1
|
Parmaksiz D, Kim Y. Navigating Central Oxytocin Transport: Known Realms and Uncharted Territories. Neuroscientist 2025; 31:234-261. [PMID: 39113465 PMCID: PMC12103645 DOI: 10.1177/10738584241268754] [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] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Complex mechanisms govern the transport and action of oxytocin (Oxt), a neuropeptide and hormone that mediates diverse physiologic processes. While Oxt exerts site-specific and rapid effects in the brain via axonal and somatodendritic release, volume transmission via CSF and the neurovascular interface can act as an additional mechanism to distribute Oxt signals across distant brain regions on a slower timescale. This review focuses on modes of Oxt transport and action in the CNS, with particular emphasis on the roles of perivascular spaces, the blood-brain barrier (BBB), and circumventricular organs in coordinating the triadic interaction among circulating blood, CSF, and parenchyma. Perivascular spaces, critical conduits for CSF flow, play a pivotal role in Oxt diffusion and distribution within the CNS and reciprocally undergo Oxt-mediated structural and functional reconstruction. While the BBB modulates the movement of Oxt between systemic and cerebral circulation in a majority of brain regions, circumventricular organs without a functional BBB can allow for diffusion, monitoring, and feedback regulation of bloodborne peripheral signals such as Oxt. Recognition of these additional transport mechanisms provides enhanced insight into the systemic propagation and regulation of Oxt activity.
Collapse
Affiliation(s)
- Deniz Parmaksiz
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
2
|
Zdon ST, Silva MSB, Navarro VM. Sexually dimorphic distribution of Kiss1 neurons in the bed nucleus of the stria terminalis. J Neuroendocrinol 2025:e70049. [PMID: 40421488 DOI: 10.1111/jne.70049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 05/02/2025] [Accepted: 05/08/2025] [Indexed: 05/28/2025]
Abstract
Kiss1 neurons play a crucial role in reproductive function and are found in distinct brain regions, including the bed nucleus of the stria terminalis (BNST). However, the sexual dimorphism of Kiss1 neurons in the BNST and their projections has not been fully characterized. This study examined the distribution and projections of Kiss1 neurons in the anterior (aBNST) and principal (prBNST) regions of the BNST in male and female Kiss1-Cre and Kiss1-Cre; tdTomatoloxP/+ mice. Neuroanatomical analysis and tracing experiments were conducted to quantify Kiss1 neurons and map their projections. Males had approximately a threefold higher number of Kiss1 neurons in the prBNST than females, while no significant sex difference was observed in the aBNST. Viral tracing experiments revealed sexually dimorphic projections of Kiss1adBNST neurons, with females displaying more diverse projections to various brain regions involved in reproduction and social behaviors. Kiss1prBNST neurons project exclusively to the zona incerta and adBNST in both sexes, while females exhibited additional projections to the RP3V and PVH. The sexually dimorphic distribution and projections of Kiss1BNST neurons suggest their potential role in modulating sex-specific behaviors and neuroendocrine functions. This neuroanatomical sexual dimorphism may contribute to sex differences in social and reproductive behaviors associated with BNST function, providing new insights into the neural basis of sex-specific behaviors and reproductive regulation.
Collapse
Affiliation(s)
- Samuel T Zdon
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Mauro S B Silva
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Victor M Navarro
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Sweet SR, Biddinger JE, Zimmermann JB, Yu GL, Simerly RB. Postnatal hyperosmolality alters development of hypothalamic feeding circuits with context-specific changes in ingestive behavior. iScience 2025; 28:112284. [PMID: 40469114 PMCID: PMC12136832 DOI: 10.1016/j.isci.2025.112284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/24/2025] [Accepted: 03/20/2025] [Indexed: 06/11/2025] Open
Abstract
Drinking and feeding are tightly coordinated homeostatic events and the paraventricular nucleus of the hypothalamus (PVH) represents a possible node of neural integration for signals related to energy and fluid homeostasis. We used TRAP2;Ai14 mice and Fos labeling to visualize neurons in the PVH and median preoptic nucleus (MEPO) responding to both water deprivation and feeding signals. We determined that structural and functional development of dehydration-sensitive inputs to the PVH precedes those of agouti-related peptide (AgRP) neurons, which convey hunger signals and are known to be developmentally programmed by nutrition. Moreover, we found that osmotic hyperstimulation of neonatal mice led to enhanced AgRP inputs to the PVH in adulthood, as well as disruptions to ingestive behaviors during high-fat diet feeding and dehydration-anorexia. Thus, development of feeding circuits is impacted not only by nutritional signals, but also by early perturbations to fluid homeostasis with context-specific consequences for coordination of ingestive behavior.
Collapse
Affiliation(s)
- Serena R. Sweet
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
| | - Jessica E. Biddinger
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
| | - Jessie B. Zimmermann
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Gina L. Yu
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Richard B. Simerly
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
4
|
Hanchate NK. Single-cell genomics meets systems neuroscience: Insights from mapping the brain circuitry of stress. J Neuroendocrinol 2025; 37:e70005. [PMID: 39956535 PMCID: PMC12045673 DOI: 10.1111/jne.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 12/26/2024] [Accepted: 02/03/2025] [Indexed: 02/18/2025]
Abstract
Responses to external and internal dangers is essential for survival and homeostatic regulation. Hypothalamic corticotropin-releasing hormone neurons (CRHNs) play a pivotal role in regulating neuroendocrine responses to fear and stress. In recent years, the application of neurogenetic tools, such as fiber photometry, chemogenetics and optogenetics, have provided new insights into the dynamic neuronal responses of CRHNs during stressful events, offering new perspectives into their functional significance in mediating neurobehavioural responses to stress. Transsynaptic viral tracers have facilitated the comprehensive mapping of neuronal inputs to CRHNs. Furthermore, the development and application of innovative single-cell genomic tools combined with viral tracing have begun to pave the way for a deeper understanding of the transcriptional profiles of neural circuit components, enabling molecular-anatomical circuit mapping. Here, I will discuss how these systems neuroscience approaches and novel single-cell genomic methods are advancing the molecular and functional mapping of stress neurocircuits, their associated challenges and future directions.
Collapse
Affiliation(s)
- Naresh K. Hanchate
- Genetics & Genomic Medicine DepartmentUCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
| |
Collapse
|
5
|
Morales L, Desfilis E, Medina L. Development of catecholaminergic neurons of Otp-lineage in the medial extended amygdala and related forebrain centers. Front Neuroanat 2025; 19:1553952. [PMID: 40177299 PMCID: PMC11961924 DOI: 10.3389/fnana.2025.1553952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Catecholaminergic (CA) neurons of the medial extended amygdala, preoptic region and adjacent alar hypothalamus have been involved in different aspects of social behavior, as well as in modulation of homeostasis in response to different stressors. Previous data suggested that at least some CA neurons of the medial extended amygdala could originate in a hypothalamic embryonic domain that expresses the transcription factor Otp. To investigate this, we used Otp-eGFP mice (with permanent labeling of GFP in Otp cells) to analyze coexpression of GFP and tyrosine hydroxylase (TH) throughout ontogenesis by way of double immunofluorescence. Our results provide evidence that some forebrain CA cells belong to the Otp lineage. In particular, we found small subpopulations of TH cells that coexpress GFP within the medial extended amygdala, the periventricular preoptic area, the paraventricular hypothalamus, the periventricular hypothalamus, as well as some subdivisions of the basal hypothalamus. In some of the Otp cells, such as those of extended amygdala, the expression of TH appears to be transitory, in agreement with previous studies. The results open interesting questions about the role of these Otp versus non-Otp catecholaminergic subpopulations during development, network integration and in modulation of different functions, including homeostasis and social behaviors.
Collapse
Affiliation(s)
- Lorena Morales
- Department of Medicina Experimental, Universitat de Lleida, Lleida, Spain
- Laboratory of Evolutionary Developmental Neurobiology, Lleida’s Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Lleida, Spain
| | - Ester Desfilis
- Department of Medicina Experimental, Universitat de Lleida, Lleida, Spain
- Laboratory of Evolutionary Developmental Neurobiology, Lleida’s Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Lleida, Spain
| | - Loreta Medina
- Department of Medicina Experimental, Universitat de Lleida, Lleida, Spain
- Laboratory of Evolutionary Developmental Neurobiology, Lleida’s Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Lleida, Spain
| |
Collapse
|
6
|
Jeong H, Chon Y, Yoon S, Choi EK, Lee N, Oh JK, Chung YA, Song IU. Structural and functional alterations in hypothalamic subregions in male patients with alcohol use disorder. Drug Alcohol Depend 2025; 268:112554. [PMID: 39848134 DOI: 10.1016/j.drugalcdep.2025.112554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/29/2024] [Accepted: 01/04/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND The hypothalamus is involved in stress regulation and reward processing, with its various nuclei exhibiting unique functions and connections. However, human neuroimaging studies on the hypothalamic subregions are limited in drug addiction. This study examined the volumes and functional connectivity of hypothalamic subregions in individuals with alcohol use disorder (AUD). METHOD The study included 24 male patients with AUD who had maintained abstinence and 24 healthy male controls, all of whom underwent brain structural and resting-state functional magnetic resonance imaging. The hypothalamus was segmented into five subunits using a deep learning-based algorithm, with comparisons of volumes and functional connectivity (FC) between the two groups. The relationships between these measures and alcohol-related characteristics were examined in the AUD group. RESULTS Findings indicated lower volumes in the anterior-superior (corrected-p < 0.001) and tuberal-superior subunits (corrected-p = 0.002) and altered FC of these and the anterior-inferior subunit among AUD patients (corrected-p < 0.05). Moreover, greater disease severity and a longer history of heavy drinking correlated with lower volumes in the anterior-superior (r = -0.42, p = 0.045) and tuberal-superior subregions (r = -0.61, p = 0.013), respectively. Conversely, a longer abstinence duration was associated with larger volumes in the anterior-superior (r = 0.56, p = 0.008) and tuberal-superior subunits (r = 0.40, p = 0.048) and with higher FC between the tuberal-superior hypothalamus and the thalamus, caudate, and anterior cingulate cortex (r = 0.55, p = 0.014). CONCLUSIONS Our results suggest that specific regional alterations within the hypothalamus, particularly the superior subregions, are associated with AUD, and more importantly, that these alterations may be reversible with prolonged abstinence.
Collapse
Affiliation(s)
- Hyeonseok Jeong
- Department of Radiology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Neurology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Younghoon Chon
- Department of Psychiatry, Incheon Chamsarang Hospital, Incheon, Republic of Korea
| | - Sujung Yoon
- Ewha Brain Institute, Ewha W. University, Seoul, Republic of Korea; Department of Brain and Cognitive Sciences, Ewha W. University, Seoul, Republic of Korea
| | - Eun Kyoung Choi
- Department of Nuclear Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Narae Lee
- Department of Psychiatry, Incheon Chamsarang Hospital, Incheon, Republic of Korea
| | - Jin Kyoung Oh
- Department of Nuclear Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Yong-An Chung
- Department of Nuclear Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - In-Uk Song
- Department of Neurology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
7
|
Patel TA, Gao L, Boomer SH, Liu X, Patel KP, Zheng H. Downregulation of neuronal nitric oxide synthase (nNOS) within the paraventricular nucleus in Ins2 Akita-type-1 diabetic mice contributes to sympatho-excitation. Nitric Oxide 2025; 154:1-7. [PMID: 39521242 PMCID: PMC11729414 DOI: 10.1016/j.niox.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/10/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Activation of both renin-angiotensin system (RAS) and the sympathetic system is the primary etiologic event in developing cardiovascular complications in diabetes mellitus (DM). However, the precise mechanisms for sympathetic activation in DM have not been elucidated. Here we attempted to investigate diabetes-linked cardiovascular dysregulation due to angiotensin II (Ang II)-mediated reduction in neuronal nitric oxide (NO) synthase (nNOS) within the paraventricular neuleus (PVN). In the present study, we used Ins2+/-Akita (a spontaneous, insulin-dependent genetic diabetic non-obese murine model) and wild-type (WT) littermates mice as controls. At 14 weeks of age, we found the Akita mice had increased renal sympathetic nerve activity and elevated levels of plasma norepinephrine. There was decreased expression of nNOS protein (Akita 0.43 ± 0.11 vs. WT 0.75 ± 0.05, P < 0.05) in the PVN of Akita mice. Akita mice had increased expression of angiotensin-converting enzyme (ACE) (Akita 0.58 ± 0.05 vs. WT 0.34 ± 0.04, P < 0.05) and Ang II type 1 receptor (Akita 0.49 ± 0.03 vs. WT 0.29 ± 0.09, P < 0.05), decreased expressions of ACE2 (Akita 0.17 ± 0.05 vs. WT 0.27 ± 0.03, P < 0.05) and angiotensin (1-7) Mas receptor (Akita 0.46 ± 0.02 vs. WT 0.77 ± 0.07, P < 0.05). Futher, there were increased protein levels of protein inhibitor of nNOS (PIN) (Akita 1.75 ± 0.08 vs. WT 0.71 ± 0.09, P < 0.05) with concomitantly decreased catalytically active dimers of nNOS (Akita 0.11 ± 0.04 vs. WT 0.19 ± 0.02, P < 0.05) in the PVN in Akita mice. Our studies suggest that activation of the excitatory arm of RAS, leads to a decrease NO, causing an over-activation of the sympathetic drive in DM.
Collapse
Affiliation(s)
- Tapan A Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198-5850, USA
| | - Lie Gao
- Department of Anesthesiology, University of Nebraska Medical Center, USA
| | - Shane H Boomer
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Xuefei Liu
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Kaushik P Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198-5850, USA
| | - Hong Zheng
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA.
| |
Collapse
|
8
|
Martins Dos Santos K, Saunders SE, Antunes VR, Boychuk CR. Insulin activates parasympathetic hepatic-related neurons of the paraventricular nucleus of the hypothalamus through mTOR signaling. J Neurophysiol 2025; 133:320-332. [PMID: 39665212 PMCID: PMC11918334 DOI: 10.1152/jn.00284.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/05/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024] Open
Abstract
Integration of autonomic and metabolic regulation, including hepatic function, is a critical role played by the brain's hypothalamic region. Specifically, the paraventricular nucleus of the hypothalamus (PVN) regulates autonomic functions related to metabolism, such as hepatic glucose production. Although insulin can act directly on hepatic tissue to inhibit hepatic glucose production, recent evidence implicates that central actions of insulin within PVN also regulate glucose metabolism. However, specific central circuits responsible for insulin signaling with relation to hepatic regulation are poorly understood. As a heterogeneous nucleus essential to controlling parasympathetic motor output with notable expression of insulin receptors, PVN is an appealing target for insulin-dependent modulation of parasympathetic activity. Here, we tested the hypothesis that insulin activates hepatic-related PVN (PVNhepatic) neurons through a parasympathetic pathway. Using transsynaptic retrograde tracing, labeling within PVN was first identified 24 h after its expression in the dorsal motor nucleus of the vagus (DMV) and 72 h after hepatic injection. Critically, nearly all labeling in medial PVN was abolished after a left vagotomy, indicating that PVNhepatic neurons in this region are part of a central circuit innervating parasympathetic motor neurons. Insulin also significantly increased the firing frequency of PVNhepatic neurons in this subregion. Mechanistically, rapamycin pretreatment inhibited insulin-dependent activation of PVNhepatic neurons. Therefore, central insulin signaling can activate a subset of PVNhepatic neurons that are part of a unique parasympathetic network in control of hepatic function. Taken together, PVNhepatic neurons related to parasympathetic output regulation could serve as a key central network in insulin's ability to control hepatic functions.NEW & NOTEWORTHY Increased peripheral insulin concentrations are known to decrease hepatic glucose production through both direct actions on hepatocytes and central autonomic networks. Despite this understanding, how (and in which brain regions) insulin exerts its action is still obscure. Here, we demonstrate that insulin activates parasympathetic hepatic-related PVN neurons (PVNhepatic) and that this effect relies on mammalian target of rapamycin (mTOR) signaling, suggesting that insulin modulates hepatic function through autonomic pathways involving insulin receptor intracellular signaling cascades.
Collapse
Affiliation(s)
- Karoline Martins Dos Santos
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, United States
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sandy E Saunders
- Dalton Cardiovascular Research Center, Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
| | - Vagner R Antunes
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carie R Boychuk
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, United States
- Dalton Cardiovascular Research Center, Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
9
|
Holt MK, Valderrama N, Polanco MJ, Hayter I, Badenoch EG, Trapp S, Rinaman L. Modulation of stress-related behaviour by preproglucagon neurons and hypothalamic projections to the nucleus of the solitary tract. Mol Metab 2025; 91:102076. [PMID: 39603502 PMCID: PMC11667184 DOI: 10.1016/j.molmet.2024.102076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/12/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024] Open
Abstract
Stress-induced behaviours are driven by complex neural circuits and some neuronal populations concurrently modulate diverse behavioural and physiological responses to stress. Glucagon-like peptide-1 (GLP-1)-producing preproglucagon (PPG) neurons within the lower brainstem caudal nucleus of the solitary tract (cNTS) are particularly sensitive to stressful stimuli and are implicated in multiple physiological and behavioural responses to interoceptive and psychogenic threats. However, the afferent inputs driving stress-induced activation of PPG neurons are largely unknown, and the role of PPG neurons in anxiety-like behaviour is controversial. Through chemogenetic manipulations we reveal that cNTS PPG neurons have the ability to moderately increase anxiety-like behaviours in mice in a sex-dependent manner. Using an intersectional approach, we show that input from the paraventricular nucleus of the hypothalamus (PVN) drives activation of both the cNTS as a whole and PPG neurons in particular in response to acute restraint stress, but that while this input is rich in corticotropin-releasing hormone (CRH), PPG neurons do not express significant levels of receptors for CRH and are not activated following lateral ventricle delivery of CRH. Finally, we demonstrate that cNTS-projecting PVN neurons are necessary for the ability of restraint stress to suppress food intake in male mice. Our findings reveal sex differences in behavioural responses to PPG neural activation and highlight a hypothalamic-brainstem pathway in stress-induced hypophagia.
Collapse
Affiliation(s)
- Marie K Holt
- Florida State University, Department of Psychology and Program in Neuroscience, Tallahassee, FL, USA; Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, UCL, London, UK; University of Warwick, School of Life Sciences, Coventry, UK.
| | - Natalia Valderrama
- Florida State University, Department of Psychology and Program in Neuroscience, Tallahassee, FL, USA
| | - Maria J Polanco
- Florida State University, Department of Psychology and Program in Neuroscience, Tallahassee, FL, USA
| | - Imogen Hayter
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, UCL, London, UK; GlaxoSmithKline Pharmaceuticals, London, UK
| | | | - Stefan Trapp
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, UCL, London, UK
| | - Linda Rinaman
- Florida State University, Department of Psychology and Program in Neuroscience, Tallahassee, FL, USA
| |
Collapse
|
10
|
Li S, Li W, Miao Y, Gao M, Jia Y, Chen Z, Chen X, Pan T, Zhang S, Xing Z, Han S, Sun XL, Wei X, Liu Z, Zhou W, Wu W, Liu F, Han L, Zhu H, Ye H, Liu L, Li Y, Zhang P, Gong J, Tian Y, Ai Y, Cao P, Wu D, Qi X, Gui S, Wu QF. Modeling craniopharyngioma for drug screening reveals a neuronal mechanism for tumor growth. Sci Transl Med 2024; 16:eadn6763. [PMID: 39693408 DOI: 10.1126/scitranslmed.adn6763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 07/15/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024]
Abstract
Tumors occurring along the hypothalamus-pituitary axis receive axonal projection from neuroendocrine neurons, but it remains unclear whether neuroendocrine neuronal activity drives tumor expansion. Craniopharyngioma is a common suprasellar tumor with a propensity for invading the hypothalamus, leading to devastating endocrine and metabolic disorders. Here, we developed two autochthonous animal models that faithfully recapitulate the molecular pathology, clinical manifestations, and transcriptomic profiles of papillary craniopharyngioma. Using high-throughput drug screening, we identified 74 compounds with potent antitumor efficacy. The administration of (S)-amlodipine besylate achieved tumor regression in vivo, potentially by abrogating calcium transients and neuron-to-tumor chemical transmission. Chemogenetic manipulation of neuroendocrine neuronal activity bidirectionally regulated tumor cell growth in our mouse model, suggesting that craniopharyngioma hijacks hypothalamic neurons to promote tumor progression. These findings deepen our understanding of suprasellar tumor biology and offer promising avenues for clinical exploration of effective chemotherapies.
Collapse
Affiliation(s)
- Si Li
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Li
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Yuqi Miao
- Department of Endocrinology, Beijing Children's Hospital, Capital Medical University, Beijing 100045, China
| | | | - Yanfei Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Zhenhua Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xi Chen
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| | | | - Shuangfeng Zhang
- National Institute of Biological Sciences, Beijing 102206, China
| | - Zhifang Xing
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Shuping Han
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xue-Lian Sun
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaochan Wei
- BGI Research, Shenzhen 518083, China
- BGI Research, Hangzhou 310030, China
| | - Zhiming Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Wentao Zhou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Wentao Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Fangzheng Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Lei Han
- BGI Research, Hangzhou 310030, China
| | | | - Hongying Ye
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | | | - Yinqing Li
- IDG/McGovern Institute for Brain Research, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing 100045, China
| | - Jian Gong
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Yongji Tian
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Youwei Ai
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Peng Cao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Di Wu
- Department of Endocrinology, Beijing Children's Hospital, Capital Medical University, Beijing 100045, China
| | - Xiangbing Qi
- National Institute of Biological Sciences, Beijing 102206, China
| | - Songbai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Qing-Feng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing 100045, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
11
|
Biddinger JE, Elson AET, Fathi PA, Sweet SR, Nishimori K, Ayala JE, Simerly RB. AgRP neurons mediate activity-dependent development of oxytocin connectivity and autonomic regulation. Proc Natl Acad Sci U S A 2024; 121:e2403810121. [PMID: 39585985 PMCID: PMC11626166 DOI: 10.1073/pnas.2403810121] [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] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/24/2024] [Indexed: 11/27/2024] Open
Abstract
During postnatal life, leptin specifies neuronal inputs to the paraventricular nucleus of the hypothalamus (PVH) and activates agouti-related peptide (AgRP) neurons in the arcuate nucleus of the hypothalamus. Activity-dependent developmental mechanisms impact refinement of sensory circuits, but whether leptin-mediated postnatal neuronal activity specifies hypothalamic neural projections is largely unexplored. Here, we used chemogenetics to manipulate the activity of AgRP neurons during a discrete postnatal critical period and evaluated the development of AgRP inputs to the PVH and descending efferent outflow to the dorsal vagal complex (DVC). In leptin-deficient mice, targeting of AgRP neuronal outgrowth to PVH oxytocin neurons was reduced, and despite the lack of leptin receptors found on oxytocin neurons in the PVH, oxytocin-containing connections to the DVC were also impaired. Activation of AgRP neurons during early postnatal life not only normalized AgRP inputs to the PVH but also oxytocin outputs to the DVC in leptin-deficient mice. Blocking AgRP neuron activity during the same postnatal period reduced the density of AgRP inputs to the PVH of wild type mice, as well as the density of oxytocin-containing DVC fibers, and these innervation deficits were associated with dysregulated autonomic function. These findings suggest that leptin-mediated AgRP neuronal activity is required for the development of PVH connectivity and represents a unique activity-dependent mechanism for specification of neural pathways involved in the hypothalamic integration of autonomic responses.
Collapse
Affiliation(s)
- Jessica E. Biddinger
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN37232
| | - Amanda E. T. Elson
- Developmental Neuroscience Program, The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA90027
| | - Payam A. Fathi
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN37232
| | - Serena R. Sweet
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN37232
| | - Katsuhiko Nishimori
- Department of Obesity and Internal Inflammation, Fukushima Medical University, Fukushima City960-1295, Japan
| | - Julio E. Ayala
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN37232
| | - Richard B. Simerly
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN37232
| |
Collapse
|
12
|
Sweet SR, Biddinger JE, Zimmermann JB, Yu GL, Simerly RB. Early perturbations to fluid homeostasis alter development of hypothalamic feeding circuits with context-specific changes in ingestive behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620307. [PMID: 39484367 PMCID: PMC11527132 DOI: 10.1101/2024.10.25.620307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Drinking and feeding are tightly coordinated homeostatic events and the paraventricular nucleus of the hypothalamus (PVH) represents a possible node of neural integration for signals related to energy and fluid homeostasis. We used TRAP2;Ai14 and Fos labeling to visualize neurons in the PVH and median preoptic nucleus (MEPO) responding to both water deprivation and hunger. Moreover, we determined that structural and functional development of dehydration-sensitive inputs to the PVH from the MEPO precedes those of agouti-related peptide (AgRP) neurons, which convey hunger signals and are known to be developmentally programmed by nutrition. We also determined that osmotic hyperstimulation of neonatal mice led to enhanced AgRP inputs to the PVH in adulthood, as well as disruptions to ingestive behaviors during high-fat diet feeding and dehydration-anorexia. Thus, development of feeding circuits is impacted not only by nutritional signals, but also by early perturbations to fluid homeostasis with context-specific consequences for coordination of ingestive behavior.
Collapse
|
13
|
Evans LC, Dailey-Krempel B, Lauar MR, Dayton A, Vulchanova L, Osborn JW. Renal interoception in health and disease. Auton Neurosci 2024; 255:103208. [PMID: 39128142 DOI: 10.1016/j.autneu.2024.103208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 08/13/2024]
Abstract
Catheter based renal denervation has recently been FDA approved for the treatment of hypertension. Traditionally, the anti-hypertensive effects of renal denervation have been attributed to the ablation of the efferent sympathetic renal nerves. In recent years the role of the afferent sensory renal nerves in the regulation of blood pressure has received increased attention. In addition, afferent renal denervation is associated with reductions in sympathetic nervous system activity. This suggests that reductions in sympathetic drive to organs other than the kidney may contribute to the non-renal beneficial effects observed in clinical trials of catheter based renal denervation. In this review we will provide an overview of the role of the afferent renal nerves in the regulation of renal function and the development of pathophysiologies, both renal and non-renal. We will also describe the central projections of the afferent renal nerves, to give context to the responses seen following their ablation and activation. Finally, we will discuss the emerging role of the kidney as an interoceptive organ. We will describe the potential role of the kidney in the regulation of interoceptive sensitivity and in this context, speculate on the possible pathological consequences of altered renal function.
Collapse
Affiliation(s)
- Louise C Evans
- Department of Surgery, University of Minnesota Medical School, Minneapolis 55455, United States of America
| | - Brianna Dailey-Krempel
- Department of Neuroscience, University of Minnesota, Minneapolis 55455, United States of America
| | - Mariana R Lauar
- Department of Surgery, University of Minnesota Medical School, Minneapolis 55455, United States of America
| | - Alex Dayton
- Division of Nephrology and Hypertension, University of Minnesota Medical School, Minneapolis 55455, United States of America
| | - Lucy Vulchanova
- Department of Neuroscience, University of Minnesota, Minneapolis 55455, United States of America
| | - John W Osborn
- Department of Surgery, University of Minnesota Medical School, Minneapolis 55455, United States of America.
| |
Collapse
|
14
|
Bonaz B, Sinniger V, Pellissier S. Role of stress and early-life stress in the pathogeny of inflammatory bowel disease. Front Neurosci 2024; 18:1458918. [PMID: 39319312 PMCID: PMC11420137 DOI: 10.3389/fnins.2024.1458918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/23/2024] [Indexed: 09/26/2024] Open
Abstract
Numerous preclinical and clinical studies have shown that stress is one of the main environmental factor playing a significant role in the pathogeny and life-course of bowel diseases. However, stressful events that occur early in life, even during the fetal life, leave different traces within the central nervous system, in area involved in stress response and autonomic network but also in emotion, cognition and memory regulation. Early-life stress can disrupt the prefrontal-amygdala circuit thus favoring an imbalance of the autonomic nervous system and the hypothalamic-pituitary adrenal axis, resulting in anxiety-like behaviors. The down regulation of vagus nerve and cholinergic anti-inflammatory pathway favors pro-inflammatory conditions. Recent data suggest that emotional abuse at early life are aggravating risk factors in inflammatory bowel disease. This review aims to unravel the mechanisms that explain the consequences of early life events and stress in the pathophysiology of inflammatory bowel disease and their mental co-morbidities. A review of therapeutic potential will also be covered.
Collapse
Affiliation(s)
- Bruno Bonaz
- Université Grenoble Alpes, Service d'Hépato-Gastroentérologie, Grenoble Institut Neurosciences, Grenoble, France
| | - Valérie Sinniger
- Université Grenoble Alpes, Service d'Hépato-Gastroentérologie, Grenoble Institut Neurosciences, Grenoble, France
| | - Sonia Pellissier
- Université Savoie Mont Blanc, Université Grenoble Alpes, LIP/PC2S, Chambéry, France
| |
Collapse
|
15
|
Biddinger JE, Elson AET, Fathi PA, Sweet SR, Nishimori K, Ayala JE, Simerly RB. AgRP neurons mediate activity-dependent development of oxytocin connectivity and autonomic regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.02.592838. [PMID: 38895484 PMCID: PMC11185571 DOI: 10.1101/2024.06.02.592838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
During postnatal life, the adipocyte-derived hormone leptin is required for proper targeting of neural inputs to the paraventricular nucleus of the hypothalamus (PVH) and impacts the activity of neurons containing agouti-related peptide (AgRP) in the arcuate nucleus of the hypothalamus. Activity-dependent developmental mechanisms are known to play a defining role during postnatal organization of neural circuits, but whether leptin-mediated postnatal neuronal activity specifies neural projections to the PVH or impacts downstream connectivity is largely unexplored. Here, we blocked neuronal activity of AgRP neurons during a discrete postnatal period and evaluated development of AgRP inputs to defined regions in the PVH, as well as descending projections from PVH oxytocin neurons to the dorsal vagal complex (DVC) and assessed their dependence on leptin or postnatal AgRP neuronal activity. In leptin-deficient mice, AgRP inputs to PVH neurons were significantly reduced, as well as oxytocin-specific neuronal targeting by AgRP. Moreover, downstream oxytocin projections from the PVH to the DVC were also impaired, despite the lack of leptin receptors found on PVH oxytocin neurons. Blocking AgRP neuron activity specifically during early postnatal life reduced the density of AgRP inputs to the PVH, as well as the density of projections from PVH oxytocin neurons to the DVC, and these innervation deficits were associated with dysregulated autonomic function. These findings suggest that postnatal targeting of descending PVH oxytocin projections to the DVC requires leptin-mediated AgRP neuronal activity, and represents a novel activity-dependent mechanism for hypothalamic specification of metabolic circuitry, with consequences for autonomic regulation. Significance statement Hypothalamic neural circuits maintain homeostasis by coordinating endocrine signals with autonomic responses and behavioral outputs to ensure that physiological responses remain in tune with environmental demands. The paraventricular nucleus of the hypothalamus (PVH) plays a central role in metabolic regulation, and the architecture of its neural inputs and axonal projections is a defining feature of how it receives and conveys neuroendocrine information. In adults, leptin regulates multiple aspects of metabolic physiology, but it also functions during development to direct formation of circuits controlling homeostatic functions. Here we demonstrate that leptin acts to specify the input-output architecture of PVH circuits through an activity-dependent, transsynaptic mechanism, which represents a novel means of sculpting neuroendocrine circuitry, with lasting effects on how the brain controls energy balance.
Collapse
|
16
|
Ivascu R, Torsin LI, Hostiuc L, Nitipir C, Corneci D, Dutu M. The Surgical Stress Response and Anesthesia: A Narrative Review. J Clin Med 2024; 13:3017. [PMID: 38792558 PMCID: PMC11121777 DOI: 10.3390/jcm13103017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/28/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
The human physiological response "to stress" includes all metabolic and hormonal changes produced by a traumatic event at the micro or macro cellular levels. The main goal of the body's first response to trauma is to keep physiological homeostasis. The perioperative non-specific adaptation response can sometimes be detrimental and can produce systemic inflammatory response syndrome (SIRS), characterized by hypermetabolism and hyper catabolism. We performed a narrative review consisting of a description of the surgical stress response's categories of changes (neurohormonal and immunological response) followed by reviewing methods found in published studies to modulate the surgical stress response perioperatively. We described various preoperative measures cited in the literature as lowering the burden of surgical trauma. This article revises the anesthetic drugs and techniques that have an impact on the surgical stress response and proven immune-modulatory effects. We also tried to name present knowledge gaps requiring future research. Our review concludes that proper preoperative measures, adequate general anesthetics, multimodal analgesia, early postoperative mobilization, and early enteral nutrition can decrease the stress response to surgery and ease patient recovery. Anesthetics and analgesics used during the perioperative period may modulate the innate and adaptive immune system and inflammatory system, with a consecutive impact on cancer recurrence and long-term outcomes.
Collapse
Affiliation(s)
- Robert Ivascu
- Department of Anesthesiology and Intensive Care, Carol Davila University of Medicine and Pharmacy, 0200021 Bucharest, Romania; (R.I.); (D.C.)
- Department of Anesthesiology and Intensive Care, Dr. Carol Davila Central Military Emergency University Hospital, 010242 Bucharest, Romania; (L.I.T.); (L.H.)
| | - Ligia I. Torsin
- Department of Anesthesiology and Intensive Care, Dr. Carol Davila Central Military Emergency University Hospital, 010242 Bucharest, Romania; (L.I.T.); (L.H.)
| | - Laura Hostiuc
- Department of Anesthesiology and Intensive Care, Dr. Carol Davila Central Military Emergency University Hospital, 010242 Bucharest, Romania; (L.I.T.); (L.H.)
| | - Cornelia Nitipir
- Department of Oncology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Dan Corneci
- Department of Anesthesiology and Intensive Care, Carol Davila University of Medicine and Pharmacy, 0200021 Bucharest, Romania; (R.I.); (D.C.)
- Department of Anesthesiology and Intensive Care, Dr. Carol Davila Central Military Emergency University Hospital, 010242 Bucharest, Romania; (L.I.T.); (L.H.)
| | - Madalina Dutu
- Department of Anesthesiology and Intensive Care, Carol Davila University of Medicine and Pharmacy, 0200021 Bucharest, Romania; (R.I.); (D.C.)
- Department of Anesthesiology and Intensive Care, Dr. Carol Davila Central Military Emergency University Hospital, 010242 Bucharest, Romania; (L.I.T.); (L.H.)
| |
Collapse
|
17
|
Li H, Jiang T, An S, Xu M, Gou L, Ren B, Shi X, Wang X, Yan J, Yuan J, Xu X, Wu QF, Luo Q, Gong H, Bian WJ, Li A, Yu X. Single-neuron projectomes of mouse paraventricular hypothalamic nucleus oxytocin neurons reveal mutually exclusive projection patterns. Neuron 2024; 112:1081-1099.e7. [PMID: 38290516 DOI: 10.1016/j.neuron.2023.12.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 11/07/2023] [Accepted: 12/29/2023] [Indexed: 02/01/2024]
Abstract
Oxytocin (OXT) plays important roles in autonomic control and behavioral modulation. However, it is unknown how the projection patterns of OXT neurons align with underlying physiological functions. Here, we present the reconstructed single-neuron, whole-brain projectomes of 264 OXT neurons of the mouse paraventricular hypothalamic nucleus (PVH) at submicron resolution. These neurons hierarchically clustered into two groups, with distinct morphological and transcriptional characteristics and mutually exclusive projection patterns. Cluster 1 (177 neurons) axons terminated exclusively in the median eminence (ME) and have few collaterals terminating within hypothalamic regions. By contrast, cluster 2 (87 neurons) sent wide-spread axons to multiple brain regions, but excluding ME. Dendritic arbors of OXT neurons also extended outside of the PVH, suggesting capability to sense signals and modulate target regions. These single-neuron resolution observations reveal distinct OXT subpopulations, provide comprehensive analysis of their morphology, and lay the structural foundation for better understanding the functional heterogeneity of OXT neurons.
Collapse
Affiliation(s)
- Humingzhu Li
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and Peking University McGovern Institute, Peking University, Beijing 100871, China
| | - Tao Jiang
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
| | - Sile An
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Mingrui Xu
- University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lingfeng Gou
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Biyu Ren
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoxue Shi
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaofei Wang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jun Yan
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Yuan
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China; Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaohong Xu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing-Feng Wu
- University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qingming Luo
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China; Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hui Gong
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China; Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wen-Jie Bian
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China; School of Life Sciences, Westlake University, Hangzhou 310024, China.
| | - Anan Li
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China; Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Xiang Yu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and Peking University McGovern Institute, Peking University, Beijing 100871, China; Chinese Institute for Brain Research, Beijing 102206, China.
| |
Collapse
|
18
|
Hendry E, McCallister B, Elman DJ, Freeman R, Borsook D, Elman I. Validity of mental and physical stress models. Neurosci Biobehav Rev 2024; 158:105566. [PMID: 38307304 PMCID: PMC11082879 DOI: 10.1016/j.neubiorev.2024.105566] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/13/2024] [Accepted: 01/27/2024] [Indexed: 02/04/2024]
Abstract
Different stress models are employed to enhance our understanding of the underlying mechanisms and explore potential interventions. However, the utility of these models remains a critical concern, as their validities may be limited by the complexity of stress processes. Literature review revealed that both mental and physical stress models possess reasonable construct and criterion validities, respectively reflected in psychometrically assessed stress ratings and in activation of the sympathoadrenal system and the hypothalamic-pituitary-adrenal axis. The findings are less robust, though, in the pharmacological perturbations' domain, including such agents as adenosine or dobutamine. Likewise, stress models' convergent- and discriminant validity vary depending on the stressors' nature. Stress models share similarities, but also have important differences regarding their validities. Specific traits defined by the nature of the stressor stimulus should be taken into consideration when selecting stress models. Doing so can personalize prevention and treatment of stress-related antecedents, its acute processing, and chronic sequelae. Further work is warranted to refine stress models' validity and customize them so they commensurate diverse populations and circumstances.
Collapse
Affiliation(s)
- Erin Hendry
- Center for Autonomic and Peripheral Nerve Disorders, Harvard Medical School, Boston, MA, USA; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Brady McCallister
- Center for Autonomic and Peripheral Nerve Disorders, Harvard Medical School, Boston, MA, USA
| | - Dan J Elman
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Roy Freeman
- Center for Autonomic and Peripheral Nerve Disorders, Harvard Medical School, Boston, MA, USA; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - David Borsook
- Departments of Psychiatry and Radiology, Massachusetts General Hospital, Harvard Medical School, Department of Anesthesiology, Harvard Medical School, Boston, MA, USA.
| | - Igor Elman
- Department of Psychiatry, Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA
| |
Collapse
|
19
|
Haaf R, Brandi ML, Albantakis L, Lahnakoski JM, Henco L, Schilbach L. Peripheral oxytocin levels are linked to hypothalamic gray matter volume in autistic adults: a cross-sectional secondary data analysis. Sci Rep 2024; 14:1380. [PMID: 38228703 PMCID: PMC10791615 DOI: 10.1038/s41598-023-50770-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 12/25/2023] [Indexed: 01/18/2024] Open
Abstract
Oxytocin (OXT) is known to modulate social behavior and cognition and has been discussed as pathophysiological and therapeutic factor for autism spectrum disorder (ASD). An accumulating body of evidence indicates the hypothalamus to be of particular importance with regard to the underlying neurobiology. Here we used a region of interest voxel-based morphometry (VBM) approach to investigate hypothalamic gray matter volume (GMV) in autistic (n = 29, age 36.03 ± 11.0) and non-autistic adults (n = 27, age 30.96 ± 11.2). Peripheral plasma OXT levels and the autism spectrum quotient (AQ) were used for correlation analyses. Results showed no differences in hypothalamic GMV in autistic compared to non-autistic adults but suggested a differential association between hypothalamic GMV and OXT levels, such that a positive association was found for the ASD group. In addition, hypothalamic GMV showed a positive association with autistic traits in the ASD group. Bearing in mind the limitations such as a relatively small sample size, a wide age range and a high rate of psychopharmacological treatment in the ASD sample, these results provide new preliminary evidence for a potentially important role of the HTH in ASD and its relationship to the OXT system, but also point towards the importance of interindividual differences.
Collapse
Affiliation(s)
- Raoul Haaf
- Independent Max Planck Research Group for Social Neuroscience, Max Planck Institute of Psychiatry, Munich, Germany.
- Graduate School, Technical University of Munich, Munich, Germany.
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany.
| | - Marie-Luise Brandi
- Independent Max Planck Research Group for Social Neuroscience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Laura Albantakis
- Independent Max Planck Research Group for Social Neuroscience, Max Planck Institute of Psychiatry, Munich, Germany
- Outpatient and Day Clinic for Disorders of Social Interaction, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Juha M Lahnakoski
- Independent Max Planck Research Group for Social Neuroscience, Max Planck Institute of Psychiatry, Munich, Germany
- Institute of Neurosciences and Medicine, Brain and Behaviour (INM-7), Research Center Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Lara Henco
- Independent Max Planck Research Group for Social Neuroscience, Max Planck Institute of Psychiatry, Munich, Germany
- Graduate School of Systemic Neurosciences, Munich, Germany
| | - Leonhard Schilbach
- Independent Max Planck Research Group for Social Neuroscience, Max Planck Institute of Psychiatry, Munich, Germany
- Outpatient and Day Clinic for Disorders of Social Interaction, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Munich, Germany
- Graduate School of Systemic Neurosciences, Munich, Germany
- Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
20
|
Zahner MR, Brown MC, Chandley MJ. Inactivation of the paraventricular nucleus attenuates the cardiogenic sympathetic afferent reflex in the spontaneously hypertensive rat. J Hypertens 2024; 42:70-78. [PMID: 37889604 PMCID: PMC10792548 DOI: 10.1097/hjh.0000000000003542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
BACKGROUND Myocardial ischemia causes the release of bradykinin, which stimulates cardiac afferents, causing sympathetic excitation and chest pain. Glutamatergic activation of the paraventricular hypothalamic nucleus (PVN) in the spontaneously hypertensive rat (SHR) drives elevated basal sympathetic activity. Thus, we tested the hypothesis that inactivation of the PVN attenuates the elevated reflex response to epicardial bradykinin in the SHR and that ionotropic PVN glutamate receptors mediate the elevated reflex. METHODS We recorded the arterial pressure and renal sympathetic nerve activity (RSNA) response to epicardial bradykinin application in anesthetized SHR and Wistar Kyoto (WKY) rats before and after PVN microinjection of GABA A agonist muscimol or ionotropic glutamate receptor antagonist kynurenic acid. RESULTS Muscimol significantly decreased the arterial pressure response to bradykinin from 180.4 ± 5.8 to 119.5 ± 6.9 mmHg in the SHR and from 111.8 ± 7.0 to 84.2 ± 8.3 mmHg in the WKY and the RSNA response from 186.2 ± 7.1 to 142.7 ± 7.3% of baseline in the SHR and from 201.0 ± 11.5 to 160.2 ± 9.3% of baseline in the WKY. Kynurenic acid significantly decreased the arterial pressure response in the SHR from 164.5 ± 5.0 to 126.2 ± 7.7 mmHg and the RSNA response from 189.9 ± 13.7to 168.5 ± 12.7% of baseline but had no effect in the WKY. CONCLUSION These results suggest that tonic PVN activity is critical for the full manifestation of the CSAR in both the WKY and SHR. Glutamatergic PVN activity contributes to the augmented CSAR observed in the SHR.
Collapse
Affiliation(s)
- Matthew R Zahner
- Department of Health Sciences, East Tennessee State University College of Public Health
| | - Mary C Brown
- Department of Health Sciences, East Tennessee State University College of Public Health
| | - Michelle J Chandley
- Department of Biomedical Science, East Tennessee State University College of Medicine, Johnson City, Tennessee, USA
| |
Collapse
|
21
|
Tsai SF, Kuo YM. The Role of Central Oxytocin in Autonomic Regulation. CHINESE J PHYSIOL 2024; 67:3-14. [PMID: 38780268 DOI: 10.4103/ejpi.ejpi-d-23-00037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/10/2023] [Indexed: 05/25/2024] Open
Abstract
Oxytocin (OXT), a neuropeptide originating from the hypothalamus and traditionally associated with peripheral functions in parturition and lactation, has emerged as a pivotal player in the central regulation of the autonomic nervous system (ANS). This comprehensive ANS, comprising sympathetic, parasympathetic, and enteric components, intricately combines sympathetic and parasympathetic influences to provide unified control. The central oversight of sympathetic and parasympathetic outputs involves a network of interconnected regions spanning the neuroaxis, playing a pivotal role in the real-time regulation of visceral function, homeostasis, and adaptation to challenges. This review unveils the significant involvement of the central OXT system in modulating autonomic functions, shedding light on diverse subpopulations of OXT neurons within the paraventricular nucleus of the hypothalamus and their intricate projections. The narrative progresses from the basics of central ANS regulation to a detailed discussion of the central controls of sympathetic and parasympathetic outflows. The subsequent segment focuses specifically on the central OXT system, providing a foundation for exploring the central role of OXT in ANS regulation. This review synthesizes current knowledge, paving the way for future research endeavors to unravel the full scope of autonomic control and understand multifaceted impact of OXT on physiological outcomes.
Collapse
Affiliation(s)
- Sheng-Feng Tsai
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Min Kuo
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
22
|
Pace SA, Myers B. Hindbrain Adrenergic/Noradrenergic Control of Integrated Endocrine and Autonomic Stress Responses. Endocrinology 2023; 165:bqad178. [PMID: 38015813 DOI: 10.1210/endocr/bqad178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/07/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Hindbrain adrenergic/noradrenergic nuclei facilitate endocrine and autonomic responses to physical and psychological challenges. Neurons that synthesize adrenaline and noradrenaline target hypothalamic structures to modulate endocrine responses while descending spinal projections regulate sympathetic function. Furthermore, these neurons respond to diverse stress-related metabolic, autonomic, and psychosocial challenges. Accordingly, adrenergic and noradrenergic nuclei are integrative hubs that promote physiological adaptation to maintain homeostasis. However, the precise mechanisms through which adrenaline- and noradrenaline-synthesizing neurons sense interoceptive and exteroceptive cues to coordinate physiological responses have yet to be fully elucidated. Additionally, the regulatory role of these cells in the context of chronic stress has received limited attention. This mini-review consolidates reports from preclinical rodent studies on the organization and function of brainstem adrenaline and noradrenaline cells to provide a framework for how these nuclei coordinate endocrine and autonomic physiology. This includes identification of hindbrain adrenaline- and noradrenaline-producing cell groups and their role in stress responding through neurosecretory and autonomic engagement. Although temporally and mechanistically distinct, the endocrine and autonomic stress axes are complementary and interconnected. Therefore, the interplay between brainstem adrenergic/noradrenergic nuclei and peripheral physiological systems is necessary for integrated stress responses and organismal survival.
Collapse
Affiliation(s)
- Sebastian A Pace
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Brent Myers
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
23
|
Song JW, Lee KH, Seong H, Shin DM, Shon WJ. Taste receptor type 1 member 3 enables western diet-induced anxiety in mice. BMC Biol 2023; 21:243. [PMID: 37926812 PMCID: PMC10626698 DOI: 10.1186/s12915-023-01723-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 10/03/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Accumulating evidence supports that the Western diet (WD), a diet high in saturated fat and sugary drinks, contributes to the pathogenesis of anxiety disorders, which are the most prevalent mental disorders worldwide. However, the underlying mechanisms by which WD causes anxiety remain unclear. Abundant expression of taste receptor type 1 member 3 (TAS1R3) has been identified in the hypothalamus, a key brain area involved in sensing peripheral nutritional signals and regulating anxiety. Thus, we investigated the influence of excessive WD intake on anxiety and mechanisms by which WD intake affects anxiety development using wild-type (WT) and Tas1r3 deficient (Tas1r3-/-) mice fed a normal diet (ND) or WD for 12 weeks. RESULTS WD increased anxiety in male WT mice, whereas male Tas1r3-/- mice were protected from WD-induced anxiety, as assessed by open field (OF), elevated plus maze (EPM), light-dark box (LDB), and novelty-suppressed feeding (NSF) tests. Analyzing the hypothalamic transcriptome of WD-fed WT and Tas1r3-/- mice, we found 1,432 genes significantly up- or down-regulated as a result of Tas1r3 deficiency. Furthermore, bioinformatic analysis revealed that the CREB/BDNF signaling-mediated maintenance of neuronal regeneration, which can prevent anxiety development, was enhanced in WD-fed Tas1r3-/- mice compared with WD-fed WT mice. Additionally, in vitro studies further confirmed that Tas1r3 knockdown prevents the suppression of Creb1 and of CREB-mediated BDNF expression caused by high levels of glucose, fructose, and palmitic acid in hypothalamic neuronal cells. CONCLUSIONS Our results imply that TAS1R3 may play a key role in WD-induced alterations in hypothalamic functions, and that inhibition of TAS1R3 overactivation in the hypothalamus could offer therapeutic targets to alleviate the effects of WD on anxiety.
Collapse
Affiliation(s)
- Jae Won Song
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Keon-Hee Lee
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Hobin Seong
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Dong-Mi Shin
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea.
- Research Institute of Human Ecology, Seoul National University, Gwanak-Gu, Seoul, 08826, Republic of Korea.
| | - Woo-Jeong Shon
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea.
- Research Institute of Human Ecology, Seoul National University, Gwanak-Gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
24
|
Tseng YT, Schaefke B, Wei P, Wang L. Defensive responses: behaviour, the brain and the body. Nat Rev Neurosci 2023; 24:655-671. [PMID: 37730910 DOI: 10.1038/s41583-023-00736-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2023] [Indexed: 09/22/2023]
Abstract
Most animals live under constant threat from predators, and predation has been a major selective force in shaping animal behaviour. Nevertheless, defence responses against predatory threats need to be balanced against other adaptive behaviours such as foraging, mating and recovering from infection. This behavioural balance in ethologically relevant contexts requires adequate integration of internal and external signals in a complex interplay between the brain and the body. Despite this complexity, research has often considered defensive behaviour as entirely mediated by the brain processing threat-related information obtained via perception of the external environment. However, accumulating evidence suggests that the endocrine, immune, gastrointestinal and reproductive systems have important roles in modulating behavioural responses to threat. In this Review, we focus on how predatory threat defence responses are shaped by threat imminence and review the circuitry between subcortical brain regions involved in mediating defensive behaviours. Then, we discuss the intersection of peripheral systems involved in internal states related to infection, hunger and mating with the neurocircuits that underlie defence responses against predatory threat. Through this process, we aim to elucidate the interconnections between the brain and body as an integrated network that facilitates appropriate defensive responses to threat and to discuss the implications for future behavioural research.
Collapse
Affiliation(s)
- Yu-Ting Tseng
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behaviour, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bernhard Schaefke
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Pengfei Wei
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Liping Wang
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- Guangdong Provincial Key Laboratory of Brain Connectome and Behaviour, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
25
|
Gruber T, Lechner F, Murat C, Contreras RE, Sanchez-Quant E, Miok V, Makris K, Le Thuc O, González-García I, García-Clave E, Althammer F, Krabichler Q, DeCamp LM, Jones RG, Lutter D, Williams RH, Pfluger PT, Müller TD, Woods SC, Pospisilik JA, Martinez-Jimenez CP, Tschöp MH, Grinevich V, García-Cáceres C. High-calorie diets uncouple hypothalamic oxytocin neurons from a gut-to-brain satiation pathway via κ-opioid signaling. Cell Rep 2023; 42:113305. [PMID: 37864798 PMCID: PMC10636643 DOI: 10.1016/j.celrep.2023.113305] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/21/2023] [Accepted: 10/04/2023] [Indexed: 10/23/2023] Open
Abstract
Oxytocin-expressing paraventricular hypothalamic neurons (PVNOT neurons) integrate afferent signals from the gut, including cholecystokinin (CCK), to adjust whole-body energy homeostasis. However, the molecular underpinnings by which PVNOT neurons orchestrate gut-to-brain feeding control remain unclear. Here, we show that mice undergoing selective ablation of PVNOT neurons fail to reduce food intake in response to CCK and develop hyperphagic obesity on a chow diet. Notably, exposing wild-type mice to a high-fat/high-sugar (HFHS) diet recapitulates this insensitivity toward CCK, which is linked to diet-induced transcriptional and electrophysiological aberrations specifically in PVNOT neurons. Restoring OT pathways in diet-induced obese (DIO) mice via chemogenetics or polypharmacology sufficiently re-establishes CCK's anorexigenic effects. Last, by single-cell profiling, we identify a specialized PVNOT neuronal subpopulation with increased κ-opioid signaling under an HFHS diet, which restrains their CCK-evoked activation. In sum, we document a (patho)mechanism by which PVNOT signaling uncouples a gut-brain satiation pathway under obesogenic conditions.
Collapse
Affiliation(s)
- Tim Gruber
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA; Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49506, USA.
| | - Franziska Lechner
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Cahuê Murat
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Raian E Contreras
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Eva Sanchez-Quant
- Helmholtz Pioneer Campus (HPC), Helmholtz Zentrum München, Neuherberg, Germany
| | - Viktorian Miok
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Konstantinos Makris
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Ophélia Le Thuc
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Ismael González-García
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Elena García-Clave
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | | | - Quirin Krabichler
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Lisa M DeCamp
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA
| | - Dominik Lutter
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Rhiannan H Williams
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute for Neurogenomics, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Paul T Pfluger
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Research Unit NeuroBiology of Diabetes, Helmholtz Zentrum München, 85764 Neuherberg, Germany; Neurobiology of Diabetes, TUM School of Medicine, Technical University Munich, 80333 Munich, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Department of Pharmacology and Experimental Therapy, Institute for Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls Hospitals and Clinics, Tübingen, Germany
| | - Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - John Andrew Pospisilik
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA; Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49506, USA
| | - Celia P Martinez-Jimenez
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49506, USA; TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität, Munich, Germany
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA.
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| |
Collapse
|
26
|
Wang J, Zhang Q, Yao L, He T, Chen X, Su Y, Sun S, Fan M, Yan J, Wang T, Zhang M, Guo F, Mo S, Lu M, Zou M, Li L, Yuan Q, Pan H, Chen Y. Modulating activity of PVN neurons prevents atrial fibrillation induced circulation dysfunction by electroacupuncture at BL15. Chin Med 2023; 18:135. [PMID: 37848944 PMCID: PMC10580609 DOI: 10.1186/s13020-023-00841-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Circulation dysfunction is a major contributing factor to thrombosis in patients with atrial fibrillation (AF) for which effective interventions are lacking. Growing evidence indicates that regulating the paraventricular nucleus (PVN), an autonomic control center, could offer a novel strategy for treating cardiovascular and circulatory diseases. Concurrently, electroacupuncture (EA) at Xinshu (BL15), a form of peripheral nerve stimulation, has shown efficacy in treating several cardiovascular conditions, although its specific mechanism remains unclear. This study aimed to assess the impact of EA at BL15 on circulatory dysfunction in a rat AF model and investigate the pivotal role of PVN neuronal activity. METHODS To mimic the onset of AF, male SD rats received tail intravenous injection of ACh-CaCl2 and were then subjected to EA at BL15, sham EA, or EA at Shenshu (BL23). Macro- and micro-circulation function were evaluated using in vivo ultrasound imaging and laser doppler testing, respectively. Vasomotricity was assessed by measuring dimension changes during vascular relaxation and contraction. Vascular endothelial function was measured using myograph, and the activation of the autonomic nerve system was evaluated through nerve activity signals. Additionally, chemogenetic manipulation was used to block PVN neuronal activation to further elucidate the role of PVN activation in the prevention of AF-induced blood circulation dysfunction through EA treatment. RESULTS Our data demonstrate that EA at BL15, but not BL23 or sham EA, effectively prevented AF-induced macro- and micro-circulation dysfunction. Furthermore, EA at BL15 restored AF-induced vasomotricity impairment. Additionally, EA treatment prevented abnormal activation of the autonomic nerve system induced by AF, although it did not address vascular endothelial dysfunction. Importantly, excessive activation of PVN neurons negated the protective effects of EA treatment on AF-induced circulation dysfunction in rats. CONCLUSION These results indicate that EA treatment at BL15 modulates PVN neuronal activity and provides protection against AF-induced circulatory dysfunction.
Collapse
Affiliation(s)
- Jingya Wang
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Qiumei Zhang
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
- Institute of Physical and Health, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
- Guangdong Chaozhou Health Vocational College, Chaozhou, 521000, People's Republic of China
| | - Lin Yao
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Teng He
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Xinyi Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Yang Su
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Shengxuan Sun
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Mengyue Fan
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Jinglan Yan
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Taiyi Wang
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Meng Zhang
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Feng Guo
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Shiqing Mo
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Manqi Lu
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Meixia Zou
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Liangjie Li
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Qing Yuan
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Huashan Pan
- Institute of Physical and Health, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China.
- Guangdong Chaozhou Health Vocational College, Chaozhou, 521000, People's Republic of China.
| | - Yongjun Chen
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
27
|
Althammer F. Heralding a new era of oxytocinergic research: New tools, new problems? J Neuroendocrinol 2023; 35:e13333. [PMID: 37621199 DOI: 10.1111/jne.13333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023]
Abstract
According to classic neuroendocrinology, hypothalamic oxytocin cells can be categorized into parvo- and magnocellular neurons. However, research in the last decade provided ample evidence that this black-and-white model of oxytocin neurons is most likely oversimplified. Novel genetic, functional and morphological studies indicate that oxytocin neurons might be organized in functional modules and suggest the existence of five or more distinct oxytocinergic subpopulations. However, many of these novel, automated high-throughput techniques might be inherently biased and interpretation of acquired data needs to be approached with caution to enable drawing sound and reliable conclusions. In addition, the recent finding that astrocytes in various brain regions express functional oxytocin receptors represents a paradigm shift and challenges the view that oxytocin primarily acts as a direct peptidergic neurotransmitter. This review highlights the latest technical advances in oxytocinergic research, puts recent studies on the oxytocin system into context and formulates various provocative ideas based on novel findings that challenges various prevailing hypotheses and dogmas about oxytocinergic modulation.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
28
|
Fortin SM, Chen JC, Petticord MC, Ragozzino FJ, Peters JH, Hayes MR. The locus coeruleus contributes to the anorectic, nausea, and autonomic physiological effects of glucagon-like peptide-1. SCIENCE ADVANCES 2023; 9:eadh0980. [PMID: 37729419 PMCID: PMC10511187 DOI: 10.1126/sciadv.adh0980] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023]
Abstract
Increasing the therapeutic potential and reducing the side effects of U.S. Food and Drug Administration-approved glucagon-like peptide-1 receptor (GLP-1R) agonists used to treat obesity require complete characterization of the central mechanisms that mediate both the food intake-suppressive and illness-like effects of GLP-1R signaling. Our studies, in the rat, demonstrate that GLP-1Rs in the locus coeruleus (LC) are pharmacologically and physiologically relevant for food intake control. Furthermore, agonism of LC GLP-1Rs induces illness-like behaviors, and antagonism of LC GLP-1Rs can attenuate GLP-1R-mediated nausea. Electrophysiological and behavioral pharmacology data support a role for LC GLP-1Rs expressed on presynaptic glutamatergic terminals in the control of feeding and malaise. Collectively, our work establishes the LC as a site of action for GLP-1 signaling and extends our understanding of the GLP-1 signaling mechanism necessary for the development of improved obesity pharmacotherapies.
Collapse
Affiliation(s)
- Samantha M. Fortin
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jack C. Chen
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marisa C. Petticord
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Forrest J. Ragozzino
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - James H. Peters
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Matthew R. Hayes
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
29
|
Makrygianni EA, Chrousos GP. Neural Progenitor Cells and the Hypothalamus. Cells 2023; 12:1822. [PMID: 37508487 PMCID: PMC10378393 DOI: 10.3390/cells12141822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 07/30/2023] Open
Abstract
Neural progenitor cells (NPCs) are multipotent neural stem cells (NSCs) capable of self-renewing and differentiating into neurons, astrocytes and oligodendrocytes. In the postnatal/adult brain, NPCs are primarily located in the subventricular zone (SVZ) of the lateral ventricles (LVs) and subgranular zone (SGZ) of the hippocampal dentate gyrus (DG). There is evidence that NPCs are also present in the postnatal/adult hypothalamus, a highly conserved brain region involved in the regulation of core homeostatic processes, such as feeding, metabolism, reproduction, neuroendocrine integration and autonomic output. In the rodent postnatal/adult hypothalamus, NPCs mainly comprise different subtypes of tanycytes lining the wall of the 3rd ventricle. In the postnatal/adult human hypothalamus, the neurogenic niche is constituted by tanycytes at the floor of the 3rd ventricle, ependymal cells and ribbon cells (showing a gap-and-ribbon organization similar to that in the SVZ), as well as suprachiasmatic cells. We speculate that in the postnatal/adult human hypothalamus, neurogenesis occurs in a highly complex, exquisitely sophisticated neurogenic niche consisting of at least four subniches; this structure has a key role in the regulation of extrahypothalamic neurogenesis, and hypothalamic and extrahypothalamic neural circuits, partly through the release of neurotransmitters, neuropeptides, extracellular vesicles (EVs) and non-coding RNAs (ncRNAs).
Collapse
Affiliation(s)
- Evanthia A Makrygianni
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
30
|
Woodson J, Bergan JF. Uncovering the brain-wide pattern of synaptic input to vasopressin-expressing neurons in the paraventricular nucleus of the hypothalamus. J Comp Neurol 2023; 531:1017-1031. [PMID: 37121600 PMCID: PMC10566340 DOI: 10.1002/cne.25476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 05/02/2023]
Abstract
Arginine vasopressin (AVP) is a neuropeptide critical for the mammalian stress response and social behavior. AVP produced in the hypothalamus regulates water osmolality and vasoconstriction in the body, and in the brain, it regulates social behavior, aggression, and anxiety. However, the circuit mechanisms that link AVP to social behavior, homeostatic function, and disease are not well understood. This study investigates the circuit configurations of AVP-expressing neurons in the rodent hypothalamus and characterizes synaptic input from the entire brain. We targeted the paraventricular nucleus (PVN) using retrograde viral tracing techniques to identify direct afferent synaptic connections made onto AVP-expressing neurons. AVP neurons in the PVN display region-specific anatomical configurations that reflect their unique contributions to homeostatic function, motor behaviors, feeding, and affiliative behavior. The afferent connections identified were similar in both sexes and subsequent molecular investigation of these inputs shows that those local hypothalamic inputs are overwhelmingly nonpeptidergic cells indicating a potential interneuron nexus between hormone cell activation and broader cortical connection. This proposed work reveals new insights into the organization of social behavior circuits in the brain, and how neuropeptides act centrally to modulate social behaviors.
Collapse
Affiliation(s)
- Jonathan Woodson
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Joseph F Bergan
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, Massachusetts, USA
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| |
Collapse
|
31
|
Zheng H, Patel TA, Liu X, Patel KP. C-type natriuretic peptide (CNP) in the paraventricular nucleus-mediated renal sympatho-inhibition. Front Physiol 2023; 14:1162699. [PMID: 37082246 PMCID: PMC10110992 DOI: 10.3389/fphys.2023.1162699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
Volume reflex produces sympatho-inhibition that is mediated by the hypothalamic paraventricular nucleus (PVN). However, the mechanisms for the sympatho-inhibitory role of the PVN and the neurochemical factors involved remain to be identified. In this study, we proposed C-type natriuretic peptide (CNP) as a potential mediator of this sympatho-inhibition within the PVN. Microinjection of CNP (1.0 μg) into the PVN significantly decreased renal sympathetic nerve activity (RSNA) (-25.8% ± 1.8% vs. -3.6% ± 1.5%), mean arterial pressure (-15.0 ± 1.9 vs. -0.1 ± 0.9 mmHg) and heart rate (-23.6 ± 3.5 vs. -0.3 ± 0.9 beats/min) compared with microinjection of vehicle. Picoinjection of CNP significantly decreased the basal discharge of extracellular single-unit recordings in 5/6 (83%) rostral ventrolateral medulla (RVLM)-projecting PVN neurons and in 6/13 (46%) of the neurons that were not antidromically activated from the RVLM. We also observed that natriuretic peptide receptor type C (NPR-C) was present on the RVLM projecting PVN neurons detected by dual-labeling with retrograde tracer. Prior NPR-C siRNA microinjection into the PVN significantly blunted the decrease in RSNA to CNP microinjections into the PVN. Volume expansion-mediated reduction in RSNA was significantly blunted by prior administration of NPR-C siRNA into the PVN. These results suggest a potential role for CNP within the PVN in regulating RSNA, specifically under physiological conditions of alterations in fluid balance.
Collapse
Affiliation(s)
- Hong Zheng
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD, United States
| | - Tapan A. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Xuefei Liu
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD, United States
| | - Kaushik P. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
32
|
Rasiah NP, Loewen SP, Bains JS. Windows into stress: a glimpse at emerging roles for CRH PVN neurons. Physiol Rev 2023; 103:1667-1691. [PMID: 36395349 DOI: 10.1152/physrev.00056.2021] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The corticotropin-releasing hormone cells in the paraventricular nucleus of the hypothalamus (CRHPVN) control the slow endocrine response to stress. The synapses on these cells are exquisitely sensitive to acute stress, leveraging local signals to leave a lasting imprint on this system. Additionally, recent work indicates that these cells also play key roles in the control of distinct stress and survival behaviors. Here we review these observations and provide a perspective on the role of CRHPVN neurons as integrative and malleable hubs for behavioral, physiological, and endocrine responses to stress.
Collapse
Affiliation(s)
- Neilen P Rasiah
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Spencer P Loewen
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jaideep S Bains
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
33
|
Acharya KD, Graham M, Raman H, Parakoyi AER, Corcoran A, Belete M, Ramaswamy B, Koul S, Sachar I, Derendorf K, Wilmer JB, Gottipati S, Tetel MJ. Estradiol-mediated protection against high-fat diet induced anxiety and obesity is associated with changes in the gut microbiota in female mice. Sci Rep 2023; 13:4776. [PMID: 36959275 PMCID: PMC10036463 DOI: 10.1038/s41598-023-31783-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/17/2023] [Indexed: 03/25/2023] Open
Abstract
Decreased estrogens during menopause are associated with increased risk of anxiety, depression, type 2 diabetes and obesity. Similarly, depleting estrogens in rodents by ovariectomy, combined with a high-fat diet (HFD), increases anxiety and adiposity. How estrogens and diet interact to affect anxiety and metabolism is poorly understood. Mounting evidence indicates that gut microbiota influence anxiety and metabolism. Here, we investigated the effects of estradiol (E) and HFD on anxiety, metabolism, and their correlation with changes in gut microbiota in female mice. Adult C57BL/6J mice were ovariectomized, implanted with E or vehicle-containing capsules and fed a standard diet or HFD. Anxiety-like behavior was assessed and neuronal activation was measured by c-fos immunoreactivity throughout the brain using iDISCO. HFD increased anxiety-like behavior, while E reduced this HFD-dependent anxiogenic effect. Interestingly, E decreased neuronal activation in brain regions involved in anxiety and metabolism. E treatment also altered gut microbes, a subset of which were associated with anxiety-like behavior. These findings provide insight into gut microbiota-based therapies for anxiety and metabolic disorders associated with declining estrogens in menopausal women.
Collapse
Affiliation(s)
- Kalpana D Acharya
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | - Madeline Graham
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | - Harshini Raman
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | | | - Alexis Corcoran
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | - Merzu Belete
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | - Bharath Ramaswamy
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | - Shashikant Koul
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | | | - Kevin Derendorf
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | - Jeremy B Wilmer
- Department of Psychology, Wellesley College, Wellesley, MA, 02481, USA
| | - Srikanth Gottipati
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | - Marc J Tetel
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA.
| |
Collapse
|
34
|
Chen RB, Wang QY, Wang YY, Wang YD, Liu JH, Liao ZZ, Xiao XH. Feeding-induced hepatokines and crosstalk with multi-organ: A novel therapeutic target for Type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1094458. [PMID: 36936164 PMCID: PMC10020511 DOI: 10.3389/fendo.2023.1094458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/15/2023] [Indexed: 03/06/2023] Open
Abstract
Hyperglycemia, which can be caused by either an insulin deficit and/or insulin resistance, is the main symptom of Type 2 diabetes, a significant endocrine metabolic illness. Conventional medications, including insulin and oral antidiabetic medicines, can alleviate the signs of diabetes but cannot restore insulin release in a physiologically normal amount. The liver detects and reacts to shifts in the nutritional condition that occur under a wide variety of metabolic situations, making it an essential organ for maintaining energy homeostasis. It also performs a crucial function in glucolipid metabolism through the secretion of hepatokines. Emerging research shows that feeding induces hepatokines release, which regulates glucose and lipid metabolism. Notably, these feeding-induced hepatokines act on multiple organs to regulate glucolipotoxicity and thus influence the development of T2DM. In this review, we focus on describing how feeding-induced cross-talk between hepatokines, including Adropin, Manf, Leap2 and Pcsk9, and metabolic organs (e.g.brain, heart, pancreas, and adipose tissue) affects metabolic disorders, thus revealing a novel approach for both controlling and managing of Type 2 diabetes as a promising medication.
Collapse
Affiliation(s)
- Rong-Bin Chen
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qi-Yu Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuan-Yuan Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ya-Di Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiang-Hua Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhe-Zhen Liao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin-Hua Xiao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
35
|
Jiang Y, Rezai-Zadeh K, Desmoulins LD, Muenzberg H, Derbenev AV, Zsombok A. GABAergic leptin receptor-expressing neurons in the dorsomedial hypothalamus project to brown adipose tissue-related neurons in the paraventricular nucleus of mice. Auton Neurosci 2023; 245:103058. [PMID: 36538864 PMCID: PMC9899324 DOI: 10.1016/j.autneu.2022.103058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/14/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Brown adipose tissue (BAT) contributes to energy homeostasis via nonshivering thermogenesis. The BAT is densely innervated by the sympathetic nervous system (SNS) and activity of pre-autonomic neurons modulates the sympathetic outflow. Leptin, an adipocyte hormone, alters energy homeostasis and thermogenesis of BAT via several neuronal circuits; however, the cellular effects of leptin on interscapular BAT (iBAT)-related neurons in the hypothalamus remain to be determined. In this study, we used pseudorabies virus (PRV) to identify iBAT-related neurons in the paraventricular nucleus (PVN) of the hypothalamus and test the hypothesis that iBAT-related PVN neurons are modulated by leptin. Inoculation of iBAT with PRV in leptin receptor reporter mice (Lepr:EGFP) demonstrated that a population of iBAT-related PVN neurons expresses Lepr receptors. Our electrophysiological findings revealed that leptin application caused hyperpolarization in some of iBAT-related PVN neurons. Bath application of leptin also modulated excitatory and inhibitory neurotransmission to most of iBAT-related PVN neurons. Using channel rhodopsin assisted circuit mapping we found that GABAergic and glutamatergic Lepr-expressing neurons in the dorsomedial hypothalamus/dorsal hypothalamic area (dDMH/DHA) project to PVN neurons; however, connected iBAT-related PVN neurons receive exclusively inhibitory signals from Lepr-expressing dDMH/DHA neurons.
Collapse
Affiliation(s)
- Yanyan Jiang
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States of America; Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America
| | - Kavon Rezai-Zadeh
- Central Leptin Signaling, Pennington Biomedical Research Center, LSU System, Baton Rouge, LA, United States of America
| | - Lucie D Desmoulins
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States of America
| | - Heike Muenzberg
- Central Leptin Signaling, Pennington Biomedical Research Center, LSU System, Baton Rouge, LA, United States of America
| | - Andrei V Derbenev
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States of America; Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America
| | - Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States of America; Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America.
| |
Collapse
|
36
|
Anjali, Vk G, Sarma L, Tripathi M, Verma MR, Verma V, Pathak MC, Samad HA, Maurya VP, Chouhan VS, Singh G. Thyroid hormone dynamics of Tharparkar and Sahiwal cattle during induced heat stress. Trop Anim Health Prod 2023; 55:57. [PMID: 36715891 DOI: 10.1007/s11250-023-03477-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 01/17/2023] [Indexed: 01/31/2023]
Abstract
Thyroid hormones and Cortisol level are the essential biomarkers in the assessment of stress condition. This study was done to estimate the metabolic hormonal profile of Tharparkar and Sahiwal during heat stress condition. The experiment was conducted on two groups consisting of Tharparkar and Sahiwal animals (5 in each group) and the experimental period comprised a 7-day acclimatization period, a heat exposure period of 21 days at control (25 °C), moderate (35 °C) and severe (42 °C) heat stress within a 9-10-day recovery period between each exposure. The hormonal concentrations of T3, T4 and cortisol were determined in serum. The serum concentration of Thyroxine (T4) and tri-iodothyronine (T3) decreases whereas cortisol level increases in both the breeds when subjected to heat stress. However, the serum level of T4 was significantly (p < 0.05) more declined in Sahiwal as compared to Tharparkar but there was no significant difference found between the two breeds in serum T3 levels. The cortisol levels were elevated in both breeds during heat stress but significantly (p < 0.05) more elevated in the Sahiwal. Hence, observations of these hormonal profiles suggest a better thermo-adaptability in Tharparkar as compared to Sahiwal.
Collapse
Affiliation(s)
- Anjali
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 UP, India
| | - Gururaj Vk
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 UP, India
| | - Lipika Sarma
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 UP, India
| | - M Tripathi
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 UP, India
| | - Med Ram Verma
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 UP, India
| | - Vinay Verma
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 UP, India
| | - M C Pathak
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 UP, India
| | - H A Samad
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 UP, India
| | - V P Maurya
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 UP, India
| | - V S Chouhan
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 UP, India
| | - Gyanendra Singh
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 UP, India.
| |
Collapse
|
37
|
Buban KN, Saperstein SE, Oyola MG, Rothwell SW, John Wu T. Alterations in the activation of corticotropin-releasing factor neurons in the paraventricular nucleus following a single or multiple days of sleep restriction. Neurosci Lett 2023; 792:136940. [PMID: 36336086 DOI: 10.1016/j.neulet.2022.136940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/17/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
Sleep disturbances are common among disorders associated with hypothalamic pituitary-adrenal (HPA) axis dysfunction, such as depression and anxiety. This comorbidity may partly be the result of the intersection between the role of the HPA axis in mediating the stress response and its involvement in sleep-wake cyclicity. Our previous work has shown that following 20 h of sleep restriction, mice show a blunting of the HPA axis in response to an acute stressor. Furthermore, these responses differ in a sex-dependent manner. This study sought to examine the effect of sleep restriction on corticotropin-releasing factor (CRF)-containing neurons in the paraventricular nucleus (PVN) of the hypothalamus. Male and female Crf-IRES-Cre: Ai14 (Tdtomato) reporter mice were sleep restricted for 20 h daily for either a single or three consecutive days using the modified multiple platform method. These mice allowed the visualization of CRF+ neurons throughout the brain. Animals were subjected to acute restraint stress, and their brains were collected to assess PVN neuronal activation via c-Fos immunohistochemistry. Analyses of cell counts revealed an ablation of the restraint-induced increase in both CRF/c-Fos colocalization and overall c-Fos expression in female mice following both a single day and three days of sleep restriction. Males showed an overall decrease in restraint-induced c-Fos levels following a single day of sleep restriction. However, male mice examined after three days of sleep restriction showed a recovery in PVN-CRF and overall PVN neuronal activation. These data suggest the sex dependent dysregulation in CRF function following sleep restriction.
Collapse
Affiliation(s)
- Katelyn N Buban
- Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Samantha E Saperstein
- Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Mario G Oyola
- Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Stephen W Rothwell
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - T John Wu
- Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
38
|
Sánchez-Lafuente CL, Romay-Tallon R, Allen J, Johnston JN, Kalynchuk LE, Caruncho HJ. Sex differences in basal reelin levels in the paraventricular hypothalamus and in response to chronic stress induced by repeated corticosterone in rats. Horm Behav 2022; 146:105267. [PMID: 36274499 DOI: 10.1016/j.yhbeh.2022.105267] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/21/2022] [Accepted: 09/24/2022] [Indexed: 11/04/2022]
Abstract
Repeated exposure to the stress hormone corticosterone results in depressive-like behaviours paralleled by the downregulation of hippocampal reelin expression. Reelin is expressed in key neural populations involved in the stress response, but whether its hypothalamic expression is sex-specific or involved in sex-specific vulnerability to stress is unknown. Female and male rats were treated with either daily vehicle or corticosterone injections (40 mg/kg) for 21 days. Thereafter, they were subjected to several behavioural tasks before being sacrificed to allow the analysis of reelin expression in hypothalamic nuclei. The basal density of reelin-positive cells in males was significantly higher in the paraventricular nucleus (19 %) and in the medial preoptic area (51 %) compared to females. Chronic corticosterone injections increased the immobility time in the forced swim test in males (107 %) and females (108 %) and decreased the exploration of the elevated plus maze in males (34 %). Corticosterone also caused a significant decrease in the density of reelin-positive cells in males, in both ventrodorsal (37 %) and ventrolateral (32 %) subdivisions of the paraventricular nucleus, while not affecting females. Moreover, in the paraventricular nucleus of males, 30 % of the basal reelin-positive cells co-expressed oxytocin while only 17.5 % did in females, showing a positive correlation between reelin and oxytocin levels. Chronic corticosterone did not significantly affect co-localization levels. For the first time, this study shows that there is a sexually dimorphic subpopulation of reelin-positive neurons in the paraventricular nucleus that can be differentially affected by chronic stress.
Collapse
Affiliation(s)
| | | | - Josh Allen
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Jenessa N Johnston
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Lisa E Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
39
|
Patel KP, Zheng H. Calcineurin Controls Hypothalamic NMDA Receptor Activity and Sympathetic Outflow. Circ Res 2022; 131:361-363. [PMID: 35926008 PMCID: PMC9351814 DOI: 10.1161/circresaha.122.321581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Kaushik P Patel
- Department of Integrative and Cellular Physiology, University of Nebraska Medical Center, Omaha (K.P.P.)
| | - Hong Zheng
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion (H.Z.)
| |
Collapse
|
40
|
Islam MT, Rumpf F, Tsuno Y, Kodani S, Sakurai T, Matsui A, Maejima T, Mieda M. Vasopressin neurons in the paraventricular hypothalamus promote wakefulness via lateral hypothalamic orexin neurons. Curr Biol 2022; 32:3871-3885.e4. [PMID: 35907397 DOI: 10.1016/j.cub.2022.07.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 06/11/2022] [Accepted: 07/08/2022] [Indexed: 01/25/2023]
Abstract
The sleep-wakefulness cycle is regulated by complicated neural networks that include many different populations of neurons throughout the brain. Arginine vasopressin neurons in the paraventricular nucleus of the hypothalamus (PVHAVP) regulate various physiological events and behaviors, such as body-fluid homeostasis, blood pressure, stress response, social interaction, and feeding. Changes in arousal level often accompany these PVHAVP-mediated adaptive responses. However, the contribution of PVHAVP neurons to sleep-wakefulness regulation has remained unknown. Here, we report the involvement of PVHAVP neurons in arousal promotion. Optogenetic stimulation of PVHAVP neurons rapidly induced transitions to wakefulness from both NREM and REM sleep. This arousal effect was dependent on AVP expression in these neurons. Similarly, chemogenetic activation of PVHAVP neurons increased wakefulness and reduced NREM and REM sleep, whereas chemogenetic inhibition of these neurons significantly reduced wakefulness and increased NREM sleep. We observed dense projections of PVHAVP neurons in the lateral hypothalamus with potential connections to orexin/hypocretin (LHOrx) neurons. Optogenetic stimulation of PVHAVP neuronal fibers in the LH immediately induced wakefulness, whereas blocking orexin receptors attenuated the arousal effect of PVHAVP neuronal activation drastically. Monosynaptic rabies-virus tracing revealed that PVHAVP neurons receive inputs from multiple brain regions involved in sleep-wakefulness regulation, as well as those involved in stress response and energy metabolism. Moreover, PVHAVP neurons mediated the arousal induced by novelty stress and a melanocortin receptor agonist melanotan-II. Thus, our data suggested that PVHAVP neurons promote wakefulness via LHOrx neurons in the basal sleep-wakefulness and some stressful conditions.
Collapse
Affiliation(s)
- Md Tarikul Islam
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Florian Rumpf
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan; Graduate School of Life Sciences, University of Würzburg, Beatrice-Edgell-Weg 21, 97074 Würzburg, Germany
| | - Yusuke Tsuno
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Shota Kodani
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Takeshi Sakurai
- Faculty of Medicine/WPI-IIIS, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Ayako Matsui
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Takashi Maejima
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan.
| |
Collapse
|
41
|
Olivares MJ, Toledo C, Ortolani D, Ortiz FC, Díaz HS, Iturriaga R, Del Río R. Sleep dysregulation in sympathetic-mediated diseases: implications for disease progression. Sleep 2022; 45:6649852. [DOI: 10.1093/sleep/zsac166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/18/2022] [Indexed: 11/12/2022] Open
Abstract
Abstract
The autonomic nervous system (ANS) plays an important role in the coordination of several physiological functions including sleep/wake process. Significant changes in ANS activity occur during wake-to-sleep transition maintaining the adequate cardiorespiratory regulation and brain activity. Since sleep is a complex homeostatic function, partly regulated by the ANS, it is not surprising that sleep disruption trigger and/or evidence symptoms of ANS impairment. Indeed, several studies suggest a bidirectional relationship between impaired ANS function (i.e. enhanced sympathetic drive), and the emergence/development of sleep disorders. Furthermore, several epidemiological studies described a strong association between sympathetic-mediated diseases and the development and maintenance of sleep disorders resulting in a vicious cycle with adverse outcomes and increased mortality risk. However, which and how the sleep/wake control and ANS circuitry becomes affected during the progression of ANS-related diseases remains poorly understood. Thus, understanding the physiological mechanisms underpinning sleep/wake-dependent sympathetic modulation could provide insights into diseases involving autonomic dysfunction. The purpose of this review is to explore potential neural mechanisms involved in both the onset/maintenance of sympathetic-mediated diseases (Rett syndrome, congenital central hypoventilation syndrome, obstructive sleep apnoea, type 2 diabetes, obesity, heart failure, hypertension, and neurodegenerative diseases) and their plausible contribution to the generation of sleep disorders in order to review evidence that may serve to establish a causal link between sleep disorders and heightened sympathetic activity.
Collapse
Affiliation(s)
- María José Olivares
- Department of Physiology, Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Camilo Toledo
- Department of Physiology, Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile , Santiago , Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes , Punta Arenas , Chile
| | - Domiziana Ortolani
- Department of Physiology, Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Fernando C Ortiz
- Mechanisms of Myelin Formation and Repair Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile , Santiago , Chile
| | - Hugo S Díaz
- Department of Physiology, Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile , Santiago , Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes , Punta Arenas , Chile
| | - Rodrigo Iturriaga
- Department of Physiology, Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile , Santiago , Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes , Punta Arenas , Chile
| | - Rodrigo Del Río
- Department of Physiology, Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile , Santiago , Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes , Punta Arenas , Chile
- Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile , Santiago , Chile
| |
Collapse
|
42
|
Picard A, Berney X, Castillo-Armengol J, Tarussio D, Jan M, Sanchez-Archidona AR, Croizier S, Thorens B. Hypothalamic Irak4 is a genetically controlled regulator of hypoglycemia-induced glucagon secretion. Mol Metab 2022; 61:101479. [PMID: 35339728 PMCID: PMC9046887 DOI: 10.1016/j.molmet.2022.101479] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVES Glucagon secretion to stimulate hepatic glucose production is the first line of defense against hypoglycemia. This response is triggered by so far incompletely characterized central hypoglycemia-sensing mechanisms, which control autonomous nervous activity and hormone secretion. The objective of this study was to identify novel hypothalamic genes controlling insulin-induced glucagon secretion. METHODS To obtain new information on the mechanisms of hypothalamic hypoglycemia sensing, we combined genetic and transcriptomic analysis of glucagon response to insulin-induced hypoglycemia in a panel of BXD recombinant inbred mice. RESULTS We identified two QTLs on chromosome 8 and chromosome 15. We further investigated the role of Irak4 and Cpne8, both located in the QTL on chromosome 15, in C57BL/6J and DBA/2J mice, the BXD mouse parental strains. We found that the poor glucagon response of DBA/2J mice was associated with higher hypothalamic expression of Irak4, which encodes a kinase acting downstream of the interleukin-1 receptor (Il-1R), and of Il-ß when compared with C57BL/6J mice. We showed that intracerebroventricular administration of an Il-1R antagonist in DBA/2J mice restored insulin-induced glucagon secretion; this was associated with increased c-fos expression in the arcuate and paraventricular nuclei of the hypothalamus and with higher activation of both branches of the autonomous nervous system. Whole body inactivation of Cpne8, which encodes a Ca++-dependent regulator of membrane trafficking and exocytosis, however, had no impact on insulin-induced glucagon secretion. CONCLUSIONS Collectively, our data identify Irak4 as a genetically controlled regulator of hypoglycemia-activated hypothalamic neurons and glucagon secretion.
Collapse
Affiliation(s)
- Alexandre Picard
- Center for Integrative Genomics, University of Lausanne, 1015, Lausanne, Switzerland
| | - Xavier Berney
- Center for Integrative Genomics, University of Lausanne, 1015, Lausanne, Switzerland
| | - Judit Castillo-Armengol
- Center for Integrative Genomics, University of Lausanne, 1015, Lausanne, Switzerland; Novo Nordisk A/S, Måløv, Denmark
| | - David Tarussio
- Center for Integrative Genomics, University of Lausanne, 1015, Lausanne, Switzerland
| | - Maxime Jan
- Center for Integrative Genomics, University of Lausanne, 1015, Lausanne, Switzerland
| | | | - Sophie Croizier
- Center for Integrative Genomics, University of Lausanne, 1015, Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, 1015, Lausanne, Switzerland.
| |
Collapse
|
43
|
Ichiyama A, Mestern S, Benigno GB, Scott KE, Allman BL, Muller L, Inoue W. State-dependent activity dynamics of hypothalamic stress effector neurons. eLife 2022; 11:76832. [PMID: 35770968 PMCID: PMC9278954 DOI: 10.7554/elife.76832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 06/17/2022] [Indexed: 11/30/2022] Open
Abstract
The stress response necessitates an immediate boost in vital physiological functions from their homeostatic operation to an elevated emergency response. However, the neural mechanisms underlying this state-dependent change remain largely unknown. Using a combination of in vivo and ex vivo electrophysiology with computational modeling, we report that corticotropin releasing hormone (CRH) neurons in the paraventricular nucleus of the hypothalamus (PVN), the effector neurons of hormonal stress response, rapidly transition between distinct activity states through recurrent inhibition. Specifically, in vivo optrode recording shows that under non-stress conditions, CRHPVN neurons often fire with rhythmic brief bursts (RB), which, somewhat counterintuitively, constrains firing rate due to long (~2 s) interburst intervals. Stressful stimuli rapidly switch RB to continuous single spiking (SS), permitting a large increase in firing rate. A spiking network model shows that recurrent inhibition can control this activity-state switch, and more broadly the gain of spiking responses to excitatory inputs. In biological CRHPVN neurons ex vivo, the injection of whole-cell currents derived from our computational model recreates the in vivo-like switch between RB and SS, providing direct evidence that physiologically relevant network inputs enable state-dependent computation in single neurons. Together, we present a novel mechanism for state-dependent activity dynamics in CRHPVN neurons.
Collapse
|
44
|
Holt MK. The ins and outs of the caudal nucleus of the solitary tract: An overview of cellular populations and anatomical connections. J Neuroendocrinol 2022; 34:e13132. [PMID: 35509189 PMCID: PMC9286632 DOI: 10.1111/jne.13132] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/25/2022] [Accepted: 03/22/2022] [Indexed: 11/29/2022]
Abstract
The body and brain are in constant two-way communication. Driving this communication is a region in the lower brainstem: the dorsal vagal complex. Within the dorsal vagal complex, the caudal nucleus of the solitary tract (cNTS) is a major first stop for incoming information from the body to the brain carried by the vagus nerve. The anatomy of this region makes it ideally positioned to respond to signals of change in both emotional and bodily states. In turn, the cNTS controls the activity of regions throughout the brain that are involved in the control of both behaviour and physiology. This review is intended to help anyone with an interest in the cNTS. First, I provide an overview of the architecture of the cNTS and outline the wide range of neurotransmitters expressed in subsets of neurons in the cNTS. Next, in detail, I discuss the known inputs and outputs of the cNTS and briefly highlight what is known regarding the neurochemical makeup and function of those connections. Then, I discuss one group of cNTS neurons: glucagon-like peptide-1 (GLP-1)-expressing neurons. GLP-1 neurons serve as a good example of a group of cNTS neurons, which receive input from varied sources and have the ability to modulate both behaviour and physiology. Finally, I consider what we might learn about other cNTS neurons from our study of GLP-1 neurons and why it is important to remember that the manipulation of molecularly defined subsets of cNTS neurons is likely to affect physiology and behaviours beyond those monitored in individual experiments.
Collapse
Affiliation(s)
- Marie K. Holt
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| |
Collapse
|
45
|
Islam MT, Maejima T, Matsui A, Mieda M. Paraventricular hypothalamic vasopressin neurons induce self-grooming in mice. Mol Brain 2022; 15:47. [PMID: 35606816 PMCID: PMC9125887 DOI: 10.1186/s13041-022-00932-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/09/2022] [Indexed: 11/24/2022] Open
Abstract
Self-grooming plays an essential role in hygiene maintenance, thermoregulation, and stress response. However, the neural populations involved in self-grooming remain largely unknown. The paraventricular hypothalamic nucleus (PVH) has been implicated in the regulation of self-grooming. Arginine vasopressin-producing neurons are among the major neuronal populations in the PVH (PVHAVP), which play important roles in water homeostasis, blood pressure regulation, feeding, and stress response. Here, we report the critical role of PVHAVP neurons in the induction of self-grooming. Optogenetic activation of PVHAVP neurons immediately induced self-grooming in freely moving mice. Chemogenetic activation of these neurons also increased time spent self-grooming. In contrast, their chemogenetic inhibition significantly reduced naturally occurring self-grooming, suggesting that PVHAVP-induced grooming has physiological relevance. Notably, optogenetic activation of PVHAVP neurons triggered self-grooming over other adaptive behaviors, such as voracious feeding induced by fasting and social interaction with female mice. Thus, our study proposes the novel role of PVHAVP neurons in regulating self-grooming behavior and, consequently, hygiene maintenance and stress response. Furthermore, uncontrolled activation of these neurons may be potentially relevant to diseases characterized by compulsive behaviors and impaired social interaction, such as autism, obsessive–compulsive disorder, and anorexia nervosa.
Collapse
Affiliation(s)
- Md Tarikul Islam
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Takashi Maejima
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Ayako Matsui
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan.
| |
Collapse
|
46
|
Patel KP, Katsurada K, Zheng H. Cardiorenal Syndrome: The Role of Neural Connections Between the Heart and the Kidneys. Circ Res 2022; 130:1601-1617. [PMID: 35549375 PMCID: PMC9179008 DOI: 10.1161/circresaha.122.319989] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The maintenance of cardiovascular homeostasis is highly dependent on tightly controlled interactions between the heart and the kidneys. Therefore, it is not surprising that a dysfunction in one organ affects the other. This interlinking relationship is aptly demonstrated in the cardiorenal syndrome. The characteristics of the cardiorenal syndrome state include alterations in neurohumoral drive, autonomic reflexes, and fluid balance. The evidence suggests that several factors contribute to these alterations. These may include peripheral and central nervous system abnormalities. However, accumulating evidence from animals with experimental models of congestive heart failure and renal dysfunction as well as humans with the cardiorenal syndrome suggests that alterations in neural pathways, from and to the kidneys and the heart, including the central nervous system are involved in regulating sympathetic outflow and may be critically important in the alterations in neurohumoral drive, autonomic reflexes, and fluid balance commonly observed in the cardiorenal syndrome. This review focuses on studies implicating neural pathways, particularly the afferent and efferent signals from the heart and the kidneys integrating at the level of the paraventricular nucleus in the hypothalamus to alter neurohumoral drive, autonomic pathways, and fluid balance. Further, it explores the potential mechanisms of action for the known beneficial use of various medications or potential novel therapeutic manipulations for the treatment of the cardiorenal syndrome. A comprehensive understanding of these mechanisms will enhance our ability to treat cardiorenal conditions and their cardiovascular complications more efficaciously and thoroughly.
Collapse
Affiliation(s)
- Kaushik P Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (K.P.P.)
| | - Kenichi Katsurada
- Division of Cardiovascular Medicine, Department of Internal Medicine (K.K.), Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan.,Division of Clinical Pharmacology, Department of Pharmacology (K.K.), Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Hong Zheng
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion (H.Z.)
| |
Collapse
|
47
|
Wee CL, Song E, Nikitchenko M, Herrera KJ, Wong S, Engert F, Kunes S. Social isolation modulates appetite and avoidance behavior via a common oxytocinergic circuit in larval zebrafish. Nat Commun 2022; 13:2573. [PMID: 35545618 PMCID: PMC9095721 DOI: 10.1038/s41467-022-29765-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 03/28/2022] [Indexed: 12/13/2022] Open
Abstract
Animal brains have evolved to encode social stimuli and transform these representations into advantageous behavioral responses. The commonalities and differences of these representations across species are not well-understood. Here, we show that social isolation activates an oxytocinergic (OXT), nociceptive circuit in the larval zebrafish hypothalamus and that chemical cues released from conspecific animals are potent modulators of this circuit's activity. We delineate an olfactory to subpallial pathway that transmits chemical social cues to OXT circuitry, where they are transformed into diverse outputs simultaneously regulating avoidance and feeding behaviors. Our data allow us to propose a model through which social stimuli are integrated within a fundamental neural circuit to mediate diverse adaptive behaviours.
Collapse
Affiliation(s)
- Caroline L Wee
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA
- Program in Neuroscience, Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Erin Song
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA
| | - Maxim Nikitchenko
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA
- Duke University, Durham, North Carolina, USA
| | - Kristian J Herrera
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA
| | - Sandy Wong
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA
| | - Florian Engert
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA.
| | - Samuel Kunes
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA.
| |
Collapse
|
48
|
Bilbao MG, Garrigos D, Martinez-Morga M, Toval A, Kutsenko Y, Bautista R, Barreda A, Ribeiro Do-Couto B, Puelles L, Ferran JL. Prosomeric Hypothalamic Distribution of Tyrosine Hydroxylase Positive Cells in Adolescent Rats. Front Neuroanat 2022; 16:868345. [PMID: 35601999 PMCID: PMC9121318 DOI: 10.3389/fnana.2022.868345] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Most of the studies on neurochemical mapping, connectivity, and physiology in the hypothalamic region were carried out in rats and under the columnar morphologic paradigm. According to the columnar model, the entire hypothalamic region lies ventrally within the diencephalon, which includes preoptic, anterior, tuberal, and mamillary anteroposterior regions, and sometimes identifying dorsal, intermediate, and ventral hypothalamic partitions. This model is weak in providing little or no experimentally corroborated causal explanation of such subdivisions. In contrast, the modern prosomeric model uses different axial assumptions based on the parallel courses of the brain floor, alar-basal boundary, and brain roof (all causally explained). This model also postulates that the hypothalamus and telencephalon jointly form the secondary prosencephalon, separately from and rostral to the diencephalon proper. The hypothalamus is divided into two neuromeric (transverse) parts called peduncular and terminal hypothalamus (PHy and THy). The classic anteroposterior (AP) divisions of the columnar hypothalamus are rather seen as dorsoventral subdivisions of the hypothalamic alar and basal plates. In this study, we offered a prosomeric immunohistochemical mapping in the rat of hypothalamic cells expressing tyrosine hydroxylase (TH), which is the enzyme that catalyzes the conversion of L-tyrosine to levodopa (L-DOPA) and a precursor of dopamine. This mapping was also combined with markers for diverse hypothalamic nuclei [agouti-related peptide (Agrp), arginine vasopressin (Avp), cocaine and amphetamine-regulated transcript (Cart), corticotropin releasing Hormone (Crh), melanin concentrating hormone (Mch), neuropeptide Y (Npy), oxytocin/neurophysin I (Oxt), proopiomelanocortin (Pomc), somatostatin (Sst), tyrosine hidroxilase (Th), and thyrotropin releasing hormone (Trh)]. TH-positive cells are particularly abundant within the periventricular stratum of the paraventricular and subparaventricular alar domains. In the tuberal region, most labeled cells are found in the acroterminal arcuate nucleus and in the terminal periventricular stratum. The dorsal retrotuberal region (PHy) contains the A13 cell group of TH-positive cells. In addition, some TH cells appear in the perimamillary and retromamillary regions. The prosomeric model proved useful for determining the precise location of TH-positive cells relative to possible origins of morphogenetic signals, thus aiding potential causal explanation of position-related specification of this hypothalamic cell type.
Collapse
Affiliation(s)
- María G. Bilbao
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Facultad de Ciencias Veterinarias, Universidad Nacional de La Pampa, General Pico, Argentina
| | - Daniel Garrigos
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain
- Institute of Biomedical Research of Murcia – IMIB, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - Marta Martinez-Morga
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain
- Institute of Biomedical Research of Murcia – IMIB, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - Angel Toval
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain
- Institute of Biomedical Research of Murcia – IMIB, Virgen de la Arrixaca University Hospital, Murcia, Spain
- PROFITH “PROmoting FITness and Health Through Physical Activity” Research Group, Department of Physical Education and Sports, Faculty of Sport Sciences, University of Granada, Granada, Spain
| | - Yevheniy Kutsenko
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain
- Institute of Biomedical Research of Murcia – IMIB, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - Rosario Bautista
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain
- Institute of Biomedical Research of Murcia – IMIB, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - Alberto Barreda
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain
- Institute of Biomedical Research of Murcia – IMIB, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - Bruno Ribeiro Do-Couto
- Institute of Biomedical Research of Murcia – IMIB, Virgen de la Arrixaca University Hospital, Murcia, Spain
- Department of Human Anatomy and Psychobiology, Faculty of Psychology, University of Murcia, Murcia, Spain
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain
- Institute of Biomedical Research of Murcia – IMIB, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - José Luis Ferran
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain
- Institute of Biomedical Research of Murcia – IMIB, Virgen de la Arrixaca University Hospital, Murcia, Spain
| |
Collapse
|
49
|
Savić B, Murphy D, Japundžić-Žigon N. The Paraventricular Nucleus of the Hypothalamus in Control of Blood Pressure and Blood Pressure Variability. Front Physiol 2022; 13:858941. [PMID: 35370790 PMCID: PMC8966844 DOI: 10.3389/fphys.2022.858941] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/15/2022] [Indexed: 12/26/2022] Open
Abstract
The paraventricular nucleus (PVN) is a highly organized structure of the hypothalamus that has a key role in regulating cardiovascular and osmotic homeostasis. Functionally, the PVN is divided into autonomic and neuroendocrine (neurosecretory) compartments, both equally important for maintaining blood pressure (BP) and body fluids in the physiological range. Neurosecretory magnocellular neurons (MCNs) of the PVN are the main source of the hormones vasopressin (VP), responsible for water conservation and hydromineral balance, and oxytocin (OT), involved in parturition and milk ejection during lactation. Further, neurosecretory parvocellular neurons (PCNs) take part in modulation of the hypothalamic–pituitary–adrenal axis and stress responses. Additionally, the PVN takes central place in autonomic adjustment of BP to environmental challenges and contributes to its variability (BPV), underpinning the PVN as an autonomic master controller of cardiovascular function. Autonomic PCNs of the PVN modulate sympathetic outflow toward heart, blood vessels and kidneys. These pre-autonomic neurons send projections to the vasomotor nucleus of rostral ventrolateral medulla and to intermediolateral column of the spinal cord, where postganglionic fibers toward target organs arise. Also, PVN PCNs synapse with NTS neurons which are the end-point of baroreceptor primary afferents, thus, enabling the PVN to modify the function of baroreflex. Neuroendocrine and autonomic parts of the PVN are segregated morphologically but they work in concert when the organism is exposed to environmental challenges via somatodendritically released VP and OT by MCNs. The purpose of this overview is to address both neuroendocrine and autonomic PVN roles in BP and BPV regulation.
Collapse
Affiliation(s)
- Bojana Savić
- Laboratory for Cardiovascular Pharmacology and Toxicology, Faculty of Medicine, Institute of Pharmacology, Clinical Pharmacology and Toxicology, University of Belgrade, Belgrade, Serbia
| | - David Murphy
- Molecular Neuroendocrinology Research Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Nina Japundžić-Žigon
- Laboratory for Cardiovascular Pharmacology and Toxicology, Faculty of Medicine, Institute of Pharmacology, Clinical Pharmacology and Toxicology, University of Belgrade, Belgrade, Serbia
- *Correspondence: Nina Japundžić-Žigon,
| |
Collapse
|
50
|
Tanaka J, Ishikawa F, Jinno T, Miyakita M, Miyamori H, Sasaki T, Yokokawa T, Goto T, Inoue K, Matsumura S. Disruption of CRTC1 and CRTC2 in Sim1 cells strongly increases high-fat diet intake in female mice but has a modest impact on male mice. PLoS One 2022; 17:e0262577. [PMID: 35020776 PMCID: PMC8754333 DOI: 10.1371/journal.pone.0262577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/30/2021] [Indexed: 01/23/2023] Open
Abstract
cAMP responsive element binding protein (CREB)-regulated transcription coactivators (CRTCs) regulate gene transcription in response to an increase in intracellular cAMP or Ca2+ levels. To date, three isoforms of CRTC have been identified in mammals. All CRTCs are widely expressed in various regions of the brain. Numerous studies have shown the importance of CREB and CRTC in energy homeostasis. In the brain, the paraventricular nucleus of the hypothalamus (PVH) plays a critical role in energy metabolism, and CRTC1 and CRTC2 are highly expressed in PVH neuronal cells. The single-minded homolog 1 gene (Sim1) is densely expressed in PVH neurons and in some areas of the amygdala neurons. To determine the role of CRTCs in PVH on energy metabolism, we generated mice that lacked CRTC1 and CRTC2 in Sim1 cells using Sim-1 cre mice. We found that Sim1 cell-specific CRTC1 and CRTC2 double-knockout mice were sensitive to high-fat diet (HFD)-induced obesity. Sim1 cell-specific CRTC1 and CRTC2 double knockout mice showed hyperphagia specifically for the HFD, but not for the normal chow diet, increased fat mass, and no change in energy expenditure. Interestingly, these phenotypes were stronger in female mice than in male mice, and a weak phenotype was observed in the normal chow diet. The lack of CRTC1 and CRTC2 in Sim1 cells changed the mRNA levels of some neuropeptides that regulate energy metabolism in female mice fed an HFD. Taken together, our findings suggest that CRTCs in Sim1 cells regulate gene expression and suppress excessive fat intake, especially in female mice.
Collapse
Affiliation(s)
- Jin Tanaka
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Fuka Ishikawa
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Tomoki Jinno
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Motoki Miyakita
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Haruka Miyamori
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Tsutomu Sasaki
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takumi Yokokawa
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Kazuo Inoue
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Shigenobu Matsumura
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
- Department of Clinical Nutrition, Graduate School of Comprehensive Rehabilitation, Osaka Prefecture University, Osaka, Japan
- * E-mail:
| |
Collapse
|