingent on the recipients' sex and sexual orientation. Neuroendocrine mechanisms underlying the effects of human chemosignals are not yet clear and await future detailed analyses.
Collapse
Affiliation(s)
- Yuting Ye
- State Key Laboratory of Brain and Cognitive Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Zhonghua Lu
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wen Zhou
- State Key Laboratory of Brain and Cognitive Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
61
|
Yip SH, Campos P, Liu X, Porteous R, Herbison AE. Innervation of GnRH Neuron Distal Projections and Activation by Kisspeptin in a New GnRH-Cre Rat Model. Endocrinology 2021; 162:bqaa186. [PMID: 33057587 DOI: 10.1210/endocr/bqaa186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Indexed: 12/12/2022]
Abstract
The neural mechanisms generating pulsatile GnRH release from the median eminence (ME) remain unclear. Studies undertaken in the mouse demonstrate that GnRH neurons extend projections to the ME that have properties of both dendrites and axons, termed "dendrons," and that the kisspeptin neuron pulse generator targets these distal dendrons to drive pulsatile GnRH secretion. It presently remains unknown whether the GnRH neuron dendron exists in other species. We report here the generation of a knock-in Gnrh1-Ires-Cre rat line with near-perfect targeting of Cre recombinase to the GnRH neuronal phenotype. More than 90% of adult male and female GnRH neurons express Cre with no ectopic expression. Adeno-associated viruses were used in adult female Gnrh1-Ires-Cre rats to target mCherry or GCAMP6 to rostral preoptic area GnRH neurons. The mCherry tracer revealed the known unipolar and bipolar morphology of GnRH neurons and their principal projection pathways to the external zone of the ME. Synaptophysin-labeling of presynaptic nerve terminals revealed that GnRH neuron distal projections received numerous close appositions as they passed through the arcuate nucleus and into the median eminence. Confocal GCaMP6 imaging in acute horizontal brain slices demonstrated that GnRH neuron distal projections lateral to the median eminence were activated by kisspeptin. These studies indicate the presence of a dendron-like arrangement in the rat with GnRH neuron distal projections receiving synaptic input and responding to kisspeptin.
Collapse
Affiliation(s)
- Siew Hoong Yip
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Pauline Campos
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Xinhuai Liu
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Robert Porteous
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| |
Collapse
|
62
|
Social Transmission and Buffering of Hippocampal Metaplasticity after Stress in Mice. J Neurosci 2020; 41:1317-1330. [PMID: 33310752 DOI: 10.1523/jneurosci.1751-20.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/16/2020] [Accepted: 11/30/2020] [Indexed: 11/21/2022] Open
Abstract
In social animals, the behavioral and hormonal responses to stress can be transmitted from one individual to another through a social transmission process, and, conversely, social support ameliorates stress responses, a phenomenon referred to as social buffering. Metaplasticity represents activity-dependent synaptic changes that modulate the ability to elicit subsequent synaptic plasticity. Authentic stress can induce hippocampal metaplasticity, but whether transmitted stress has the same ability remains unknown. Here, using an acute restraint-tailshock stress paradigm, we report that both authentic and transmitted stress in adult male mice trigger metaplastic facilitation of long-term depression (LTD) induction at hippocampal CA1 synapses. Using LTD as a readout of persistent synaptic consequences of stress, our findings demonstrate that, in a male-male dyad, stress transmission happens in nearly half of naive partners and stress buffering occurs in approximately half of male stressed mice that closely interact with naive partners. By using a social-confrontation tube test to assess the dominant-subordinate relationship in a male-male dyad, we found that stressed subordinate mice are not buffered by naive dominant partners and that stress transmission is exhibited in ∼60% of dominant naive partners. Furthermore, the appearance of stress transmission correlates with more time spent in sniffing the anogenital area of stressed mice, and the appearance of stress buffering correlates with more time engaged in allogrooming from naive partners. Chemical ablation of the olfactory epithelium with dichlobenil or physical separation between social contacts diminishes stress transmission. Together, our data demonstrate that transmitted stress can elicit metaplastic facilitation of LTD induction as authentic stress.SIGNIFICANCE STATEMENT Social animals can acquire information about their environment through interactions with conspecifics. Stress can induce enduring changes in neural activity and synaptic function. Current studies are already unraveling the transmission and buffering of stress responses between individuals, but little is known about the relevant synaptic changes associated with social transmission and buffering of stress. Here, we show that authentic and transmitted stress can prime glutamatergic synapses onto hippocampal CA1 neurons to undergo long-term depression. This hippocampal metaplasticity is bufferable following social interactions with naive partners. Hierarchical status of naive partners strongly affects the social buffering effect on synaptic consequences of stress. This work provides novel insights into the conceptual framework for synaptic changes with social transmission and buffering of stress.
Collapse
|
63
|
Trova S, Bovetti S, Pellegrino G, Bonzano S, Giacobini P, Peretto P. HPG-Dependent Peri-Pubertal Regulation of Adult Neurogenesis in Mice. Front Neuroanat 2020; 14:584493. [PMID: 33328903 PMCID: PMC7732626 DOI: 10.3389/fnana.2020.584493] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/29/2020] [Indexed: 11/13/2022] Open
Abstract
Adult neurogenesis, a striking form of neural plasticity, is involved in the modulation of social stimuli driving reproduction. Previous studies on adult neurogenesis have shown that this process is significantly modulated around puberty in female mice. Puberty is a critical developmental period triggered by increased secretion of the gonadotropin releasing hormone (GnRH), which controls the activity of the hypothalamic-pituitary-gonadal axis (HPG). Secretion of HPG-axis factors at puberty participates to the refinement of neural circuits that govern reproduction. Here, by exploiting a transgenic GnRH deficient mouse model, that progressively loses GnRH expression during postnatal development (GnRH::Cre;Dicer loxP/loxP mice), we found that a postnatally-acquired dysfunction in the GnRH system affects adult neurogenesis selectively in the subventricular-zone neurogenic niche in a sexually dimorphic way. Moreover, by examining adult females ovariectomized before the onset of puberty, we provide important evidence that, among the HPG-axis secreting factors, the circulating levels of gonadal hormones during pre-/peri-pubertal life contribute to set-up the proper adult subventricular zone-olfactory bulb neurogenic system.
Collapse
Affiliation(s)
- Sara Trova
- Department of Life Sciences and Systems Biology, Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy.,Univ.Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, Laboratory of the Development and Plasticity of Neuroendocrine Brain, Lille, France
| | - Serena Bovetti
- Department of Life Sciences and Systems Biology, Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy
| | - Giuliana Pellegrino
- Univ.Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, Laboratory of the Development and Plasticity of Neuroendocrine Brain, Lille, France
| | - Sara Bonzano
- Department of Life Sciences and Systems Biology, Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy
| | - Paolo Giacobini
- Univ.Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, Laboratory of the Development and Plasticity of Neuroendocrine Brain, Lille, France
| | - Paolo Peretto
- Department of Life Sciences and Systems Biology, Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy
| |
Collapse
|
64
|
Taroc EZM, Katreddi RR, Forni PE. Identifying Isl1 Genetic Lineage in the Developing Olfactory System and in GnRH-1 Neurons. Front Physiol 2020; 11:601923. [PMID: 33192618 PMCID: PMC7609815 DOI: 10.3389/fphys.2020.601923] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/30/2020] [Indexed: 01/04/2023] Open
Abstract
During embryonic development, symmetric ectodermal thickenings [olfactory placodes (OP)] give rise to several cell types that comprise the olfactory system, such as those that form the terminal nerve ganglion (TN), gonadotropin releasing hormone-1 neurons (GnRH-1ns), and other migratory neurons in rodents. Even though the genetic heterogeneity among these cell types is documented, unidentified cell populations arising from the OP remain. One candidate to identify placodal derived neurons in the developing nasal area is the transcription factor Isl1, which was recently identified in GnRH-3 neurons of the terminal nerve in fish, as well as expression in neurons of the nasal migratory mass (MM). Here, we analyzed the Isl1 genetic lineage in chemosensory neuronal populations in the nasal area and migratory GnRH-1ns in mice using in situ hybridization, immunolabeling a Tamoxifen inducible Isl1CreERT and a constitutive Isl1Cre knock-in mouse lines. In addition, we also performed conditional Isl1 ablation in developing GnRH neurons. We found Isl1 lineage across non-sensory cells of the respiratory epithelium and sustentacular cells of OE and VNO. We identified a population of transient embryonic Isl1 + neurons in the olfactory epithelium and sparse Isl1 + neurons in postnatal VNO. Isl1 is expressed in almost all GnRH neurons and in approximately half of the other neuron populations in the MM. However, Isl1 conditional ablation alone does not significantly compromise GnRH-1 neuronal migration or GnRH-1 expression, suggesting compensatory mechanisms. Further studies will elucidate the functional and mechanistic role of Isl1 in development of migratory endocrine neurons.
Collapse
Affiliation(s)
- Ed Zandro M Taroc
- Department of Biological Sciences, The RNA Institute, and the Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY, United States
| | - Raghu Ram Katreddi
- Department of Biological Sciences, The RNA Institute, and the Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY, United States
| | - Paolo E Forni
- Department of Biological Sciences, The RNA Institute, and the Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY, United States
| |
Collapse
|
65
|
Abstract
Gonadal hormones contribute to the sexual differentiation of brain and behavior throughout the lifespan, from initial neural patterning to "activation" of adult circuits. Sexual behavior is an ideal system in which to investigate the mechanisms underlying hormonal activation of neural circuits. Sexual behavior is a hormonally regulated, innate social behavior found across species. Although both sexes seek out and engage in sexual behavior, the specific actions involved in mating are sexually dimorphic. Thus, the neural circuits mediating sexual motivation and behavior in males and females are overlapping yet distinct. Furthermore, sexual behavior is strongly dependent on circulating gonadal hormones in both sexes. There has been significant recent progress on elucidating how gonadal hormones modulate physiological properties within sexual behavior circuits with consequences for behavior. Therefore, in this mini-review we review the neural circuits of male and female sexual motivation and behavior, from initial sensory detection of pheromones to the extended amygdala and on to medial hypothalamic nuclei and reward systems. We also discuss how gonadal hormones impact the physiology and functioning of each node within these circuits. By better understanding the myriad of ways in which gonadal hormones impact sexual behavior circuits, we can gain a richer and more complete appreciation for the neural substrates of complex behavior.
Collapse
Affiliation(s)
- Kimberly J Jennings
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| |
Collapse
|
66
|
Vanacker C, Trova S, Shruti S, Casoni F, Messina A, Croizier S, Malone S, Ternier G, Hanchate NK, Rasika S, Bouret SG, Ciofi P, Giacobini P, Prevot V. Neuropilin-1 expression in GnRH neurons regulates prepubertal weight gain and sexual attraction. EMBO J 2020; 39:e104633. [PMID: 32761635 PMCID: PMC7527814 DOI: 10.15252/embj.2020104633] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/01/2020] [Accepted: 07/13/2020] [Indexed: 12/26/2022] Open
Abstract
Hypothalamic neurons expressing gonadotropin-releasing hormone (GnRH), the "master molecule" regulating reproduction and fertility, migrate from their birthplace in the nose to their destination using a system of guidance cues, which include the semaphorins and their receptors, the neuropilins and plexins, among others. Here, we show that selectively deleting neuropilin-1 in new GnRH neurons enhances their survival and migration, resulting in excess neurons in the hypothalamus and in their unusual accumulation in the accessory olfactory bulb, as well as an acceleration of mature patterns of activity. In female mice, these alterations result in early prepubertal weight gain, premature attraction to male odors, and precocious puberty. Our findings suggest that rather than being influenced by peripheral energy state, GnRH neurons themselves, through neuropilin-semaphorin signaling, might engineer the timing of puberty by regulating peripheral adiposity and behavioral switches, thus acting as a bridge between the reproductive and metabolic axes.
Collapse
Affiliation(s)
- Charlotte Vanacker
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Sara Trova
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Sonal Shruti
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Filippo Casoni
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Andrea Messina
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Sophie Croizier
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | - Samuel Malone
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Gaetan Ternier
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Naresh Kumar Hanchate
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - S Rasika
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Sebastien G Bouret
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Philippe Ciofi
- Inserm U1215Neurocentre MagendieBordeauxFrance
- Université de BordeauxBordeauxFrance
| | - Paolo Giacobini
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| |
Collapse
|
67
|
Rozenkrantz L, Weissgross R, Weiss T, Ravreby I, Frumin I, Shushan S, Gorodisky L, Reshef N, Holzman Y, Pinchover L, Endevelt-Shapira Y, Mishor E, Soroka T, Finkel M, Tagania L, Ravia A, Perl O, Furman-Haran E, Carp H, Sobel N. Unexplained repeated pregnancy loss is associated with altered perceptual and brain responses to men's body-odor. eLife 2020; 9:e55305. [PMID: 32988456 PMCID: PMC7524551 DOI: 10.7554/elife.55305] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 08/18/2020] [Indexed: 01/24/2023] Open
Abstract
Mammalian olfaction and reproduction are tightly linked, a link less explored in humans. Here, we asked whether human unexplained repeated pregnancy loss (uRPL) is associated with altered olfaction, and particularly altered olfactory responses to body-odor. We found that whereas most women with uRPL could identify the body-odor of their spouse, most control women could not. Moreover, women with uRPL rated the perceptual attributes of men's body-odor differently from controls. These pronounced differences were accompanied by an only modest albeit significant advantage in ordinary, non-body-odor-related olfaction in uRPL. Next, using structural and functional brain imaging, we found that in comparison to controls, most women with uRPL had smaller olfactory bulbs, yet increased hypothalamic response in association with men's body-odor. These findings combine to suggest altered olfactory perceptual and brain responses in women experiencing uRPL, particularly in relation to men's body-odor. Whether this link has any causal aspects to it remains to be explored.
Collapse
Affiliation(s)
- Liron Rozenkrantz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Reut Weissgross
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Tali Weiss
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Inbal Ravreby
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Idan Frumin
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Sagit Shushan
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
- Department of Otolaryngology & Head and Neck Surgery, Edith Wolfson Medical Center, Holon, Israel
| | - Lior Gorodisky
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Netta Reshef
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Yael Holzman
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Liron Pinchover
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Yaara Endevelt-Shapira
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Eva Mishor
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Timna Soroka
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Maya Finkel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Liav Tagania
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Aharon Ravia
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Ofer Perl
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| | - Edna Furman-Haran
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Howard Carp
- Department of Obstetrics & Gynecology, Sheba Medical Center, Tel Hashomer, Israel
| | - Noam Sobel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Rehovot, Israel
| |
Collapse
|
68
|
An Y, Guan X, Ni Y, Zhao Y, Chen Z, Chen Y, Zhang J. Reversible olfactory dysfunction impaired learning and memory with impaired hippocampal synaptic plasticity and increased corticosterone release in mice. Neurochem Int 2020; 138:104774. [DOI: 10.1016/j.neuint.2020.104774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/18/2020] [Accepted: 05/22/2020] [Indexed: 10/24/2022]
|
69
|
Wang L, Guo W, Shen X, Yeo S, Long H, Wang Z, Lyu Q, Herbison AE, Kuang Y. Different dendritic domains of the GnRH neuron underlie the pulse and surge modes of GnRH secretion in female mice. eLife 2020; 9:53945. [PMID: 32644040 PMCID: PMC7347383 DOI: 10.7554/elife.53945] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 06/18/2020] [Indexed: 12/26/2022] Open
Abstract
The gonadotropin-releasing hormone (GnRH) neurons exhibit pulse and surge modes of activity to control fertility. They also exhibit an unusual bipolar morphology comprised of a classical soma-proximal dendritic zone and an elongated secretory process that can operate as both a dendrite and an axon, termed a 'dendron'. We show using expansion microscopy that the highest density of synaptic inputs to a GnRH neuron exists at its distal dendron. In vivo, selective chemogenetic inhibition of the GnRH neuron distal dendron abolishes the luteinizing hormone (LH) surge and markedly dampens LH pulses. In contrast, inhibitory chemogenetic and optogenetic strategies targeting the GnRH neuron soma-proximal dendritic zone abolish the LH surge but have no effect upon LH pulsatility. These observations indicate that electrical activity at the soma-proximal dendrites of the GnRH neuron is only essential for the LH surge while the distal dendron represents an autonomous zone where synaptic integration drives pulsatile GnRH secretion.
Collapse
Affiliation(s)
- Li Wang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Wenya Guo
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xi Shen
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Shel Yeo
- Centre for Neuroendocrinology, Department of Physiology, University of Otago, Dunedin, New Zealand.,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Hui Long
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Zhexuan Wang
- School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qifeng Lyu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Allan E Herbison
- Centre for Neuroendocrinology, Department of Physiology, University of Otago, Dunedin, New Zealand.,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
70
|
Chemogenetic Suppression of GnRH Neurons during Pubertal Development Can Alter Adult GnRH Neuron Firing Rate and Reproductive Parameters in Female Mice. eNeuro 2020; 7:ENEURO.0223-20.2020. [PMID: 32513661 PMCID: PMC7363480 DOI: 10.1523/eneuro.0223-20.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/02/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) neurons control anterior pituitary, and thereby gonadal, function. GnRH neurons are active before outward indicators of puberty appear. Prenatal androgen (PNA) exposure mimics reproductive dysfunction of the common fertility disorder polycystic ovary syndrome (PCOS) and reduces prepubertal GnRH neuron activity. Early neuron activity can play a critical role in establishing circuitry and adult function. We tested the hypothesis that changing prepubertal GnRH neuron activity programs adult GnRH neuron activity and reproduction independent of androgen exposure in female mice. Activating (3Dq) or inhibitory (4Di) designer receptors exclusively activated by designer drugs (DREADDs) were targeted to GnRH neurons using Cre-lox technology. In control studies, the DREADD ligand clozapine n-oxide (CNO) produced the expected changes in GnRH neuron activity in vitro and luteinizing hormone (LH) release in vivo. CNO was administered to control or PNA mice between two and three weeks of age, when GnRH neuron firing rate is reduced in PNA mice. In controls, reducing prepubertal GnRH neuron activity with 4Di increased adult GnRH neuron firing rate and days in diestrus but did not change puberty onset or GABA transmission to these cells. In contrast, activating GnRH neurons had no effect on reproductive parameters or firing rate and did not rescue reproductive phenotypes in PNA mice. These studies support the hypothesis that prepubertal neuronal activity sculpts elements of the adult reproductive neuroendocrine axis and cyclicity but indicate that other PNA-induced programming actions are required for full reproductive phenotypes and/or that compensatory mechanisms overcome activity-mediated changes to mitigate reproductive changes in adults.
Collapse
|
71
|
Yang L, Demetriou L, Wall MB, Mills EG, Zargaran D, Sykes M, Prague JK, Abbara A, Owen BM, Bassett PA, Rabiner EA, Comninos AN, Dhillo WS. Kisspeptin enhances brain responses to olfactory and visual cues of attraction in men. JCI Insight 2020; 5:133633. [PMID: 32051344 PMCID: PMC7098781 DOI: 10.1172/jci.insight.133633] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/18/2019] [Indexed: 01/07/2023] Open
Abstract
Successful reproduction is a fundamental physiological process that relies on the integration of sensory cues of attraction with appropriate emotions and behaviors and the reproductive axis. However, the factors responsible for this integration remain largely unexplored. Using functional neuroimaging, hormonal, and psychometric analyses, we demonstrate that the reproductive hormone kisspeptin enhances brain activity in response to olfactory and visual cues of attraction in men. Furthermore, the brain regions enhanced by kisspeptin correspond to areas within the olfactory and limbic systems that govern sexual behavior and perception of beauty as well as overlap with its endogenous expression pattern. Of key functional and behavioral significance, we observed that kisspeptin was most effective in men with lower sexual quality-of-life scores. As such, our results reveal a previously undescribed attraction pathway in humans activated by kisspeptin and identify kisspeptin signaling as a new therapeutic target for related reproductive and psychosexual disorders. Kisspeptin enhances brain processing in response to olfactory and visual cues of attraction and is most effective in men with lower sexual quality of life.
Collapse
Affiliation(s)
- Lisa Yang
- Section of Endocrinology & Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, United Kingdom
| | - Lysia Demetriou
- Section of Endocrinology & Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, United Kingdom.,Invicro, Hammersmith Hospital, London, United Kingdom
| | - Matthew B Wall
- Invicro, Hammersmith Hospital, London, United Kingdom.,Division of Brain Sciences, Faculty of Medicine, Imperial College London, United Kingdom.,Clinical Psychopharmacology Unit, University College London, United Kingdom
| | - Edouard Ga Mills
- Section of Endocrinology & Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, United Kingdom
| | - David Zargaran
- Section of Endocrinology & Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, United Kingdom
| | - Mark Sykes
- Section of Endocrinology & Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, United Kingdom
| | - Julia K Prague
- Section of Endocrinology & Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, United Kingdom
| | - Ali Abbara
- Section of Endocrinology & Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, United Kingdom
| | - Bryn M Owen
- Section of Endocrinology & Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, United Kingdom
| | | | - Eugenii A Rabiner
- Invicro, Hammersmith Hospital, London, United Kingdom.,Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College, London, United Kingdom
| | - Alexander N Comninos
- Section of Endocrinology & Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, United Kingdom.,Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Waljit S Dhillo
- Section of Endocrinology & Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, United Kingdom
| |
Collapse
|
72
|
Han SY, Cheong I, McLennan T, Herbison AE. Neural Determinants of Pulsatile Luteinizing Hormone Secretion in Male Mice. Endocrinology 2020; 161:5697320. [PMID: 31907531 DOI: 10.1210/endocr/bqz045] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 01/03/2020] [Indexed: 11/19/2022]
Abstract
The gonadotrophin-releasing hormone (GnRH) pulse generator drives pulsatile luteinizing hormone (LH) secretion essential for fertility. However, the constraints within which the pulse generator operates to drive efficient LH pulsatility remain unclear. We used optogenetic activation of the arcuate nucleus kisspeptin neurons, recently identified as the GnRH pulse generator, to assess the efficiency of different pulse generator frequencies in driving pulsatile LH secretion in intact freely behaving male mice. Activating the pulse generator at 45-minute intervals generated LH pulses similar to those observed in intact male mice while 9-minute interval stimulation generated LH profiles indistinguishable from gonadectomized (GDX) male mice. However, more frequent activation of the pulse generator resulted in disordered LH secretion. Optogenetic experiments directly activating the distal projections of the GnRH neuron gave the exact same results, indicating the pituitary to be the locus of the high frequency decoding. To evaluate the state-dependent behavior of the pulse generator, the effects of high-frequency activation of the arcuate kisspeptin neurons were compared in GDX and intact mice. The same stimulus resulted in an overall inhibition of LH release in GDX mice but stimulation in intact males. These studies demonstrate that the GnRH pulse generator is the primary determinant of LH pulse profile and that a nonlinear relationship exists between pulse generator frequency and LH pulse frequency. This may underlie the ability of stimulatory inputs to the pulse generator to have opposite effects on LH secretion in intact and GDX animals.
Collapse
Affiliation(s)
- Su Young Han
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Isaiah Cheong
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Tim McLennan
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| |
Collapse
|
73
|
Camargo AP, Nakahara TS, Firmino LER, Netto PHM, do Nascimento JBP, Donnard ER, Galante PAF, Carazzolle MF, Malnic B, Papes F. Uncovering the mouse olfactory long non-coding transcriptome with a novel machine-learning model. DNA Res 2020; 26:365-378. [PMID: 31321403 PMCID: PMC6704403 DOI: 10.1093/dnares/dsz015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 06/18/2019] [Indexed: 02/07/2023] Open
Abstract
Very little is known about long non-coding RNAs (lncRNAs) in the mammalian olfactory sensory epithelia. Deciphering the non-coding transcriptome in olfaction is relevant because these RNAs have been shown to play a role in chromatin modification and nuclear architecture reorganization, processes that accompany olfactory differentiation and olfactory receptor gene choice, one of the most poorly understood gene regulatory processes in mammals. In this study, we used a combination of in silico and ex vivo approaches to uncover a comprehensive catalogue of olfactory lncRNAs and to investigate their expression in the mouse olfactory organs. Initially, we used a novel machine-learning lncRNA classifier to discover hundreds of annotated and unannotated lncRNAs, some of which were predicted to be preferentially expressed in the main olfactory epithelium and the vomeronasal organ, the most important olfactory structures in the mouse. Moreover, we used whole-tissue and single-cell RNA sequencing data to discover lncRNAs expressed in mature sensory neurons of the main epithelium. Candidate lncRNAs were further validated by in situ hybridization and RT-PCR, leading to the identification of lncRNAs found throughout the olfactory epithelia, as well as others exquisitely expressed in subsets of mature olfactory neurons or progenitor cells.
Collapse
Affiliation(s)
- Antonio P Camargo
- Department of Genetics and Evolution, Institute of Biology, University of Campinas, Campinas, SP, Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Thiago S Nakahara
- Department of Genetics and Evolution, Institute of Biology, University of Campinas, Campinas, SP, Brazil.,Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Luiz E R Firmino
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Paulo H M Netto
- Department of Genetics and Evolution, Institute of Biology, University of Campinas, Campinas, SP, Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - João B P do Nascimento
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Elisa R Donnard
- Molecular Oncology Center, Hospital Sirio-Libanes, Sao Paulo, SP, Brazil
| | - Pedro A F Galante
- Molecular Oncology Center, Hospital Sirio-Libanes, Sao Paulo, SP, Brazil
| | - Marcelo F Carazzolle
- Department of Genetics and Evolution, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Bettina Malnic
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Fabio Papes
- Department of Genetics and Evolution, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
74
|
Structural, morphometric and immunohistochemical study of the rabbit accessory olfactory bulb. Brain Struct Funct 2019; 225:203-226. [PMID: 31802255 DOI: 10.1007/s00429-019-01997-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 11/23/2019] [Indexed: 10/25/2022]
Abstract
The accessory olfactory bulb (AOB) is the first neural integrative centre of the vomeronasal system (VNS), which is associated primarily with the detection of semiochemicals. Although the rabbit is used as a model for the study of chemocommunication, these studies are hampered by the lack of knowledge regarding the topography, lamination, and neurochemical properties of the rabbit AOB. To fill this gap, we have employed histological stainings: lectin labelling with Ulex europaeus (UEA-I), Bandeiraea simplicifolia (BSI-B4), and Lycopersicon esculentum (LEA) agglutinins, and a range of immunohistochemical markers. Anti-G proteins Gαi2/Gαo, not previously studied in the rabbit AOB, are expressed following an antero-posterior zonal pattern. This places Lagomorpha among the small groups of mammals that conserve a double-path vomeronasal reception. Antibodies against olfactory marker protein (OMP), growth-associated protein-43 (GAP-43), glutaminase (GLS), microtubule-associated protein-2 (MAP-2), glial fibrillary-acidic protein (GFAP), calbindin (CB), and calretinin (CR) characterise the strata and the principal components of the BOA, demonstrating several singular features of the rabbit AOB. This diversity is accentuated by the presence of a unique organisation: four neuronal clusters in the accessory bulbar white matter, two of them not previously characterised in any species (the γ and δ groups). Our morphometric study of the AOB has found significant differences between sexes in the numerical density of principal cells, with larger values in females, a pattern completely opposite to that found in rats. In summary, the rabbit possesses a highly developed AOB, with many specific features that highlight the significant role played by chemocommunication among this species.
Collapse
|
75
|
The Homeodomain Transcription Factors Vax1 and Six6 Are Required for SCN Development and Function. Mol Neurobiol 2019; 57:1217-1232. [PMID: 31705443 DOI: 10.1007/s12035-019-01781-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/12/2019] [Indexed: 12/13/2022]
Abstract
The brain's primary circadian pacemaker, the suprachiasmatic nucleus (SCN), is required to translate day-length and circadian rhythms into neuronal, hormonal, and behavioral rhythms. Here, we identify the homeodomain transcription factor ventral anterior homeobox 1 (Vax1) as required for SCN development, vasoactive intestinal peptide expression, and SCN output. Previous work has shown that VAX1 is required for gonadotropin-releasing hormone (GnRH/LHRH) neuron development, a neuronal population controlling reproductive status. Surprisingly, the ectopic expression of a Gnrh-Cre allele (Gnrhcre) in the SCN confirmed the requirement of both VAX1 (Vax1flox/flox:Gnrhcre, Vax1Gnrh-cre) and sine oculis homeobox protein 6 (Six6flox/flox:Gnrhcre, Six6Gnrh-cre) in SCN function in adulthood. To dissociate the role of Vax1 and Six6 in GnRH neuron and SCN function, we used another Gnrh-cre allele that targets GnRH neurons, but not the SCN (Lhrhcre). Both Six6Lhrh-cre and Vax1Lhrh-cre were infertile, and in contrast to Vax1Gnrh-cre and Six6Gnrh-cre mice, Six6Lhrh-cre and Vax1Lhrh-cre had normal circadian behavior. Unexpectedly, ~ 1/4 of the Six6Gnrh-cre mice were unable to entrain to light, showing that ectopic expression of Gnrhcre impaired function of the retino-hypothalamic tract that relays light information to the brain. This study identifies VAX1, and confirms SIX6, as transcription factors required for SCN development and function and demonstrates the importance of understanding how ectopic CRE expression can impact the results.
Collapse
|
76
|
Li J, Liu T, Dong Y, Kondoh K, Lu Z. Trans-synaptic Neural Circuit-Tracing with Neurotropic Viruses. Neurosci Bull 2019; 35:909-920. [PMID: 31004271 PMCID: PMC6754522 DOI: 10.1007/s12264-019-00374-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/15/2018] [Indexed: 12/19/2022] Open
Abstract
A central objective in deciphering the nervous system in health and disease is to define the connections of neurons. The propensity of neurotropic viruses to spread among synaptically-linked neurons makes them ideal for mapping neural circuits. So far, several classes of viral neuronal tracers have become available and provide a powerful toolbox for delineating neural networks. In this paper, we review the recent developments of neurotropic viral tracers and highlight their unique properties in revealing patterns of neuronal connections.
Collapse
Affiliation(s)
- Jiamin Li
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Taian Liu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yun Dong
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Kunio Kondoh
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institute of Natural Sciences, Myodaiji, Okazaki, Aichi, 444-8585, Japan.
- Japan Science and Technology Agency, PRESTO, Myodaiji, Okazaki, Aichi, 444-8585, Japan.
| | - Zhonghua Lu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
77
|
Karaoglan M, Çolakoğlu Er H. The relationship between the olfactory bulb and precocious puberty: from the nose to the pituitary. J Pediatr Endocrinol Metab 2019; 32:1013-1021. [PMID: 31377742 DOI: 10.1515/jpem-2018-0534] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 06/17/2019] [Indexed: 11/15/2022]
Abstract
Background/objective The olfactory bulb (OB) and pubertal development have a close relationship as they share a common ontogenetic origin. The aim of this study was to analyze the potential relationship between precocious puberty (PP) in girls as a sign of early pubertal timing and their OB volume as an indicator of its functional activity. Design In the study group (n = 125), OB volume, pituitary height (PH), body mass index (BMI) and body surface (S) variables were retrospectively investigated in 49 girls included in the PP group and 76 healthy girls constituting the control group. Volumetric and length measurements were performed on a magnetic resonance imaging (MRI) scan by using manual segmentation of slices. Results The mean OB volume (73.41 ± 17.21 mm3) and PH (4.96 ± 1.01 mm) were significantly higher in the PP group (p = 0.001 and p = 0.001, respectively). The mean volume difference between the right and left bulbs (1.52 ± 1.87) was higher in the PP group (p = 0.03). The body surface (1.05 ± 0.16 m2) was larger in the PP group (p = 0.09). There was a high correlation between the OB volume and PH (r125 = 0.716). There was a moderate correlation between the body surface and OB volume (r125 = 654), and a weak correlation between the former (S) and the PH (r125 = 452). Conclusions This study showed that there is a strong correlation between increased OB volume and PH in cases with PP. It indicates that increased OB volume may be a strong clue that olfactory functions play a role in pubertal timing in humans, although it does not show definitive proof of a causal relationship.
Collapse
Affiliation(s)
- Murat Karaoglan
- Department of Pediatric Endocrinology, Gaziantep University Faculty of Medicine, 27070 Gaziantep, Turkey
| | - Hale Çolakoğlu Er
- Department of Radiology, Gaziantep University Faculty of Medicine, Gaziantep, Turkey
| |
Collapse
|
78
|
Bittman EL. Circadian Function in Multiple Cell Types Is Necessary for Proper Timing of the Preovulatory LH Surge. J Biol Rhythms 2019; 34:622-633. [PMID: 31530063 DOI: 10.1177/0748730419873511] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The timing of the preovulatory surge of luteinizing hormone (LH), which occurs on the evening of proestrus in female mice, is determined by the circadian system. The identity of cells that control the phase of the LH surge is unclear: evidence supports a role of arginine vasopressin (AVP) cells of the suprachiasmatic nucleus (SCN), but it is not known whether vasopressinergic neurons are necessary or sufficient to account for circadian control of ovulation. Among other cell types, evidence also suggests important roles of circadian function of kisspeptin cells of the anteroventral periventricular nucleus (AvPV) and gonadotropin-releasing hormone (GnRH) neurons of the preoptic area (POA), whose discharge is immediately responsible for the discharge of LH from the anterior pituitary. The present studies used an ovariectomized, estradiol-treated preparation to determine critical cell types whose clock function is critical to the timing of LH secretion. As expected, the LH surge occurred at or shortly after ZT12 in control mice. In further confirmation of circadian control, the surge was advanced by 2 h in tau mutant animals. The timing of the surge was altered to varying degrees by conditional deletion of Bmal1 in AVPCre, KissCreBAC, and GnRHCreBAC mice. Excision of the mutant Cnsk1e (tau) allele in AVP neurons resulted in a reversion of the surge to the ZT12. Conditional deletion of Bmal1 in Kiss1 or GnRH neurons had no noticeable effect on locomotor rhythms, but targeting of AVP neurons produced variable effects on circadian period that did not always correspond to changes in the phase of LH secretion. The results indicate that circadian function in multiple cell types is necessary for proper timing of the LH surge.
Collapse
Affiliation(s)
- Eric L Bittman
- Department of Biology and Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts
| |
Collapse
|
79
|
Pandolfi EC, Tonsfeldt KJ, Hoffmann HM, Mellon PL. Deletion of the Homeodomain Protein Six6 From GnRH Neurons Decreases GnRH Gene Expression, Resulting in Infertility. Endocrinology 2019; 160:2151-2164. [PMID: 31211355 PMCID: PMC6821215 DOI: 10.1210/en.2019-00113] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/11/2019] [Indexed: 12/12/2022]
Abstract
Hypothalamic GnRH (luteinizing hormone-releasing hormone) neurons are crucial for the hypothalamic-pituitary-gonadal (HPG) axis, which regulates mammalian fertility. Insufficient GnRH disrupts the HPG axis and is often associated with the genetic condition idiopathic hypogonadotropic hypogonadism (IHH). The homeodomain protein sine oculis-related homeobox 6 (Six6) is required for the development of GnRH neurons. Although it is known that Six6 is specifically expressed within a more mature GnRH neuronal cell line and that overexpression of Six6 induces GnRH transcription in these cells, the direct role of Six6 within the GnRH neuron in vivo is unknown. Here we find that global Six6 knockout (KO) embryos show apoptosis of GnRH neurons beginning at embryonic day 14.5 with 90% loss of GnRH neurons by postnatal day 1. We sought to determine whether the hypogonadism and infertility reported in the Six6KO mice are generated via actions within the GnRH neuron in vivo by creating a Six6-flox mouse and crossing it with the LHRHcre mouse. Loss of Six6 specifically within the GnRH neuron abolished GnRH expression in ∼0% of GnRH neurons. We further demonstrated that deletion of Six6 only within the GnRH neuron leads to infertility, hypogonadism, hypogonadotropism, and delayed puberty. We conclude that Six6 plays distinct roles in maintaining fertility in the GnRH neuron vs in the migratory environment of the GnRH neuron by maintaining expression of GnRH and survival of GnRH neurons, respectively. These results increase knowledge of the role of Six6 in the brain and may offer insight into the mechanism of IHH.
Collapse
Affiliation(s)
- Erica C Pandolfi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California
| | - Karen J Tonsfeldt
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California
| | - Hanne M Hoffmann
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California
- Department of Animal Science, Michigan State University, East Lansing, Michigan
| | - Pamela L Mellon
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
80
|
Muir ER, Biju KC, Cong L, Rogers WE, Torres Hernandez E, Duong TQ, Clark RA. Functional MRI of the mouse olfactory system. Neurosci Lett 2019; 704:57-61. [PMID: 30951799 PMCID: PMC11113078 DOI: 10.1016/j.neulet.2019.03.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/18/2019] [Accepted: 03/31/2019] [Indexed: 10/27/2022]
Abstract
Although olfactory dysfunction is an early warning sign of Alzheimer's and Parkinson's diseases, and is commonly present in a range of other neurodegenerative disorders, the mechanisms for its pathogenesis are not yet clear. Since fMRI allows the mapping of spatial and temporal patterns of activity in multiple brain regions simultaneously, it serves as a powerful tool to study olfactory dysfunction in animal models of neurodegenerative diseases. Nonetheless, there have been no reports to date of mapping odor-induced activation patterns beyond the olfactory bulb to the extended networks of olfactory and limbic archicortex, likely due to the small size of the mouse brain. Therefore, using an 11.7 T magnet and a blood volume-weighted fMRI technique, we mapped the functional neuroanatomy of the mouse olfactory system. Consistent with reports on imaging of the much larger human brain, we mapped activity in regions of the olfactory bulb, as well as olfactory and limbic archicortex. By using two distinct odorants, we further demonstrated odorant-specific activation patterns. Our work thus provides a methodological framework for fMRI studies of olfactory dysfunction in mouse models of neurodegeneration.
Collapse
Affiliation(s)
- Eric R Muir
- Research Imaging Institute, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, United States; Department of Ophthalmology, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, United States
| | - K C Biju
- Department of Medicine, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, United States
| | - Linlin Cong
- Research Imaging Institute, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, United States; Department of Biomedical Engineering, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, United States
| | - William E Rogers
- Research Imaging Institute, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, United States
| | - Enrique Torres Hernandez
- Department of Medicine, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, United States
| | - Timothy Q Duong
- Research Imaging Institute, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, United States; Department of Ophthalmology, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, United States; South Texas Veterans Health Care System, 7400 Merton Minter Blvd, San Antonio, TX, 78229, United States
| | - Robert A Clark
- Department of Medicine, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, United States; South Texas Veterans Health Care System, 7400 Merton Minter Blvd, San Antonio, TX, 78229, United States.
| |
Collapse
|
81
|
Yang L, Jiang H, Wang Y, Lei Y, Chen J, Sun N, Lv W, Wang C, Near TJ, He S. Expansion of vomeronasal receptor genes ( OlfC) in the evolution of fright reaction in Ostariophysan fishes. Commun Biol 2019; 2:235. [PMID: 31263779 PMCID: PMC6588630 DOI: 10.1038/s42003-019-0479-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 05/28/2019] [Indexed: 12/15/2022] Open
Abstract
Ostariophysans are the most diverse group of freshwater fishes and feature a pheromone-elicited fright reaction. However, the genetic basis of fright reaction is unclear. Here, we compared vomeronasal type 2 receptor-like (OlfC) genes from fishes having and lacking fright reaction, to provide insight into evolution of pheromonal olfaction in fishes. We found OlfC genes expanded remarkably in ostariophysans having fright reaction compared with fishes lacking fright reaction. Phylogenetic analysis indicates OlfC subfamily 9 expanded specifically in ostariophysans having fright reaction. Principle component and phylogenetic logistic regression analysis partitioned fishes by ecotype (having or lacking fright reaction) and identified OlfC subfamily 9 as being an important factor for fright reaction. Expression levels of expanded OlfC subfamily genes after fright reaction in zebrafish changed more than did genes that had not expanded. Furthermore, evidence of positive selection was found in the expanded OlfC proteins in ostariophysan fishes having fright reaction. These results provide new insight into the genetic basis of fright reaction in ostariophysan fish and will enable future research into the mechanism of action of OlfC proteins.
Collapse
Affiliation(s)
- Liandong Yang
- The Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072 People’s Republic of China
| | - Haifeng Jiang
- The Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072 People’s Republic of China
- University of Chinese Academy of Sciences, 100049 Beijing, People’s Republic of China
| | - Ying Wang
- School of Life Sciences, Jianghan University, 430056 Wuhan, People’s Republic of China
| | - Yi Lei
- The Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072 People’s Republic of China
- University of Chinese Academy of Sciences, 100049 Beijing, People’s Republic of China
| | - Juan Chen
- The Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072 People’s Republic of China
- University of Chinese Academy of Sciences, 100049 Beijing, People’s Republic of China
| | - Ning Sun
- The Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072 People’s Republic of China
- University of Chinese Academy of Sciences, 100049 Beijing, People’s Republic of China
| | - Wenqi Lv
- The Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072 People’s Republic of China
- University of Chinese Academy of Sciences, 100049 Beijing, People’s Republic of China
| | - Cheng Wang
- The Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072 People’s Republic of China
- University of Chinese Academy of Sciences, 100049 Beijing, People’s Republic of China
| | - Thomas J. Near
- Department of Ecology and Evolutionary Biology and Peabody Museum of Natural History, Yale University, New Haven, CT 06520 USA
| | - Shunping He
- The Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072 People’s Republic of China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223 Kunming, People’s Republic of China
| |
Collapse
|
82
|
Yao W, Liu W, Deng K, Wang Z, Wang DH, Zhang XY. GnRH expression and cell proliferation are associated with seasonal breeding and food hoarding in Mongolian gerbils (Meriones unguiculatus). Horm Behav 2019; 112:42-53. [PMID: 30922890 DOI: 10.1016/j.yhbeh.2019.03.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 03/18/2019] [Accepted: 03/22/2019] [Indexed: 12/14/2022]
Abstract
Seasonal brain plasticity contributes to a variety of physiological and behavioral processes. We hypothesized that variations in GnRH expression and cell proliferation facilitated seasonal breeding and food hoarding. Here, we reported seasonal changes in sexual and social behavior, GnRH expression and brain cell proliferation, and the role of photoperiod in inducing seasonal breeding and brain plasticity in Mongolian gerbils (Meriones unguiculatus). The gerbils captured in April and July had more mature sexual development, higher exploratory behavior, and preferred novelty much more than those captured in September. Male gerbils captured in April and July had consistently higher GnRH expression than those captured in September. GnRH expression was also found to be suppressed by food-induced hoarding behavior in the breeding season. Both subadult and adult gerbils from April and July had higher cell proliferation in SVZ, hypothalamus and amygdala compared to those in September. However, adult gerbils captured in September preferred familiar objects, and no seasonal differences were found in cell proliferation in hippocampal dentate gyrus among the three seasons. The laboratory study showed that photoperiod alone did not alter reproductive traits, behavior, cell proliferation or cell survival in the detected brain regions. These findings suggest that the structural variations in GnRH expression in hypothalamus and cell proliferation in hypothalamus, amygdala and hippocampus are associated with seasonal breeding and food hoarding in gerbils. It gives a new insight into the proximate physiological and neural basis for these seasonal life-history traits of breeding and food hoarding in small mammals.
Collapse
Affiliation(s)
- Wei Yao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Health Sciences, Anhui University, Hefei, Anhui 230601, China
| | - Wei Liu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ke Deng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zuoxin Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306-1270, USA
| | - De-Hua Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Xue-Ying Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
83
|
Ramzan F, Phung T, Swift-Gallant A, Coome LA, Holmes MM, Monks DA. Both neural and global androgen receptor overexpression affect sexual dimorphism in the mouse brain. J Neuroendocrinol 2019; 31:e12715. [PMID: 30920021 DOI: 10.1111/jne.12715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 03/14/2019] [Accepted: 03/22/2019] [Indexed: 01/28/2023]
Abstract
Testosterone is the main endocrine mechanism mediating sexual differentiation of the mammalian brain, although testosterone signalling is complex and important mechanistic questions remain. Notably, the extent to which testosterone acts via androgen receptors (AR) in this process remains unknown and it is also not clear where testosterone acts in the body to produce sexual dimorphisms in neuroanatomy. To address these questions, we used a transgenic mouse model of Cre/loxP-driven AR overexpression in which AR was induced selectively in neural tissue (Nestin-cre) or in all tissues (CMV-cre). We then studied sexually dimorphic features of several well-characterised sexual dimorphisms: calbindin-immunoreactive neurones in the medial preoptic area (CALB-SDN), tyrosine hydroxylase neurones in the anteroventral periventricular nucleus, and vasopressin-immunoreactive neurones originating in the bed nucleus of the stria terminalis and their projections in the lateral septum. We additionally evaluated oestrogen receptor α immunoreactivity in these nuclei. Briefly, we found that global but not neural overexpression of AR resulted in masculinisation of CALB-SDN nucleus volume, cell number and cell size in transgenic females. Furthermore, neural AR overexpression resulted in increased oestrogen receptor α staining in females compared to males in the medial preoptic area. AR overexpression did not affect other measures. Overall, the results of the present study provide support for the hypothesis that androgenic mechanisms external to the nervous system can affect sexual differentiation of the brain.
Collapse
Affiliation(s)
- Firyal Ramzan
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Thanh Phung
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Ashlyn Swift-Gallant
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Lindsay A Coome
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Melissa M Holmes
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - D Ashley Monks
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
84
|
Meinsohn MC, Smith OE, Bertolin K, Murphy BD. The Orphan Nuclear Receptors Steroidogenic Factor-1 and Liver Receptor Homolog-1: Structure, Regulation, and Essential Roles in Mammalian Reproduction. Physiol Rev 2019; 99:1249-1279. [DOI: 10.1152/physrev.00019.2018] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Nuclear receptors are intracellular proteins that act as transcription factors. Proteins with classic nuclear receptor domain structure lacking identified signaling ligands are designated orphan nuclear receptors. Two of these, steroidogenic factor-1 (NR5A1, also known as SF-1) and liver receptor homolog-1 (NR5A2, also known as LRH-1), bind to the same DNA sequences, with different and nonoverlapping effects on targets. Endogenous regulation of both is achieved predominantly by cofactor interactions. SF-1 is expressed primarily in steroidogenic tissues, LRH-1 in tissues of endodermal origin and the gonads. Both receptors modulate cholesterol homeostasis, steroidogenesis, tissue-specific cell proliferation, and stem cell pluripotency. LRH-1 is essential for development beyond gastrulation and SF-1 for genesis of the adrenal, sexual differentiation, and Leydig cell function. Ovary-specific depletion of SF-1 disrupts follicle development, while LRH-1 depletion prevents ovulation, cumulus expansion, and luteinization. Uterine depletion of LRH-1 compromises decidualization and pregnancy. In humans, SF-1 is present in endometriotic tissue, where it regulates estrogen synthesis. SF-1 is underexpressed in ovarian cancer cells and overexpressed in Leydig cell tumors. In breast cancer cells, proliferation, migration and invasion, and chemotherapy resistance are regulated by LRH-1. In conclusion, the NR5A orphan nuclear receptors are nonredundant factors that are crucial regulators of a panoply of biological processes, across multiple reproductive tissues.
Collapse
Affiliation(s)
- Marie-Charlotte Meinsohn
- Centre de Recherche en Reproduction et Fertilité, Université de Montréal, St-Hyacinthe, Québec, Canada
| | - Olivia E. Smith
- Centre de Recherche en Reproduction et Fertilité, Université de Montréal, St-Hyacinthe, Québec, Canada
| | - Kalyne Bertolin
- Centre de Recherche en Reproduction et Fertilité, Université de Montréal, St-Hyacinthe, Québec, Canada
| | - Bruce D. Murphy
- Centre de Recherche en Reproduction et Fertilité, Université de Montréal, St-Hyacinthe, Québec, Canada
| |
Collapse
|
85
|
Yeo SH, Kyle V, Blouet C, Jones S, Colledge WH. Mapping neuronal inputs to Kiss1 neurons in the arcuate nucleus of the mouse. PLoS One 2019; 14:e0213927. [PMID: 30917148 PMCID: PMC6436706 DOI: 10.1371/journal.pone.0213927] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/04/2019] [Indexed: 12/20/2022] Open
Abstract
The normal function of the mammalian reproductive axis is strongly influenced by physiological, metabolic and environmental factors. Kisspeptin neuropeptides, encoded by the Kiss1 gene, are potent regulators of the mammalian reproductive axis by stimulating gonadodropin releasing hormone secretion from the hypothalamus. To understand how the reproductive axis is modulated by higher order neuronal inputs we have mapped the afferent circuits into arcuate (ARC) Kiss1 neurons. We used a transgenic mouse that expresses the CRE recombinase in Kiss1 neurons for conditional viral tracing with genetically modified viruses. CRE-mediated activation of these viruses in Kiss1 neurons allows the virus to move transynaptically to label neurons with primary or secondary afferent inputs into the Kiss1 neurons. Several regions of the brain showed synaptic connectivity to arcuate Kiss1 neurons including proopiomelanocortin neurons in the ARC itself, kisspeptin neurons in the anteroventral periventricular nucleus, vasopressin neurons in the supraoptic and suprachiasmatic nuclei, thyrotropin releasing neurons in the paraventricular nucleus and unidentified neurons in other regions including the subfornical organ, amygdala, interpeduncular nucleus, ventral premammilary nucleus, basal nucleus of stria terminalis and the visual, somatosensory and piriform regions of the cortex. These data provide an insight into how the activity of Kiss1 neurons may be regulated by metabolic signals and provide a detailed neuroanatomical map for future functional studies.
Collapse
Affiliation(s)
- Shel-Hwa Yeo
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Victoria Kyle
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Clemence Blouet
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Susan Jones
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - William Henry Colledge
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
86
|
Bayless DW, Yang T, Mason MM, Susanto AAT, Lobdell A, Shah NM. Limbic Neurons Shape Sex Recognition and Social Behavior in Sexually Naive Males. Cell 2019; 176:1190-1205.e20. [PMID: 30712868 PMCID: PMC6453703 DOI: 10.1016/j.cell.2018.12.041] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/13/2018] [Accepted: 12/21/2018] [Indexed: 12/18/2022]
Abstract
Sexually naive animals have to distinguish between the sexes because they show species-typical interactions with males and females without meaningful prior experience. However, central neural pathways in naive mammals that recognize sex of other individuals remain poorly characterized. We examined the role of the principal component of the bed nucleus of stria terminalis (BNSTpr), a limbic center, in social interactions in mice. We find that activity of aromatase-expressing BNSTpr (AB) neurons appears to encode sex of other animals and subsequent displays of mating in sexually naive males. Silencing these neurons in males eliminates preference for female pheromones and abrogates mating success, whereas activating them even transiently promotes male-male mating. Surprisingly, female AB neurons do not appear to control sex recognition, mating, or maternal aggression. In summary, AB neurons represent sex of other animals and govern ensuing social behaviors in sexually naive males.
Collapse
Affiliation(s)
- Daniel W Bayless
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Taehong Yang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Matthew M Mason
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Albert A T Susanto
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Alexandra Lobdell
- Department of Psychiatry and Behavioral Sciences, 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.
| |
Collapse
|
87
|
Ye Y, Zhuang Y, Smeets MAM, Zhou W. Human chemosignals modulate emotional perception of biological motion in a sex-specific manner. Psychoneuroendocrinology 2019; 100:246-253. [PMID: 30390523 DOI: 10.1016/j.psyneuen.2018.10.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/15/2018] [Accepted: 10/17/2018] [Indexed: 01/05/2023]
Abstract
Androsta-4,16,-dien-3-one and estra-1,3,5(10),16-tetraen-3-ol have previously been shown to communicate opposite sex information that is differently effective to the two sex groups. The current study critically examines if the two human steroids could facilitate interactions with potential mates rather than competitors by acting on the recipients' emotional perception in a sex-appropriate manner. Using dynamic point-light displays that portray the gaits of walkers whose emotional states are digitally morphed along the valence and the arousal axes, we show that smelling androstadienone subconsciously biases heterosexual women, but not men, towards perceiving the male, but not female, walkers as happier and more relaxed. By contrast, smelling estratetraenol subconsciously biases heterosexual men, but not women, towards perceiving the female, but not male, walkers as happier and more relaxed. These findings indicate that androstadienone and estratetraenol prime the identification of emotionally receptive states for the potential mates with whom they are associated, in manners contingent upon not only the recipients' own sex but also their sex perception of other individuals that ensure sex-appropriate behavior.
Collapse
Affiliation(s)
- Yuting Ye
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuan Zhuang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Monique A M Smeets
- Unilever R&D, Vlaardingen, 3133 AT, The Netherlands; Department of Social, Health & Organizational Psychology, Utrecht University, Utrecht, 3584 CS, The Netherlands
| | - Wen Zhou
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
88
|
da Silva Pacheco S, Araujo Rondini T, Cioni Bittencourt J, Fuzeti Elias C. Neurons expressing estrogen receptor α differentially innervate the periaqueductal gray matter of female rats. J Chem Neuroanat 2019; 97:33-42. [PMID: 30703434 DOI: 10.1016/j.jchemneu.2019.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 11/19/2022]
Abstract
The periaqueductal gray matter (PAG) is a brainstem site involved in distinct autonomic and behavioral responses. Among them, the motor control of female sexual behavior, including lordosis, is well described. Lordosis reflex is highly dependent on increasing levels of estradiol that occur in the afternoon of the proestrus day in normally cycling females. This effect is thought to be mediated primarily via actions in the ventromedial nucleus of the hypothalamus (VMH). By binding to estrogen receptor α (ERα), estradiol changes the activity of VMH neurons that project to the PAG. Evidence also exists for the coordination of PAG outputs by estradiol-responsive neurons outside the VMH. However, a comprehensive analysis of these circuitries is not available. Using stereotaxic injection of the retrograde tracer Fluorogold in distinct columns of the PAG we performed a systematic mapping of neurons innervating the PAG and those coexpressing ERα immunoreactivity. We found that the forebrain projections to PAG columns are largely segregated and that most of the ERα expressing neurons preferentially target the lateral and the ventrolateral columns. Dual labeled neurons were mostly found in the intermediate subdivision of the lateral septal nucleus, the posterior aspect of the medial bed nucleus of the stria terminalis, the medial preoptic nucleus, the striohypothalamic nucleus and the ventrolateral VMH. Few dual labeled neurons were also observed in the arcuate nucleus, in the posterodorsal subdivision of the medial nucleus of the amygdala and in the ventral premammillary nucleus. Our findings indicate that ERα modulates sexual behavior in female rats via an integrated neural network that differentially innervate the columns of the PAG.
Collapse
Affiliation(s)
- Silvana da Silva Pacheco
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-900, Brazil; University Hospital, University of São Paulo, São Paulo, SP 05508-900, Brazil
| | | | - Jackson Cioni Bittencourt
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-900, Brazil
| | - Carol Fuzeti Elias
- Departments of Molecular and Integrative Physiology and of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109-5622, United States.
| |
Collapse
|
89
|
Hoffmann HM, Larder R, Lee JS, Hu RJ, Trang C, Devries BM, Clark DD, Mellon PL. Differential CRE Expression in Lhrh-cre and GnRH-cre Alleles and the Impact on Fertility in Otx2-Flox Mice. Neuroendocrinology 2019; 108:328-342. [PMID: 30739114 PMCID: PMC6753941 DOI: 10.1159/000497791] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 02/06/2019] [Indexed: 12/12/2022]
Abstract
There is an increasing trend in studies utilizing cell-specific deletion of genes through conditional gene deletion by CRE recombination. Despite numerous advantages, this strategy also has limitations such as ectopic CRE-expression and germline recombination. Two commonly used gonadotropin-releasing hormone (Gnrh)-driven CRE-expressing mice both target GnRH neurons. However, a direct comparison of the cells targeted and their phenotypic outcome have not yet been presented. To compare where recombination takes place, we crossed the Gnrh-cre and Lhrh-cre lines with the Rosa26-LacZ reporter mouse. Lhrh-cre allowed recombination of the Rosa26-LacZ gene in ∼700 cells, which is comparable to the GnRH neuronal population. Surprisingly, there were > 20 times more LacZ expressing cells in the adult Gnrh-cre:Rosa26-LacZ than the Lhrh-cre:Rosa26-LacZ brain. The greatest differences in targeting of the Gnrh-cre and Lhrh-cre lines were found in the septum, the suprachiasmatic nucleus, and the septohypothalamic area. This difference in cells targeted was present from embryonic day 12. A prior study using the Gnrh-cre to delete the transcription factor Otx2 found fewer GnRH neurons, leading to male and female subfertility. To recapitulate this study, we performed a fertility assay in Otx2:Lhrh-cre mice. We confirmed the requirement for Otx2 in GnRH neuron development, fertility and correct gonadotropin hormone release in Otx2:Lhrh-cre males, but the subfertility was more modest than in Otx2:Gnrh-cre and absent in female Otx2:Lhrh-cre. This suggests that ectopic expression of Gnrh-cre contributes to the reproductive phenotype observed. Finally, the Cre alleles caused germline recombination of the flox allele when transmitted from either parent, generating embryonic lethal knock-out offspring, producing smaller live litters.
Collapse
Affiliation(s)
- Hanne M Hoffmann
- Department of Obstetrics and Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, California, USA
- Department of Animal Science, Michigan State University, East Lansing, Michigan, USA
| | - Rachel Larder
- Department of Obstetrics and Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, California, USA
| | - Jessica S Lee
- Department of Obstetrics and Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, California, USA
| | - Rachael J Hu
- Department of Obstetrics and Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, California, USA
| | - Crystal Trang
- Department of Obstetrics and Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, California, USA
| | - Brooke M Devries
- Department of Animal Science, Michigan State University, East Lansing, Michigan, USA
| | - Daniel D Clark
- Department of Obstetrics and Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, California, USA
| | - Pamela L Mellon
- Department of Obstetrics and Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, California, USA,
| |
Collapse
|
90
|
Costa RA, Ferreira IR, Cintra HA, Gomes LHF, Guida LDC. Genotype-Phenotype Relationships and Endocrine Findings in Prader-Willi Syndrome. Front Endocrinol (Lausanne) 2019; 10:864. [PMID: 31920975 PMCID: PMC6923197 DOI: 10.3389/fendo.2019.00864] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022] Open
Abstract
Prader-Willi syndrome (PWS) is a complex imprinting disorder related to genomic errors that inactivate paternally-inherited genes on chromosome 15q11-q13 with severe implications on endocrine, cognitive and neurologic systems, metabolism, and behavior. The absence of expression of one or more genes at the PWS critical region contributes to different phenotypes. There are three molecular mechanisms of occurrence: paternal deletion of the 15q11-q13 region; maternal uniparental disomy 15; or imprinting defects. Although there is a clinical diagnostic consensus criteria, DNA methylation status must be confirmed through genetic testing. The endocrine system can be the most affected in PWS, and growth hormone replacement therapy provides improvement in growth, body composition, and behavioral and physical attributes. A key feature of the syndrome is the hypothalamic dysfunction that may be the basis of several endocrine symptoms. Clinical and molecular complexity in PWS enhances the importance of genetic diagnosis in therapeutic definition and genetic counseling. So far, no single gene mutation has been described to contribute to this genetic disorder or related to any exclusive symptoms. Here we proposed to review individually disrupted genes within the PWS critical region and their reported clinical phenotypes related to the syndrome. While genes such as MKRN3, MAGEL2, NDN, or SNORD115 do not address the full spectrum of PWS symptoms and are less likely to have causal implications in PWS major clinical signs, SNORD116 has emerged as a critical, and possibly, a determinant candidate in PWS, in the recent years. Besides that, the understanding of the biology of the PWS SNORD genes is fairly low at the present. These non-coding RNAs exhibit all the hallmarks of RNA methylation guides and can be incorporated into ribonucleoprotein complexes with possible hypothalamic and endocrine functions. Also, DNA conservation between SNORD sequences across placental mammals strongly suggests that they have a functional role as RNA entities on an evolutionary basis. The broad clinical spectrum observed in PWS and the absence of a clear genotype-phenotype specific correlation imply that the numerous genes involved in the syndrome have an additive deleterious effect on different phenotypes when deficiently expressed.
Collapse
|
91
|
Moffitt JR, Bambah-Mukku D, Eichhorn SW, Vaughn E, Shekhar K, Perez JD, Rubinstein ND, Hao J, Regev A, Dulac C, Zhuang X. Molecular, spatial, and functional single-cell profiling of the hypothalamic preoptic region. Science 2018; 362:eaau5324. [PMID: 30385464 PMCID: PMC6482113 DOI: 10.1126/science.aau5324] [Citation(s) in RCA: 761] [Impact Index Per Article: 108.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/21/2018] [Indexed: 12/23/2022]
Abstract
The hypothalamus controls essential social behaviors and homeostatic functions. However, the cellular architecture of hypothalamic nuclei-including the molecular identity, spatial organization, and function of distinct cell types-is poorly understood. Here, we developed an imaging-based in situ cell-type identification and mapping method and combined it with single-cell RNA-sequencing to create a molecularly annotated and spatially resolved cell atlas of the mouse hypothalamic preoptic region. We profiled ~1 million cells, identified ~70 neuronal populations characterized by distinct neuromodulatory signatures and spatial organizations, and defined specific neuronal populations activated during social behaviors in male and female mice, providing a high-resolution framework for mechanistic investigation of behavior circuits. The approach described opens a new avenue for the construction of cell atlases in diverse tissues and organisms.
Collapse
Affiliation(s)
- Jeffrey R Moffitt
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Dhananjay Bambah-Mukku
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Stephen W Eichhorn
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Eric Vaughn
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Karthik Shekhar
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Julio D Perez
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Nimrod D Rubinstein
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Junjie Hao
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Aviv Regev
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
- Koch Institute of Integrative Cancer Biology, Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Catherine Dulac
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA.
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
92
|
Pandolfi EC, Hoffmann HM, Schoeller EL, Gorman MR, Mellon PL. Haploinsufficiency of SIX3 Abolishes Male Reproductive Behavior Through Disrupted Olfactory Development, and Impairs Female Fertility Through Disrupted GnRH Neuron Migration. Mol Neurobiol 2018; 55:8709-8727. [PMID: 29589282 PMCID: PMC6156938 DOI: 10.1007/s12035-018-1013-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 03/16/2018] [Indexed: 12/17/2022]
Abstract
Mating behavior in males and females is dependent on olfactory cues processed through both the main olfactory epithelium (MOE) and the vomeronasal organ (VNO). Signaling through the MOE is critical for the initiation of male mating behavior, and the loss of MOE signaling severely compromises this comportment. Here, we demonstrate that dosage of the homeodomain gene Six3 affects the degree of development of MOE but not the VNO. Anomalous MOE development in Six3 heterozygote mice leads to hyposmia, specifically disrupting male mounting behavior by impairing detection of volatile female estrus pheromones. Six3 is highly expressed in the MOE, main olfactory bulb (MOB), and hypothalamus; all regions essential in the proper migration of the gonadotropin-releasing hormone (GnRH) neurons, a key reproductive neuronal population that migrates along olfactory axons from the developing nose into the brain. Interestingly, we find that the reduction in Six3 expression in Six3 heterozygote mice compromises development of the MOE and MOB, resulting in mis-migration of GnRH neurons due to improper olfactory axon targeting. This reduction in the hypothalamic GnRH neuron population, by 45% in adulthood, leads to female subfertility, but does not impact male hormone levels, suggesting that male infertility is not related to GnRH neuron numbers, but exclusively linked to abnormal olfaction. We here determine that Six3 is haploinsufficient for MOE development, GnRH neuron migration, and fertility, and represents a novel candidate gene for Kallmann syndrome, a form of inherited infertility.
Collapse
Affiliation(s)
- Erica C Pandolfi
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0674, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Hanne M Hoffmann
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0674, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Erica L Schoeller
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0674, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Michael R Gorman
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Psychology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Pamela L Mellon
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0674, USA.
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
93
|
Hoffmann HM, Pandolfi EC, Larder R, Mellon PL. Haploinsufficiency of Homeodomain Proteins Six3, Vax1, and Otx2 Causes Subfertility in Mice via Distinct Mechanisms. Neuroendocrinology 2018; 109:200-207. [PMID: 30261489 PMCID: PMC6437011 DOI: 10.1159/000494086] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 09/26/2018] [Indexed: 12/17/2022]
Abstract
Haploinsufficiency occurs when loss of one copy of a diploid gene (hemizygosity) causes a phenotype. It is relatively rare, in that most genes can produce sufficient mRNA and protein from a single copy to prevent any loss of normal activity and function. Reproduction is a complex process relying on migration of GnRH neurons from the olfactory placode to the hypothalamus during development. We have studied 3 different homeodomain genes Otx2, Vax1, and Six3 and found that the deletion of one allele for any of these genes in mice produces subfertility or infertility in one or both sexes, despite the presence of one intact allele. All 3 heterozygous mice have reduced numbers of GnRH neurons, but the mechanisms of subfertility differ significantly. This review compares the subfertility phenotypes and their mechanisms.
Collapse
Affiliation(s)
- Hanne M Hoffmann
- Department of Obstetrics, Gynecology, and Reproductive Sciences and the Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California, USA
- Department of Animal Science, Michigan State University, East Lansing, Michigan, USA
| | - Erica C Pandolfi
- Department of Obstetrics, Gynecology, and Reproductive Sciences and the Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California, USA
| | - Rachel Larder
- Department of Obstetrics, Gynecology, and Reproductive Sciences and the Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California, USA
| | - Pamela L Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences and the Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California, USA,
| |
Collapse
|
94
|
Dynamic and Sex-Specific Changes in Gonadotropin-Releasing Hormone Neuron Activity and Excitability in a Mouse Model of Temporal Lobe Epilepsy. eNeuro 2018; 5:eN-NWR-0273-18. [PMID: 30255128 PMCID: PMC6153338 DOI: 10.1523/eneuro.0273-18.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 12/28/2022] Open
Abstract
Reproductive endocrine disorders are prominent comorbidities of temporal lobe epilepsy (TLE) in both men and women. The neural mechanisms underlying these comorbidities remain unclear, but hypothalamic gonadotropin-releasing hormone (GnRH) neurons may be involved. Here, we report the first direct demonstrations of aberrant GnRH neuron function in an animal model of epilepsy. Recordings of GnRH neuron firing and excitability were made in acute mouse brain slices prepared two months after intrahippocampal injection of kainate (KA) or control saline, a well-established TLE model in which most females develop comorbid estrous cycle disruption. GnRH neurons from control females showed elevated firing and excitability on estrus compared with diestrus. By contrast, cells from KA-injected females that developed prolonged, disrupted estrous cycles (KA-long) showed the reverse pattern. Firing rates of cells from KA-injected females that maintained regular cycles (KA-regular) were not different from controls on diestrus, but were reduced on estrus. In KA-injected males, only GnRH neurons in the medial septum displayed elevated firing. In contrast to the diestrus versus estrus and sex-specific changes in firing, GnRH neuron intrinsic excitability was elevated in all KA-injected groups, indicating a role for afferent synaptic and neuromodulatory inputs in shaping overall changes in firing activity. Furthermore, KA-injected females showed cycle-stage-specific changes in circulating sex steroids on diestrus and estrus that also differed between KA-long and KA-regular groups. Together, these findings reveal that the effects of epilepsy on the neural control of reproduction are dynamic across the estrous cycle, distinct in association with comorbid estrous cycle disruption severity, and sex-specific.
Collapse
|
95
|
Moore AM, Prescott M, Czieselsky K, Desroziers E, Yip SH, Campbell RE, Herbison AE. Synaptic Innervation of the GnRH Neuron Distal Dendron in Female Mice. Endocrinology 2018; 159:3200-3208. [PMID: 30010812 DOI: 10.1210/en.2018-00505] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 07/05/2018] [Indexed: 11/19/2022]
Abstract
GnRH neuron cell bodies are scattered throughout the basal forebrain but funnel their projections to the median eminence to release GnRH into the pituitary portal system to control fertility. Prior studies have shown that GnRH neurons located in the anterior hypothalamus send projections to the median eminence that have characteristics of both dendrites and axons. These unusual structures have been termed "dendrons." To address whether the dendron is unique to anterior hypothalamic GnRH neurons or is also a characteristic of more rostral GnRH neurons, we used viral vector‒mediated GnRH neuron‒specific tract-tracing coupled with CLARITY optical clearing. Individual rostral preoptic area GnRH neurons in female mice were identified to elaborate processes up to 4 mm in length that exhibited spines and projected all the way to the median eminence before branching into multiple short axons. The synaptic innervation patterns of distal GnRH neuron dendrons and their short axons in the vicinity of the median eminence were examined using electron microscopy. This revealed the presence of a high density of synaptic inputs to distal dendrons at the border of the median eminence. In contrast, no synapses were detected on any GnRH neuron axons. These studies demonstrate that GnRH neurons in the rostral preoptic area project dendrons to the edge of the median eminence, whereupon they branch into multiple short axons responsible for GnRH secretion. The dense synaptic innervation of these distal dendrons likely represents an efficient mechanism for controlling GnRH secretion required for fertility.
Collapse
Affiliation(s)
- Aleisha M Moore
- Centre for Neuroendocrinology, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Mel Prescott
- Centre for Neuroendocrinology, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Katja Czieselsky
- Centre for Neuroendocrinology, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Elodie Desroziers
- Centre for Neuroendocrinology, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Siew Hoong Yip
- Centre for Neuroendocrinology, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
96
|
Spergel DJ. Neuropeptidergic modulation of GnRH neuronal activity and GnRH secretion controlling reproduction: insights from recent mouse studies. Cell Tissue Res 2018; 375:179-191. [DOI: 10.1007/s00441-018-2893-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 07/06/2018] [Indexed: 12/18/2022]
|
97
|
Seo JH, Pyo S, Shin YK, Nam BG, Kang JW, Kim KP, Lee HY, Cho SR. The Effect of Environmental Enrichment on Glutathione-Mediated Xenobiotic Metabolism and Antioxidation in Normal Adult Mice. Front Neurol 2018; 9:425. [PMID: 30022966 PMCID: PMC6039562 DOI: 10.3389/fneur.2018.00425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 05/22/2018] [Indexed: 12/12/2022] Open
Abstract
Olfactory bulb (OB) plays an important role in protecting against harmful substances via the secretion of antioxidant and detoxifying enzymes. Environmental enrichment (EE) is a common rehabilitation method and known to have beneficial effects in the central nervous system. However, the effects of EE in the OB still remain unclear. At 6 weeks of age, CD-1® (ICR) mice were assigned to standard cages or EE cages. After 2 months, we performed proteomic analysis. Forty-four up-regulated proteins were identified in EE mice compared to the control mice. Gene Ontology analysis and Kyoto Encyclopedia of Genes and Genomes Pathway demonstrated that the upregulated proteins were mainly involved in metabolic pathways against xenobiotics. Among those upregulated proteins, 9 proteins, which participate in phase I or II of the xenobiotic metabolizing process and are known to be responsible for ROS detoxification, were validated by qRT-PCR. To explore the effect of ROS detoxification mediated by EE, glutathione activity was measured by an ELISA assay. The ratio of reduced glutathione to oxidized glutathione was significantly increased in EE mice. Based on a linear regression analysis, GSTM2 and UGT2A1 were found to be the most influential genes in ROS detoxification. For further analysis of neuroprotection, the level of iNOS and the ratio of Bax to Bcl-2 were significantly decreased in EE mice. While TUNEL+ cells were significantly decreased, Ki67+ cells were significantly increased in EE mice, implicating that EE creates an optimal state for xenobiotic metabolism and antioxidant activity. Taken together, our results suggested that EE protects olfactory layers via the upregulation of glutathione-related antioxidant and xenobiotic metabolizing enzymes, eventually lowering ROS-mediated inflammation and apoptosis and increasing neurogenesis. This study may provide an opportunity for a better understanding of the beneficial effects of EE in the OB.
Collapse
Affiliation(s)
- Jung Hwa Seo
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Soonil Pyo
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Yoon-Kyum Shin
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Bae-Geun Nam
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea.,Graduate Program of NanoScience and Technology, Yonsei University, Seoul, South Korea
| | - Jeong Won Kang
- Department of Applied Chemistry, Kyung Hee University, Yongin, South Korea.,Precision Medicine Branch, Research Institute, National Cancer Center, Goyang, South Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, Kyung Hee University, Yongin, South Korea
| | - Hoo Young Lee
- Department of Medicine, The Graduate School of Yonsei University, Seoul, South Korea.,TBI Rehabilitation Center, National Traffic Injury Rehabilitation Hospital, Yangpyeong, South Korea.,Department of Rehabilitation Medicine, School of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Rae Cho
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea.,Graduate Program of NanoScience and Technology, Yonsei University, Seoul, South Korea.,Department of Medicine, The Graduate School of Yonsei University, Seoul, South Korea.,Yonsei Stem Cell Research Center, Avison Biomedical Research Center, Seoul, South Korea.,Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
98
|
Russo C, Russo A, Pellitteri R, Stanzani S. Ghrelin-containing neurons in the olfactory bulb send collateralized projections into medial amygdaloid and arcuate hypothalamic nuclei: neuroanatomical study. Exp Brain Res 2018; 236:2223-2229. [DOI: 10.1007/s00221-018-5298-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 05/23/2018] [Indexed: 10/16/2022]
|
99
|
Tan CL, Sheard PW, Jasoni CL. Developing neurites from mouse basal forebrain gonadotropin-releasing hormone neurons use Sonic hedgehog to modulate their growth. Int J Dev Neurosci 2018; 68:89-97. [PMID: 29787797 DOI: 10.1016/j.ijdevneu.2018.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/14/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023] Open
Abstract
Hypothalamic gonadotropin-releasing hormone (GnRH) neurons are required for fertility in all mammalian species studied to date. GnRH neuron cell bodies reside in the basal forebrain, and most extend long neurites in the caudal direction to terminate at the median eminence (ME), the site of hormone secretion. Using in vitro neurite growth assays, histological methods, and genetic deletion strategies in mice we have analysed the role of the morphogen and neurite growth and guidance molecule, Sonic hedgehog (Shh), in the growth of GnRH neurites to their target. Immunohistochemistry revealed that Shh was present in the basal forebrain, the preoptic area (POA) and mediobasal hypothalamus (MBH) at gestational day 14.5 (GD 14.5), a time when GnRH neurites grow towards the ME. Furthermore, in situ hybridization revealed that mRNA encoding the Shh receptor, Smoothened (Smo), was present in GnRH neurons from GD 15.5, when the first GnRH neurites are extending towards the MBH. In vitro neurite growth assays using hypothalamic explants from GD 15.5 fetuses in 3-D collagen gels showed that Shh was able to significantly stimulate GnRH neurite outgrowth. Finally, genetic deletion of Smo specifically from GnRH neurons in vivo, using Cre-loxP technology, resulted in a significant decrease in GnRH neurites innervating the ME. These experiments demonstrate that GnRH neurites use Shh for their neurite development, provide further understanding of the mechanisms by which GnRH nerve terminals arrive at their site of hormone secretion, and identify an additional hypothalamic neuronal population for which Shh/Smo signaling is developmentally important.
Collapse
Affiliation(s)
- C L Tan
- Department of Anatomy, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand; Centre for Neuroendocrinology, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand.
| | - P W Sheard
- Department of Physiology, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand.
| | - C L Jasoni
- Department of Anatomy, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand; Centre for Neuroendocrinology, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand.
| |
Collapse
|
100
|
Burger LL, Vanacker C, Phumsatitpong C, Wagenmaker ER, Wang L, Olson DP, Moenter SM. Identification of Genes Enriched in GnRH Neurons by Translating Ribosome Affinity Purification and RNAseq in Mice. Endocrinology 2018; 159. [PMID: 29522155 PMCID: PMC6287592 DOI: 10.1210/en.2018-00001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons are a nexus of fertility regulation. We used translating ribosome affinity purification coupled with RNA sequencing to examine messenger RNAs of GnRH neurons in adult intact and gonadectomized (GDX) male and female mice. GnRH neuron ribosomes were tagged with green fluorescent protein (GFP) and GFP-labeled polysomes isolated by immunoprecipitation, producing one RNA fraction enhanced for GnRH neuron transcripts and one RNA fraction depleted. Complementary DNA libraries were created from each fraction and 50-base, paired-end sequencing done and differential expression (enhanced fraction/depleted fraction) determined with a threshold of >1.5- or <0.66-fold (false discovery rate P ≤ 0.05). A core of ∼840 genes was differentially expressed in GnRH neurons in all treatments, including enrichment for Gnrh1 (∼40-fold), and genes critical for GnRH neuron and/or gonadotrope development. In contrast, non-neuronal transcripts were not enriched or were de-enriched. Several epithelial markers were also enriched, consistent with the olfactory epithelial origins of GnRH neurons. Interestingly, many synaptic transmission pathways were de-enriched, in accordance with relatively low innervation of GnRH neurons. The most striking difference between intact and GDX mice of both sexes was a marked downregulation of genes associated with oxidative phosphorylation and upregulation of glucose transporters in GnRH neurons from GDX mice. This may suggest that GnRH neurons switch to an alternate fuel to increase adenosine triphosphate production in the absence of negative feedback when GnRH release is elevated. Knowledge of the GnRH neuron translatome and its regulation can guide functional studies and can be extended to disease states, such as polycystic ovary syndrome.
Collapse
Affiliation(s)
- Laura L Burger
- Department of Molecular and Integrative Physiology, University of Michigan, Ann
Arbor, Michigan
| | - Charlotte Vanacker
- Department of Molecular and Integrative Physiology, University of Michigan, Ann
Arbor, Michigan
| | | | - Elizabeth R Wagenmaker
- Department of Molecular and Integrative Physiology, University of Michigan, Ann
Arbor, Michigan
| | - Luhong Wang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann
Arbor, Michigan
| | - David P Olson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann
Arbor, Michigan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Suzanne M Moenter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann
Arbor, Michigan
- Department of Internal Medicine, University of Michigan, Ann Arbor,
Michigan
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor,
Michigan
- Correspondence: Laura L. Burger, PhD, University of Michigan, 7725 Med Sci II, 1137 E. Catherine
Street, Ann Arbor, Michigan 48109-5622. E-mail:
| |
Collapse
|