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
|
Zhang X, Wu Z, Yang S, Wang Y, Hu S, Ji Y, Zhang Q, Bu Y, Jiang C, Huang J, Wang H, Wang D, Huang C, Jiang P, Liu C, Yang X, Yang C, Yang L, Jiang R. CD38-mediated oxytocin signaling in paraventricular nucleus contributes to empathic pain. Neuropharmacology 2025; 267:110301. [PMID: 39814130 DOI: 10.1016/j.neuropharm.2025.110301] [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: 10/21/2024] [Revised: 12/23/2024] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
Empathy plays a crucial role in social communication and the perception of affective states and behavioral processes. In this study, we observed that empathic interaction with a mouse experiencing pain resulted in decreased mechanical pain thresholds and anxiety-like behaviors in its bystander, though the underlying mechanisms remain unknown. We demonstrated that CD38 expression in the paraventricular nucleus (PVN) was upregulated during empathic pain, and the pain and emotions of CD38 knockout (CD38KO) mice as bystanders were not affected. Furthermore, fiber photometry recordings indicated that calcium activities of PVN neurons were increased during empathic pain. Interestingly, direct chemogenetic inhibition of PVN neurons attenuated the hyperalgesia and anxiety-like behaviors associated with empathic pain. In contrast, activating PVN neurons through chemogenetics in CD38KO mice induced hyperalgesia and anxiety-like effects in empathic pain. Oxytocin levels in PVN were upregulated during empathic pain, while CD38KO mice inhibit the upregulation in OXT levels, confirming that CD38 is involved in releasing brain OXT and that the CD38-OXT system in the PVN plays a role in empathic pain. Collectively, CD38-mediated oxytocin signaling in PVN is closely linked to empathic pain through its effect on the activation of PVN neurons, and it could be viable targets for novel empathic behavior interventions.
Collapse
Affiliation(s)
- Xinying Zhang
- Department of Anesthesiology, The People's Hospital of Rugao, Rugao Hospital Affiliated to Nantong University, Rugao, 226500, China; Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zifeng Wu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Siqi Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yuanyuan Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Suwan Hu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yawei Ji
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Qi Zhang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yuchen Bu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chenqi Jiang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jingyao Huang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Haoran Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Di Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chaoli Huang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Peng Jiang
- Department of Anesthesiology, Affiliated Hospital of Jiangsu University, Nanjing, 212000, China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiaolin Yang
- Department of Anesthesiology, The People's Hospital of Rugao, Rugao Hospital Affiliated to Nantong University, Rugao, 226500, China
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Ling Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China.
| | - Riyue Jiang
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
3
|
Iremonger KJ, Power EM. The paraventricular nucleus of the hypothalamus: a key node in the control of behavioural states. J Physiol 2025; 603:2231-2243. [PMID: 40119815 PMCID: PMC12013795 DOI: 10.1113/jp288366] [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: 01/07/2025] [Accepted: 03/05/2025] [Indexed: 03/24/2025] Open
Abstract
The paraventricular nucleus (PVN) of the hypothalamus contains diverse populations of neuropeptide-producing neurons. These include neurons that synthesise oxytocin, vasopressin, corticotropin-releasing hormone, thyrotropin-releasing hormone and somatostatin. While it is well established that these neurons control the secretion of neuroendocrine hormones, there is growing evidence that they also control the expression of important homeostatic behaviours. Here we review recent data showing a critical role of PVN neurons in controlling arousal, social behaviour, defensive behaviour and pain. Collectively, this suggests that the PVN is a key node in a wider neural network controlling behavioural states.
Collapse
Affiliation(s)
- Karl J. Iremonger
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| | - Emmet M. Power
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| |
Collapse
|
4
|
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
|
5
|
Bonaldo B, Casile A, Bettarelli M, Marraudino M, Gotti S. Perinatal exposure to bisphenol A or S alters differently sexual behavior and kisspeptin system in mice. ENVIRONMENTAL RESEARCH 2025; 269:120888. [PMID: 39828186 DOI: 10.1016/j.envres.2025.120888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/09/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
The effects of bisphenol A (BPA), a highly diffused endocrine-disrupting chemical found mainly in plastics, on neural circuits and behaviors are well-known. However, the effects of its substitutes have not been fully investigated. Thus, in the present study, we compare the effects of perinatal exposure to bisphenol A or S (BPS) on reproductive behaviors and related hypothalamic kisspeptin system in mice. C57BL/6J dams were orally treated with 4 μg/kg body weight/day of BPA, BPS, or vehicle from mating until the weaning of the offspring. In the adult offspring, we performed the two-bedding T-Maze test, and we observed the spontaneous sexual behavior. Exposure to BPA caused a delay in puberty onset in females, while BPS caused anticipation in males, and both altered the estrous cycle in females. The sexual and sexual-related behaviors were partially altered in males, especially in the BPA-exposed ones. Regarding the kisspeptin immunoreactivity in the analyzed hypothalamic nuclei, in BPA- or BPS-treated females, we observed an increase within the rostral periventricular area, while BPA led to an increase in the paraventricular nucleus, and BPS induced a reduction compared to control females. Among males, we observed a significant increase in the arcuate nucleus of BPA-treated males and a significant decrease in the paraventricular nucleus of BPS-treated ones. These results support the idea that perinatal exposure to low doses of either BPA or BPS is altering, in a sexually differentiated way, some reproductive-relevant parameters, sexual behaviors, and kisspeptin hypothalamic nuclei.
Collapse
Affiliation(s)
- Brigitta Bonaldo
- Department of Health Sciences and Research Center on Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy; Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043, Orbassano, Turin, Italy.
| | - Antonino Casile
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043, Orbassano, Turin, Italy; School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 9, Camerino, 62032, Italy
| | - Martina Bettarelli
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043, Orbassano, Turin, Italy
| | - Marilena Marraudino
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043, Orbassano, Turin, Italy; Department of Neuroscience "Rita Levi-Montalcini", University of Turin, Via Cherasco 15, 10126, Turin, Italy
| | - Stefano Gotti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043, Orbassano, Turin, Italy; Department of Neuroscience "Rita Levi-Montalcini", University of Turin, Via Cherasco 15, 10126, Turin, Italy
| |
Collapse
|
6
|
Broderick MZL, Khan Q, Moradikor N. Understanding the connection between stress and sleep: From underlying mechanisms to therapeutic solutions. PROGRESS IN BRAIN RESEARCH 2025; 291:137-159. [PMID: 40222777 DOI: 10.1016/bs.pbr.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
The objective of this chapter is to navigate through the nexus between stress and sleep, highlighting the neurobiological systems that connect them. Starting with an overview of neuroanatomy and physiology of stress and sleep, with a further detailed breakdown of sleep stages and key neuroanatomical centers that are responsible for sleep and wakefulness. Starting with suprachiasmatic nuclei (SCN) in circadian rhythm and sleep regulation overview, with a center point on the molecular systems including the CLOCK/CRY and BMAL1/2/PER1/2 feedback loops. Following this is the neurobiological of stress, specifically the hypothalamic-pituitary-adrenal (HPA) axis and sympathetic-adrenal (SPA) axis and influence on sleep. Vital neural circuits connecting stress and sleep are examined with the attention of the ventral tegmental area (VTA) GABA-somatostatin neurons and the locus coerules in sleep regulation in response to stress. In addition, neuroinflammation's role occurs through the cytokines IL-1β and TNF-α are investigated as a mediator of sleep disturbances caused by stress. It concludes by summarizing the implications of neuroinflammatory modulation in stress-related psychopathologies, emphasizing the opening this provides for interventions that target this inflammation helping to lighten sleep disorder.
Collapse
Affiliation(s)
| | - Qadir Khan
- Faculty of Medicine and Stomatology, Tbilisi State Medical University, Tbilisi, Georgia
| | - Nasrollah Moradikor
- International Center for Neuroscience Research, Institute for Intelligent Research, Tbilisi, Georgia.
| |
Collapse
|
7
|
Astudillo-Guerrero C, Paredes AH, Escobar J, Fernandois D, Barra R, Cruz G. Metabolic control of ovarian function through the sympathetic nervous system: role of leptin. Front Endocrinol (Lausanne) 2025; 15:1484939. [PMID: 39963180 PMCID: PMC11830616 DOI: 10.3389/fendo.2024.1484939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/26/2024] [Indexed: 02/20/2025] Open
Abstract
The link between metabolism and reproduction is well-known. Both undernutrition and obesity affect the reproductive system. Metabolic status influences reproductive physiology by regulating gonadotropin secretion and affecting reproductive organs through hormonal signals. On the other hand, the autonomic nervous system controls follicle development and ovulation in the female reproductive system. This system is regulated by hypothalamic areas associated with metabolism as the Arcuate nuclei (ARC) and paraventricular nuclei (PVN). Metabolic signals, such as nutrients and hormones, acting on the hypothalamus may play a crucial role in modulating sympathetic innervation of the ovary and other reproductive organs. Some of these hormones are leptin, insulin, and GLP-1 that act directly in the hypothalamus to activate the sympathetic nervous system. In this minireview, we propose that leptin could be an important regulator of sympathetic innervation in reproductive tissues. Leptin may affect the density or activity of sympathetic nerves, thereby affecting reproductive function. We also speculate that other hormones such as insulin and GLP-1 may activate sympathetic nerves to the ovary. Additionally, we explore how early-onset obesity can cause lasting changes in the autonomic control of metabolic and reproductive organs, especially in the ovary. This suggests that the hyperactivation of sympathetic nerves in adulthood, due to metabolic programming, could be a possible cause of reproductive and metabolic disorders, such as polycystic ovary syndrome.
Collapse
Affiliation(s)
- Camila Astudillo-Guerrero
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Alfonso H. Paredes
- Center for Neurobiochemical Studies in Endocrine Diseases, Laboratory of Neurobiochemistry, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - Jorge Escobar
- Laboratorio de Química Biológica, Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Daniela Fernandois
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, Lille, France
| | - Rafael Barra
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Gonzalo Cruz
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
8
|
Smith W, Azevedo EP. Hunger Games: A Modern Battle Between Stress and Appetite. J Neurochem 2025; 169:e70006. [PMID: 39936619 DOI: 10.1111/jnc.70006] [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: 11/21/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 02/13/2025]
Abstract
Stress, an evolutionarily adaptive mechanism, has become a pervasive challenge in modern life, significantly impacting feeding-relevant circuits that play a role in the development and pathogenesis of eating disorders (EDs). Stress activates the hypothalamic-pituitary-adrenal (HPA) axis, disrupts specific neural circuits, and dysregulates key brain regions, including the hypothalamus, hippocampus, and lateral septum. These particular structures are interconnected and key in integrating stress and feeding signals, modulating hunger, satiety, cognition, and emotional coping behaviors. Here we discuss the interplay between genetic predispositions and environmental factors that may exacerbate ED vulnerability. We also highlight the most commonly used animal models to study the mechanisms driving EDs and recent rodent studies that emphasize the discovery of novel cellular and molecular mechanisms integrating stress and feeding signals within the hippocampus-lateral septum-hypothalamus axis. In this review, we discuss the role of gut microbiome, an emerging area of research in the field of EDs and unanswered questions that persist and hinder the scientific progress, such as why some individuals remain resilient to stress while others become at high risk for the development of EDs. We finally discuss the need for future research delineating the impact of specific stressors on neural circuits, clarifying the relevance and functionality of hippocampal-septal-hypothalamic connectivity, and investigating the role of key neuropeptides such as CRH, oxytocin, and GLP-1 in human ED pathogenesis. Emerging tools like single-cell sequencing and advanced human imaging could uncover cellular and circuit-level changes in brain areas relevant for feeding in ED patients. Ultimately, by integrating basic and clinical research, science offers promising avenues for developing personalized, mechanism-based treatments targeting maladaptive eating behavior for patients suffering from EDs.
Collapse
Affiliation(s)
- Whitnei Smith
- Laboratory of Neurobiology of Behavior, Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Estefania P Azevedo
- Laboratory of Neurobiology of Behavior, Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
9
|
Campos-Sánchez JC, Cabrera-Álvarez MJ, Saraiva JL. Review of Fish Neuropeptides: A Novel Perspective on Animal Welfare. J Comp Neurol 2025; 533:e70029. [PMID: 40008573 DOI: 10.1002/cne.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 10/09/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025]
Abstract
Neuropeptides are highly variable but widely conserved molecules, the main functions of which are the regulation and coordination of physiological processes and behaviors. They are synthesized in the nervous system and generally act on other neuronal and non-neuronal tissues or organs. In recent years, diverse neuropeptide isoforms and their receptors have been identified in different fish species, regulating functions in the neuroendocrine (e.g., corticotropin-releasing hormone and arginine vasotocin), immune (e.g., vasoactive intestinal polypeptide and somatostatin), digestive (e.g., neuropeptide Y), and reproductive (e.g., isotocin) systems, as well as in the commensal microbiota. Interestingly, all these processes carried out by neuropeptides are integrated into the nervous system and are manifested externally in the behavior and affective states of fish, thus having an impact on the modulation of these actions. In this sense, the monitoring of neuropeptides may represent a new approach to assess animal welfare, targeting both physiological and affective aspects in fish. Therefore, although there are many studies investigating the action of neuropeptides in a wide range of paradigms, especially in mammals, their study within a fish welfare framework is scarce. To the best of our knowledge, this is the first review that gathers and integrates up-to-date information on neuropeptides from an animal welfare perspective. In this review, we summarize current findings on neuropeptides in fish and discuss their possible implication in the physiological and emotional state of fish, and therefore in their welfare.
Collapse
Affiliation(s)
- Jose Carlos Campos-Sánchez
- Immunobiology for Aquaculture group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, Murcia, Spain
- Fish Ethology and Welfare Group, Centro de Ciências do Mar (CCMAR), Campus de Gambelas, Edificio 7, Universidade do Algarve - CCMAR/CIMAR-LA, Faro, Portugal
| | - María José Cabrera-Álvarez
- Fish Ethology and Welfare Group, Centro de Ciências do Mar (CCMAR), Campus de Gambelas, Edificio 7, Universidade do Algarve - CCMAR/CIMAR-LA, Faro, Portugal
- FishEthoGroup Association, Incubadora de Empresas da Universidade do Algarve Campus de Gambelas, pavilhão B1 8005-226, Faro, Portugal
| | - Joao L Saraiva
- Fish Ethology and Welfare Group, Centro de Ciências do Mar (CCMAR), Campus de Gambelas, Edificio 7, Universidade do Algarve - CCMAR/CIMAR-LA, Faro, Portugal
- FishEthoGroup Association, Incubadora de Empresas da Universidade do Algarve Campus de Gambelas, pavilhão B1 8005-226, Faro, Portugal
| |
Collapse
|
10
|
Tavares MR, Dos Santos WO, Amaral AG, List EO, Kopchick JJ, Alves GA, Frazao R, Dos Santos JDM, Cruz AG, Camporez JP, Donato J. Growth hormone receptor in VGLUT2 or Sim1 cells regulates glycemia and insulin sensitivity. Proc Natl Acad Sci U S A 2024; 121:e2407225121. [PMID: 39700135 DOI: 10.1073/pnas.2407225121] [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: 04/10/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024] Open
Abstract
Growth hormone (GH) has several metabolic effects, including a profound impact on glucose homeostasis. For example, GH oversecretion induces insulin resistance and increases the risk of developing diabetes mellitus. Here, we show that GH receptor (GHR) ablation in vesicular glutamate transporter 2 (VGLUT2)-expressing cells, which comprise a subgroup of glutamatergic neurons, led to a slight decrease in lean body mass without inducing changes in body adiposity. VGLUT2∆GHR mice exhibited reduced glycemia and improved glucose tolerance and insulin sensitivity. Among different glutamatergic neuronal populations, we found that GHR inactivation in Sim1-expressing cells recapitulated the phenotype observed in VGLUT2∆GHR mice. Furthermore, Sim1∆GHR mice exhibited reduced endogenous glucose production and improved hepatic insulin sensitivity without alterations in whole-body or muscle glucose uptake. Sim1∆GHR mice were protected against acute but not chronic diabetogenic effects of exogenous GH administration. Pharmacological activation of ATP-sensitive potassium channels in the brain normalized blood glucose levels in Sim1∆GHR mice. In conclusion, the absence of GHR signaling in VGLUT2/Sim1-expressing cells causes a persistent reduction in glycemia and improves hepatic insulin sensitivity. Central glucose-sensing mechanisms are likely involved in the reduced glycemia exhibited by Sim1∆GHR mice. The current findings uncover a mechanism involved in the effects of GHR signaling in regulating glucose homeostasis.
Collapse
Affiliation(s)
- Mariana R Tavares
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Willian O Dos Santos
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Andressa G Amaral
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Edward O List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701
| | - Guilherme A Alves
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-900, Brazil
| | - Renata Frazao
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-900, Brazil
| | - Jessica D M Dos Santos
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil
| | - Alessandra G Cruz
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil
| | - João Paulo Camporez
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| |
Collapse
|
11
|
Bansal P, Roitman MF, Jung EE. d-Amphetamine and Feeding States Cohesively Affect Locomotion and Motor Neuron Response in Zebrafish Larvae. Brain Behav 2024; 14:e70173. [PMID: 39643450 PMCID: PMC11624004 DOI: 10.1002/brb3.70173] [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: 07/31/2024] [Revised: 10/05/2024] [Accepted: 11/08/2024] [Indexed: 12/09/2024] Open
Abstract
PURPOSE Amphetamine (AMPH) increases locomotor activities in animals, and the locomotor response to AMPH is further modulated by caloric deficits such as food deprivation and restriction. The increment in locomotor activity regulated by AMPH-caloric deficit concomitance can be further modulated by varying feeding schedules (e.g., acute and chronic food deprivation and acute feeding after chronic food deprivation). However, the effects of different feeding schedules on AMPH-induced locomotor activity are yet to be explicated. Here, we have explored the stimulatory responses of acutely administered D-amphetamine in locomotion under systematically varying feeding states (fed/sated and food deprivation) and schedules (chronic and acute) in zebrafish larvae. METHOD We exposed wild-type and transgenic [Tg(mnx1:GCaMP5)] zebrafish larvae to 0.7 µM concentration of AMPH and measured swimming activity and spinal motor neuron activity in vivo in real time. The analysis involved time-elapsed and cumulative manner pre- and post-AMPH treatment in four different caloric states including acute and chronic schedules of feeding and hunger. Both locomotor and motor neuron activities were compared in all four states in both fish lines. FINDINGS Our results show that locomotion and motor neuron activity increased in both chronic and acute food deprivation post-AMPH treatment cumulatively. A steady increase in locomotion was observed in acute food deprivation compared to an immediate abrupt increase in chronic food-deprivation state. The ad libitum-fed larvae exhibited a moderate increase both in locomotion and motor neuron activity. Conversely to all other caloric states, food-sated (acute feeding after chronic food deprivation) larvae moved moderately less and exhibited a mild decrease in motor neuron activity after AMPH treatment. CONCLUSION These results reveal the importance of cohesive effects of feeding schedule and AMPH treatment by revealing the changes in stimulatory characteristics of AMPH on locomotion and motor neuron activity in acute and chronic feeding states.
Collapse
Affiliation(s)
- Pushkar Bansal
- Department of Mechanical and Industrial EngineeringThe University of Illinois at ChicagoChicagoIllinoisUSA
| | - Mitchell F. Roitman
- Department of PsychologyThe University of Illinois at ChicagoChicagoIllinoisUSA
| | - Erica E. Jung
- Department of Mechanical and Industrial EngineeringThe University of Illinois at ChicagoChicagoIllinoisUSA
- Department of BioengineeringThe University of Illinois at ChicagoChicagoIllinoisUSA
| |
Collapse
|
12
|
Farmer AL, Febo M, Wilkes BJ, Lewis MH. Environmental Enrichment Attenuates Repetitive Behavior and Alters the Functional Connectivity of Pain and Sensory Pathways in C58 Mice. Cells 2024; 13:1933. [PMID: 39682680 PMCID: PMC11640393 DOI: 10.3390/cells13231933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 12/18/2024] Open
Abstract
Restricted repetitive behaviors (RRB) encompass a variety of inflexible behaviors, which are diagnostic for autism spectrum disorder (ASD). Despite being requisite diagnostic criteria, the neurocircuitry of these behaviors remains poorly understood, limiting treatment development. Studies in translational animal models show environmental enrichment (EE) reduces the expression of RRB, although the underlying mechanisms are largely unknown. This study used functional magnetic resonance imaging to identify functional connectivity alterations associated with RRB and its attenuation by EE in C58 mice, an animal model of RRB. Extensive differences were observed between C58 mice and C57BL/6 control mice. Higher RRB was associated with altered connectivity between the somatosensory network and reticular thalamic nucleus and between striatal and sensory processing regions. Animals housed in EE displayed increased connectivity between the somatosensory network and the anterior pretectal nucleus and hippocampus, as well as reduced connectivity between the visual network and area prostriata. These results suggest aberrant sensory perception is associated with RRB in C58 mice. EE may reduce RRB by altering functional connectivity in pain and visual networks. This study raises questions about the role of sensory processing and pain in RRB development and identifies new potential intervention targets.
Collapse
Affiliation(s)
- Anna L. Farmer
- Department of Psychology, University of Florida, Gainesville, FL 32603, USA;
| | - Marcelo Febo
- Department of Psychiatry, University of Florida, Gainesville, FL 32611, USA;
| | - Bradley J. Wilkes
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32608, USA;
| | - Mark H. Lewis
- Department of Psychology, University of Florida, Gainesville, FL 32603, USA;
- Department of Psychiatry, University of Florida, Gainesville, FL 32611, USA;
| |
Collapse
|
13
|
Lu Y, Wang YD, Xu TQ, Zhao XH, Zhou J, Jin LH, Liu JJ. Pyridostigmine attenuates hypertension by inhibiting activation of the renin-angiotensin system in the hypothalamic paraventricular nucleus. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7995-8007. [PMID: 38767671 DOI: 10.1007/s00210-024-03156-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024]
Abstract
Activation of the renin-angiotensin system (RAS) triggers oxidative stress and an inflammatory response in the hypothalamic paraventricular nucleus (PVN), in turn increasing the sympathetic hyperactivity that is a major cause of hypertension. Pyridostigmine has cardioprotective effects by suppressing the RAS of myocardial tissue. However, whether pyridostigmine attenuates hypertension by inhibiting the RAS of the PVN remains unclear. We thus investigated the effect and mechanism of pyridostigmine on two-kidney one-clip (2K1C)-induced hypertension. 2K1C rats received pyridostigmine, or not, for 8 weeks. Cardiovascular function, hemodynamic parameters, and autonomic activity were measured. The PVN levels of pro-/anti-inflammatory cytokines, oxidative stress, and RAS signaling molecules were evaluated. Our results showed that hypertension was accompanied by cardiovascular dysfunction and an autonomic imbalance characterized by enhanced sympathetic but diminished vagal activity. The PVN levels of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), reactive oxygen species (ROS), NOX-2, and malondialdehyde (MDA) increased; those of IL-10 and superoxide dismutase (SOD) decreased. Moreover, the RAS signaling pathway was activated, as evidenced by increased levels of the angiotensin-converting enzyme (ACE), angiotensin II (Ang II), and the Ang II type 1 receptor (AT1R) and a decreased AT2R level. Pyridostigmine lowered blood pressure and improved cardiovascular function, associated with restoration of the autonomic balance. Meanwhile, pyridostigmine decreased PVN IL-6, TNF-α, ROS, NOX-2, and MDA levels and increased IL-10 and SOD levels. Additionally, pyridostigmine suppressed PVN ACE, Ang II, and AT1R levels and increased AT2R expression. Pyridostigmine attenuated hypertension by inhibiting PVN oxidative stress and inflammation induced by the RAS.
Collapse
Affiliation(s)
- Yi Lu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Pharmacy, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Yi-Dong Wang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tian-Qi Xu
- Department of Physiology and Pathophysiology, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, China
| | - Xu-He Zhao
- Department of Physiology and Pathophysiology, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, China
| | - Jun Zhou
- Department of Pharmacology, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, China
| | - Lian-Hai Jin
- Low Pressure and Low Oxygen Environment and Health Intervention Innovation Center, Jilin Medical University, Jilin, China
| | - Jin-Jun Liu
- Department of Physiology and Pathophysiology, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, China.
| |
Collapse
|
14
|
O’Reilly-Fong J, Simpson NJ, Thirouin ZS, Bastone PA, Zaelzer C, Murtaz A, Bourque CW. Acute and Reversible Hypothalamic Symptoms in a Lateral Head Impact Mouse Model of Mild Traumatic Brain Injury. Neurotrauma Rep 2024; 5:749-759. [PMID: 39184177 PMCID: PMC11342051 DOI: 10.1089/neur.2024.0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024] Open
Abstract
Central autonomic and endocrine dysfunctions following traumatic brain injury (TBI) are believed to involve the hypothalamus; however, underlying mechanisms are unknown. Although chronic deficits might be caused by irreversible tissue damage, various neuroendocrine and autonomic symptoms are only observed transiently, suggesting they might result from a temporary alteration in the activity of hypothalamic neurons. We therefore examined if a mouse model of mild TBI could induce reversible autonomic phenotypes and cause acute changes in c-Fos expression within corresponding regions of the hypothalamus. Adult C57Bl/6 male mice were lightly anesthetized with isoflurane and subjected to TBI by lateral head impact using a Gothenburg impactor. Mice treated the same way, but without the head impact served as controls (shams). We monitored body weight and core body temperature by infrared thermography and performed immunohistochemistry against c-Fos in various regions of the hypothalamus. We determined that a projectile velocity of 9 m/s significantly delayed recovery from the anesthesia without inducing skull fractures and signs of discomfort disappeared within 3 h, as assessed by grimace scale. Compared with shams, TBI mice displayed a rapid decrease in core body temperature which resolved within 48 h. Daily body weight gain was also significantly lower in TBI mice on the day following injury but recovered thereafter. c-Fos analysis revealed a significantly higher density of c-Fos-positive cells in the paraventricular nucleus and a significantly lower density in the median preoptic nucleus and medial preoptic area. We conclude that mild TBI induced by a single lateral head impact in mice at 9 m/s produces acute and reversible symptoms associated with hypothalamic dysfunction accompanied by significant changes in c-Fos expression within relevant areas of the hypothalamus. These findings support the hypothesis that a temporary alteration of neuronal activity may underlie the expression of reversible central autonomic and neuroendocrine symptoms.
Collapse
Affiliation(s)
- Julie O’Reilly-Fong
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Nick J. Simpson
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Zahra S. Thirouin
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Paolo A. Bastone
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Cristian Zaelzer
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Anzala Murtaz
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Charles W. Bourque
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| |
Collapse
|
15
|
Baker CE, Marta AG, Zimmerman ND, Korade Z, Mathy NW, Wilton D, Simeone T, Kochvar A, Kramer KL, Stessman HAF, Shibata A. CPT2 Deficiency Modeled in Zebrafish: Abnormal Neural Development, Electrical Activity, Behavior, and Schizophrenia-Related Gene Expression. Biomolecules 2024; 14:914. [PMID: 39199302 PMCID: PMC11353230 DOI: 10.3390/biom14080914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 09/01/2024] Open
Abstract
Carnitine palmitoyltransferase 2 (CPT2) is an inner mitochondrial membrane protein of the carnitine shuttle and is involved in the beta-oxidation of long chain fatty acids. Beta-oxidation provides an alternative pathway of energy production during early development and starvation. CPT2 deficiency is a genetic disorder that we recently showed can be associated with schizophrenia. We hypothesize that CPT2 deficiency during early brain development causes transcriptional, structural, and functional abnormalities that may contribute to a CNS environment that is susceptible to the emergence of schizophrenia. To investigate the effect of CPT2 deficiency on early vertebrate development and brain function, CPT2 was knocked down in a zebrafish model system. CPT2 knockdown resulted in abnormal lipid utilization and deposition, reduction in body size, and abnormal brain development. Axonal projections, neurotransmitter synthesis, electrical hyperactivity, and swimming behavior were disrupted in CPT2 knockdown zebrafish. RT-qPCR analyses showed significant increases in the expression of schizophrenia-associated genes in CPT2 knockdown compared to control zebrafish. Taken together, these data demonstrate that zebrafish are a useful model for studying the importance of beta-oxidation for early vertebrate development and brain function. This study also presents novel findings linking CPT2 deficiency to the regulation of schizophrenia and neurodegenerative disease-associated genes.
Collapse
Affiliation(s)
- Carly E. Baker
- Department of Biomedical Sciences, Creighton University, Omaha, NE 68178, USA; (C.E.B.); (K.L.K.)
| | - Aaron G. Marta
- Department of Biology, Creighton University, Omaha, NE 68178, USA; (A.G.M.); (N.D.Z.); (N.W.M.); (D.W.); (A.K.)
| | - Nathan D. Zimmerman
- Department of Biology, Creighton University, Omaha, NE 68178, USA; (A.G.M.); (N.D.Z.); (N.W.M.); (D.W.); (A.K.)
| | - Zeljka Korade
- Department of Pediatrics, Department of Biochemistry & Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68178, USA;
| | - Nicholas W. Mathy
- Department of Biology, Creighton University, Omaha, NE 68178, USA; (A.G.M.); (N.D.Z.); (N.W.M.); (D.W.); (A.K.)
| | - Delaney Wilton
- Department of Biology, Creighton University, Omaha, NE 68178, USA; (A.G.M.); (N.D.Z.); (N.W.M.); (D.W.); (A.K.)
| | - Timothy Simeone
- Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE 68178, USA; (T.S.); (H.A.F.S.)
| | - Andrew Kochvar
- Department of Biology, Creighton University, Omaha, NE 68178, USA; (A.G.M.); (N.D.Z.); (N.W.M.); (D.W.); (A.K.)
| | - Kenneth L. Kramer
- Department of Biomedical Sciences, Creighton University, Omaha, NE 68178, USA; (C.E.B.); (K.L.K.)
| | - Holly A. F. Stessman
- Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE 68178, USA; (T.S.); (H.A.F.S.)
| | - Annemarie Shibata
- Department of Biology, Creighton University, Omaha, NE 68178, USA; (A.G.M.); (N.D.Z.); (N.W.M.); (D.W.); (A.K.)
| |
Collapse
|
16
|
Feighan KM, Nesan D, Kurrasch DM. Gestational bisphenol A exposure alters energy homeostasis and adult hypothalamic neurogenesis in female mice. Sci Rep 2024; 14:16082. [PMID: 38992091 PMCID: PMC11239822 DOI: 10.1038/s41598-024-66726-2] [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: 09/29/2023] [Accepted: 07/03/2024] [Indexed: 07/13/2024] Open
Abstract
Regulation of physiological homeostasis, including energy balance, is thought to be modified by low levels of adult neurogenesis in the hypothalamus. Hormones such as oestradiol can influence both embryonic and adult hypothalamic neurogenic programs, demonstrating a sensitivity of hypothalamic neural progenitor cells to endogenous hormones. Previously we showed that gestational exposure to environmental levels of the xenoestrogen bisphenol A (BPA) changed neural progenitor cell behaviors in the embryo; however, we did not examine if these changes were permanent to affect adult neurogenesis. Here we investigated whether adult neuro- and/or gliogenesis were altered in mice prenatally exposed to BPA and placed on a high-fat diet challenge. Gestationally exposed adult female mice on a standard diet gained less weight than non-BPA controls, whereas gestationally exposed BPA females on a high-fat diet gained more weight than controls. Males exposed to gestational BPA showed no differences in weight gain relative to control males. Concomitantly, adult neurogenesis was increased in the VMH, DMH, and PVN of adult female mice exposed to BPA on standard diet, suggesting that disrupted adult neurogenesis might perturb normal energy balance regulation in females. These results add to growing evidence that low-dose BPA exposure in utero causes changes to adult hypothalamic function.
Collapse
Affiliation(s)
- Kira M Feighan
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Dinushan Nesan
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
17
|
Boomer SH, Liu X, Zheng H. Effects of regulator of G protein signaling 2 (RGS2) overexpression in the paraventricular nucleus on blood pressure in rats with angiotensin II-induced hypertension. Front Physiol 2024; 15:1401768. [PMID: 38974519 PMCID: PMC11224644 DOI: 10.3389/fphys.2024.1401768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 05/09/2024] [Indexed: 07/09/2024] Open
Abstract
The hypothalamic paraventricular nucleus (PVN) regulates sympathetic activity and blood pressure. The regulator of G protein signaling 2 (RGS2) is a negative G protein regulator, which selectively regulates G⍺q signaling, a potential cause of hypertension. This study aimed to examine angiotensin II (ANG II)-G protein-RGS2 signaling on the central mechanisms of blood pressure control, sympathetic activation, and kidney function. The Sprague Dawley rats were infused with ANG II (200 ng/kg/min) via osmotic mini pump to induce hypertension. Adenovirus (AV) vectors encoding RGS2 was transfected into the PVN in vivo. By radio telemetry measurements, we found AV-RGS2 transfection to the PVN significantly attenuated the increase of mean arterial pressure in ANG II infusion rats from days 2-7 of the 2-week experiment (Day 7: ANG II + AV-RGS2 141.3 ± 10.0 mmHg vs. ANG II 166.9 ± 9.3 mmHg, p < 0.05). AV-RGS2 transfection significantly reduced the serum norepinephrine level and acute volume reflex and increased daily urine volume and sodium excretion in ANG II-infused hypertensive rats. AV-RGS2 transfection significantly reduced G⍺q and PKC protein expressions within the PVN in ANG II infusion rats. In cultured mouse hypothalamic cells, real-time PCR study showed ANG II treatment increased mRNA expression of G⍺q, G⍺s, and RGS2, and AV-RGS2 treatment decreased ANG II-induced mRNA expression of G⍺q and G⍺s. Using confocal imagery, we found that AV-RGS2 attenuated the increase of calcium influx in ANG II-treated cells. Our results suggest that central overexpression of RGS2 in the PVN attenuated the increase of blood pressure and sympathetic outflow, and improves kidney excretory function in hypertensive rats. This may be via the alteration of ANG II-G-protein-RGS2 signaling in the central nervous system.
Collapse
Affiliation(s)
| | | | - Hong Zheng
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| |
Collapse
|
18
|
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
|
19
|
YE J, LI X, PAN Z, WU Z, ZHU Y, ZHANG W, LU J, XU S, QIN P, LIU Y, LI Y, LING Y, FANG F. Grid1 regulates the onset of puberty in female rats. J Vet Med Sci 2024; 86:497-506. [PMID: 38479882 PMCID: PMC11144544 DOI: 10.1292/jvms.23-0208] [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: 05/12/2023] [Accepted: 02/26/2024] [Indexed: 05/08/2024] Open
Abstract
The study aimed to investigate the effect of Grid1, encoding the glutamate ionotropic receptor delta type subunit 1 (GluD1), on puberty onset in female rats. Grid1 mRNA and protein expression was detected in the hypothalamus of female rats at prepuberty and puberty. The levels of Grid1 mRNA in the hypothalamus, the fluorescence intensity in the arcuate nucleus and paraventricular nucleus of the prepubertal rats was significantly lower than pubertal. Additionally, the expression of Grid1 was suppressed in primary hypothalamus cells and prepubertal rat. Finally, investigated the effect of Grid1 knockdown on puberty onset and reproductive performance. Treatment of hypothalamic neurons with LV-Grid1 decreased the level of Grid1 and Rfrp-3 (encoding RFamide-related peptide 3) mRNA expression, but increased the Gnrh (encoding gonadotropin-releasing hormone) mRNA levels. After an ICV injection, the time for the rat vaginal opening occurred earlier. Moreover, Gnrh mRNA expression was increased, whereas Rfrp-3 mRNA expression was decreased in the hypothalamus. The concentration of progesterone (P4) in the serum was significantly decreased compare with control group. Ovary hematoxylin-eosin staining revealed that the LV-Grid1 group mainly contained primary and secondary follicles. The reproductive performance of the rats was not affected by the Grid1 knockdown. Therefore, Grid1 may affect the onset of puberty in female rats by regulating the levels of Gnrh, and Rfrp-3 in the hypothalamus, as well as the concentrations of P4, but not reproduction performance.
Collapse
Affiliation(s)
- Jing YE
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Anhui,
China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, Anhui, China
| | - Xiaoqian LI
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Anhui,
China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, Anhui, China
| | - Zhihao PAN
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Anhui,
China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, Anhui, China
| | - Zhuoya WU
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Anhui,
China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, Anhui, China
| | - Yanyun ZHU
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Anhui,
China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, Anhui, China
| | - Wei ZHANG
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Anhui,
China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, Anhui, China
| | - Juntai LU
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, Anhui, China
| | - Shuangshuang XU
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Anhui,
China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, Anhui, China
| | - Ping QIN
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Anhui,
China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, Anhui, China
| | - Ya LIU
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Anhui,
China
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, Anhui, China
| | - Yunsheng LI
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Anhui,
China
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, Anhui, China
| | - Yinghui LING
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Anhui,
China
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Anhui, China
| | - Fugui FANG
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Anhui,
China
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, Anhui, China
| |
Collapse
|
20
|
Shankey NT, Cohen RE. Neural control of reproduction in reptiles. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2024; 341:307-321. [PMID: 38247297 DOI: 10.1002/jez.2783] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/23/2024]
Abstract
Reptiles display considerable diversity in reproductive behavior, making them great models to study the neuroendocrine control of reproductive behavior. Many reptile species are seasonally breeding, such that they become reproductively active during their breeding season and regress to a nonreproductive state during their nonbreeding season, with this transition often prompted by environmental cues. In this review, we will focus on summarizing the neural and neuroendocrine mechanisms controlling reproductive behavior. Three major areas of the brain are involved in reproductive behavior: the preoptic area (POA), amygdala, and ventromedial hypothalamus (VMH). The POA and VMH are sexually dimorphic areas, regulating behaviors in males and females respectively, and all three areas display seasonal plasticity. Lesions to these areas disrupt the onset and maintenance of reproductive behaviors, but the exact roles of these regions vary between sexes and species. Different hormones influence these regions to elicit seasonal transitions. Circulating testosterone (T) and estradiol (E2) peak during the breeding season and their influence on reproduction is well-documented across vertebrates. The conversion of T into E2 and 5α-dihydrotestosterone can also affect behavior. Melatonin and corticosterone have generally inhibitory effects on reproductive behavior, while serotonin and other neurohormones seem to stimulate it. In general, there is relatively little information on the neuroendocrine control of reproduction in reptiles compared to other vertebrate groups. This review highlights areas that should be considered for future areas of research.
Collapse
Affiliation(s)
- Nicholas T Shankey
- Department of Biological Sciences, Minnesota State University, Mankato, Mankato, Minnesota, USA
| | - Rachel E Cohen
- Department of Biological Sciences, Minnesota State University, Mankato, Mankato, Minnesota, USA
| |
Collapse
|
21
|
Jensen DEA, Ebmeier KP, Suri S, Rushworth MFS, Klein-Flügge MC. Nuclei-specific hypothalamus networks predict a dimensional marker of stress in humans. Nat Commun 2024; 15:2426. [PMID: 38499548 PMCID: PMC10948785 DOI: 10.1038/s41467-024-46275-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 02/21/2024] [Indexed: 03/20/2024] Open
Abstract
The hypothalamus is part of the hypothalamic-pituitary-adrenal axis which activates stress responses through release of cortisol. It is a small but heterogeneous structure comprising multiple nuclei. In vivo human neuroimaging has rarely succeeded in recording signals from individual hypothalamus nuclei. Here we use human resting-state fMRI (n = 498) with high spatial resolution to examine relationships between the functional connectivity of specific hypothalamic nuclei and a dimensional marker of prolonged stress. First, we demonstrate that we can parcellate the human hypothalamus into seven nuclei in vivo. Using the functional connectivity between these nuclei and other subcortical structures including the amygdala, we significantly predict stress scores out-of-sample. Predictions use 0.0015% of all possible brain edges, are specific to stress, and improve when using nucleus-specific compared to whole-hypothalamus connectivity. Thus, stress relates to connectivity changes in precise and functionally meaningful subcortical networks, which may be exploited in future studies using interventions in stress disorders.
Collapse
Affiliation(s)
- Daria E A Jensen
- Department of Experimental Psychology, University of Oxford, Tinsley Building, Mansfield Road, Oxford, OX1 3TA, UK.
- Wellcome Centre for Integrative Neuroimaging (WIN), Centre for Functional MRI of the Brain (FMRIB, University of Oxford, Nuffield Department of Clinical Neurosciences, Level 6, West Wing, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, UK.
- Clinic of Cognitive Neurology, University Medical Center Leipzig and Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstrasse 1a, 04103, Leipzig, Germany.
| | - Klaus P Ebmeier
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, UK
| | - Sana Suri
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, UK
- Wellcome Centre for Integrative Neuroimaging (WIN), Oxford Centre for Human Brain Activity (OHBA), University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, UK
| | - Matthew F S Rushworth
- Department of Experimental Psychology, University of Oxford, Tinsley Building, Mansfield Road, Oxford, OX1 3TA, UK
- Wellcome Centre for Integrative Neuroimaging (WIN), Centre for Functional MRI of the Brain (FMRIB, University of Oxford, Nuffield Department of Clinical Neurosciences, Level 6, West Wing, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Miriam C Klein-Flügge
- Department of Experimental Psychology, University of Oxford, Tinsley Building, Mansfield Road, Oxford, OX1 3TA, UK.
- Wellcome Centre for Integrative Neuroimaging (WIN), Centre for Functional MRI of the Brain (FMRIB, University of Oxford, Nuffield Department of Clinical Neurosciences, Level 6, West Wing, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, UK.
| |
Collapse
|
22
|
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
|
23
|
Hare MT, Carter ME, Swoap SJ. Activation of oxytocinergic neurons enhances torpor in mice. J Comp Physiol B 2024; 194:95-104. [PMID: 38170253 DOI: 10.1007/s00360-023-01528-y] [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: 09/24/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 01/05/2024]
Abstract
Mus musculus enters a torpid state in response to caloric restriction in sub-thermoneutral ambient temperatures. This torpid state is characterized by an adaptive and controlled decrease in metabolic rate, heart rate, body temperature, and activity. Previous research has identified the paraventricular nucleus (PVN) within the hypothalamus, a region containing oxytocin neurons, as a location that is active during torpor onset. We hypothesized that oxytocin neurons within the PVN are part of this neural circuit and that activation of oxytocin neurons would deepen and lengthen torpor bouts. We report that activation of oxytocin neurons alone is not sufficient to induce a torpor-like state in the fed mouse, with no significant difference in body temperature or heart rate upon activation of oxytocin neurons. However, we found that activation of oxytocin neurons prior to the onset of daily torpor both deepens and lengthens the subsequent bout, with a 1.7 ± 0.4 °C lower body temperature and a 135 ± 32 min increase in length. We therefore conclude that oxytocin neurons are involved in the neural circuitry controlling daily torpor in the mouse.
Collapse
Affiliation(s)
- Maia T Hare
- Department of Biology, Williams College, Williamstown, MA, 01267, USA
- Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY, 11549, USA
| | - Matthew E Carter
- Department of Biology, Williams College, Williamstown, MA, 01267, USA
| | - Steven J Swoap
- Department of Biology, Williams College, Williamstown, MA, 01267, USA.
| |
Collapse
|
24
|
Kang K, Shi K, Liu J, Li N, Wu J, Zhao X. Autonomic dysfunction and treatment strategies in intracerebral hemorrhage. CNS Neurosci Ther 2024; 30:e14544. [PMID: 38372446 PMCID: PMC10875714 DOI: 10.1111/cns.14544] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/15/2023] [Accepted: 11/17/2023] [Indexed: 02/20/2024] Open
Abstract
AIMS Autonomic dysfunction with central autonomic network (CAN) damage occurs frequently after intracerebral hemorrhage (ICH) and contributes to a series of adverse outcomes. This review aims to provide insight and convenience for future clinical practice and research on autonomic dysfunction in ICH patients. DISCUSSION We summarize the autonomic dysfunction in ICH from the aspects of potential mechanisms, clinical significance, assessment, and treatment strategies. The CAN structures mainly include insular cortex, anterior cingulate cortex, amygdala, hypothalamus, nucleus of the solitary tract, ventrolateral medulla, dorsal motor nucleus of the vagus, nucleus ambiguus, parabrachial nucleus, and periaqueductal gray. Autonomic dysfunction after ICH is closely associated with neurological functional outcomes, cardiac complications, blood pressure fluctuation, immunosuppression and infection, thermoregulatory dysfunction, hyperglycemia, digestive dysfunction, and urogenital disturbances. Heart rate variability, baroreflex sensitivity, skin sympathetic nerve activity, sympathetic skin response, and plasma catecholamine concentration can be used to assess the autonomic functional activities after ICH. Risk stratification of patients according to autonomic functional activities, and development of intervention approaches based on the restoration of sympathetic-parasympathetic balance, would potentially improve clinical outcomes in ICH patients. CONCLUSION The review systematically summarizes the evidence of autonomic dysfunction and its association with clinical outcomes in ICH patients, proposing that targeting autonomic dysfunction could be potentially investigated to improve the clinical outcomes.
Collapse
Affiliation(s)
- Kaijiang Kang
- Department of NeurologyBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Center of StrokeBeijing Institute for Brain DisordersBeijingChina
| | - Kaibin Shi
- Department of NeurologyBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Center of StrokeBeijing Institute for Brain DisordersBeijingChina
| | - Jiexin Liu
- Department of NeurologyBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Center of StrokeBeijing Institute for Brain DisordersBeijingChina
| | - Na Li
- Department of NeurologyBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Center of StrokeBeijing Institute for Brain DisordersBeijingChina
| | - Jianwei Wu
- Department of NeurologyBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Center of StrokeBeijing Institute for Brain DisordersBeijingChina
| | - Xingquan Zhao
- Department of NeurologyBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Center of StrokeBeijing Institute for Brain DisordersBeijingChina
- Research Unit of Artificial Intelligence in Cerebrovascular DiseaseChinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
25
|
Chapp AD, Shan Z, Chen QH. Acetic Acid: An Underestimated Metabolite in Ethanol-Induced Changes in Regulating Cardiovascular Function. Antioxidants (Basel) 2024; 13:139. [PMID: 38397737 PMCID: PMC10886048 DOI: 10.3390/antiox13020139] [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: 12/20/2023] [Revised: 01/13/2024] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
Acetic acid is a bioactive short-chain fatty acid produced in large quantities from ethanol metabolism. In this review, we describe how acetic acid/acetate generates oxidative stress, alters the function of pre-sympathetic neurons, and can potentially influence cardiovascular function in both humans and rodents after ethanol consumption. Our recent findings from in vivo and in vitro studies support the notion that administration of acetic acid/acetate generates oxidative stress and increases sympathetic outflow, leading to alterations in arterial blood pressure. Real-time investigation of how ethanol and acetic acid/acetate modulate neural control of cardiovascular function can be conducted by microinjecting compounds into autonomic control centers of the brain and measuring changes in peripheral sympathetic nerve activity and blood pressure in response to these compounds.
Collapse
Affiliation(s)
- Andrew D. Chapp
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zhiying Shan
- Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, MI 49931, USA;
| | - Qing-Hui Chen
- Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, MI 49931, USA;
| |
Collapse
|
26
|
Savani R, Park E, Busannagari N, Lu Y, Kwon H, Wang L, Pang Z. Metabolic and behavioral alterations associated with viral vector-mediated toxicity in the paraventricular hypothalamic nucleus. Biosci Rep 2024; 44:BSR20231846. [PMID: 38227343 PMCID: PMC10830444 DOI: 10.1042/bsr20231846] [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: 10/25/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 01/17/2024] Open
Abstract
OBJECTIVE Combining adeno-associated virus (AAV)-mediated expression of Cre recombinase with genetically modified floxed animals is a powerful approach for assaying the functional role of genes in regulating behavior and metabolism. Extensive research in diverse cell types and tissues using AAV-Cre has shown it can save time and avoid developmental compensation as compared to using Cre driver mouse line crossings. We initially sought to study the impact of ablation of corticotropin-releasing hormone (CRH) in the paraventricular hypothalamic nucleus (PVN) using intracranial AAV-Cre injection in adult animals. METHODS In this study, we stereotactically injected AAV8-hSyn-Cre or a control AAV8-hSyn-GFP both Crh-floxed and wild-type mouse PVN to assess behavioral and metabolic impacts. We then used immunohistochemical markers to systematically evaluate the density of hypothalamic peptidergic neurons and glial cells. RESULTS We found that delivery of one specific preparation of AAV8-hSyn-Cre in the PVN led to the development of obesity, hyperphagia, and anxiety-like behaviors. This effect occurred independent of sex and in both floxed and wild-type mice. We subsequently found that AAV8-hSyn-Cre led to neuronal cell death and gliosis at the site of viral vector injections. These behavioral and metabolic deficits were dependent on injection into the PVN. An alternatively sourced AAV-Cre did not reproduce the same results. CONCLUSIONS Our findings reveal that delivery of a specific batch of AAV-Cre could lead to cellular toxicity and lesions in the PVN that cause robust metabolic and behavioral impacts. These alterations can complicate the interpretation of Cre-mediated gene knockout and highlight the need for rigorous controls.
Collapse
Affiliation(s)
- Rohan Savani
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
- Department of Cell Biology and Neuroscience, Undergraduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
| | - Erin Park
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
- Department of Cell Biology and Neuroscience, Undergraduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
| | - Nidhi Busannagari
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
- Department of Cell Biology and Neuroscience, Undergraduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
| | - Yi Lu
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
| | - Hyokjoon Kwon
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, U.S.A
| | - Le Wang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
| | - Zhiping P. Pang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
- Department of Neuroscience and Cell Biology, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, U.S.A
- Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, U.S.A
| |
Collapse
|
27
|
Mitchell CS, Campbell EJ, Fisher SD, Stanton LM, Burton NJ, Pearl AJ, McNally GP, Bains JS, Füzesi T, Graham BA, Manning EE, Dayas CV. Optogenetic recruitment of hypothalamic corticotrophin-releasing-hormone (CRH) neurons reduces motivational drive. Transl Psychiatry 2024; 14:8. [PMID: 38191479 PMCID: PMC10774335 DOI: 10.1038/s41398-023-02710-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 11/20/2023] [Accepted: 12/12/2023] [Indexed: 01/10/2024] Open
Abstract
Impaired motivational drive is a key feature of depression. Chronic stress is a known antecedent to the development of depression in humans and depressive-like states in animals. Whilst there is a clear relationship between stress and motivational drive, the mechanisms underpinning this association remain unclear. One hypothesis is that the endocrine system, via corticotropin-releasing hormone (CRH) in the paraventricular nucleus of the hypothalamus (PVN; PVNCRH), initiates a hormonal cascade resulting in glucocorticoid release, and that excessive glucocorticoids change brain circuit function to produce depression-related symptoms. Another mostly unexplored hypothesis is that the direct activity of PVNCRH neurons and their input to other stress- and reward-related brain regions drives these behaviors. To further understand the direct involvement of PVNCRH neurons in motivation, we used optogenetic stimulation to activate these neurons 1 h/day for 5 consecutive days and showed increased acute stress-related behaviors and long-lasting deficits in the motivational drive for sucrose. This was associated with increased Fos-protein expression in the lateral hypothalamus (LH). Direct stimulation of the PVNCRH inputs in the LH produced a similar pattern of effects on sucrose motivation. Together, these data suggest that PVNCRH neuronal activity may be directly responsible for changes in motivational drive and that these behavioral changes may, in part, be driven by PVNCRH synaptic projections to the LH.
Collapse
Affiliation(s)
- Caitlin S Mitchell
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia
| | - Erin J Campbell
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia
| | - Simon D Fisher
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia
| | - Laura M Stanton
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia
| | - Nicholas J Burton
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia
| | - Amy J Pearl
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia
| | - Gavan P McNally
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia
| | - Jaideep S Bains
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Tamás Füzesi
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Brett A Graham
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia
| | - Elizabeth E Manning
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia.
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia.
| | - Christopher V Dayas
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia.
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia.
| |
Collapse
|
28
|
Savani R, Park E, Busannagari N, Lu Y, Kwon H, Wang L, Pang ZP. Metabolic and behavioral alterations associated with viral vector-mediated toxicity in the paraventricular hypothalamic nucleus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.26.564009. [PMID: 37961695 PMCID: PMC10634907 DOI: 10.1101/2023.10.26.564009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Objective Combining adeno-associated virus (AAV)-mediated expression of Cre recombinase with genetically modified floxed animals is a powerful approach for assaying the functional role of genes in regulating behavior and metabolism. Extensive research in diverse cell types and tissues using AAV-Cre has shown it can save time and avoid developmental compensation as compared to using Cre driver mouse line crossings. We initially sought to study the impact of ablation of corticotropin-releasing hormone (CRH) in the paraventricular hypothalamic nucleus (PVN) using intracranial AAV-Cre injection in adult animals. Methods In this study, we stereotactically injected AAV8-hSyn-Cre or a control AAV8-hSyn-GFP both Crh-floxed and wild-type mouse PVN to assess behavioral and metabolic impacts. We then used immunohistochemical markers to systematically evaluate the density of hypothalamic peptidergic neurons and glial cells. Results We found that delivery of one specific preparation of AAV8-hSyn-Cre in the PVN led to the development of obesity, hyperphagia, and anxiety-like behaviors. This effect occurred independent of sex and in both floxed and wild-type mice. We subsequently found that AAV8-hSyn-Cre led to neuronal cell death and gliosis at the site of viral vector injections. These behavioral and metabolic deficits were dependent on injection into the PVN. An alternatively sourced AAV-Cre did not reproduce the same results. Conclusions Our findings reveal that delivery of a specific batch of AAV-Cre could lead to cellular toxicity and lesions in the PVN that cause robust metabolic and behavioral impacts. These alterations can complicate the interpretation of Cre-mediated gene knockout and highlight the need for rigorous controls.
Collapse
Affiliation(s)
- Rohan Savani
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Department of Cell Biology and Neuroscience, Undergraduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Erin Park
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Department of Cell Biology and Neuroscience, Undergraduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Nidhi Busannagari
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Department of Cell Biology and Neuroscience, Undergraduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Yi Lu
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Hyokjoon Kwon
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Le Wang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Zhiping P. Pang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
29
|
Akbari A, Jelodar G, Hosseinzadeh S. Injection of resistin into the paraventricular nucleus produces a cardiovascular response that may be mediated by glutamatergic transmission in the rostral ventrolateral medulla. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:39-48. [PMID: 38164476 PMCID: PMC10722481 DOI: 10.22038/ijbms.2023.69324.15110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 07/25/2023] [Indexed: 01/03/2024]
Abstract
Objectives High levels of resistin are associated with metabolic diseases and their complications, including hypertension. The paraventricular nucleus (PVN) is also involved in metabolic disorders and cardiovascular diseases, such as hypertension. Therefore, this study aimed to study cardiovascular (CV) responses evoked by the injection of resistin into the lateral ventricle (LV) and PVN and determine the mechanism of these responses in the rostral ventrolateral medulla (RVLM). Materials and Methods Arterial pressure (AP) and heart rate (HR) were evaluated in urethane-anesthetized male rats (1.4 g/kg intraperitoneally) before and after all injections. This study was carried out in two stages. Resistin was injected into LV at the first stage, and AP and HR were evaluated. After that, the paraventricular, supraoptic, and dorsomedial nuclei of the hypothalamus were chosen to evaluate the gene expression of c-Fos. Afterward, resistin was injected into PVN, and cardiovascular responses were monitored. Then to detect possible neural mechanisms of resistin action, agonists or antagonists of glutamatergic, GABAergic, cholinergic, and aminergic transmissions were injected into RVLM. Results Resistin injection into LV or PVN could increase AP and HR compared to the control group and before injection. Resistin injection into LV also increases the activity of RVLM, paraventricular, supraoptic, and dorsomedial areas. Moreover, the CV reflex created by the administration of resistin in PVN is probably mediated by glutamatergic transmission within RVLM. Conclusion It can be concluded that hypothalamic nuclei, including paraventricular, are important central areas for resistin actions, and glutamatergic transmission in RVLM may be one of the therapeutic targets for high AP in obese people or with metabolic syndrome.
Collapse
Affiliation(s)
- Abolfazl Akbari
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Gholamali Jelodar
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Saeid Hosseinzadeh
- Department of Food Hygiene and Public Health, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
30
|
Luan JC, Zhang QJ, Zhou X, Zhou X, Gu Q, Xia JD, Song NH. Orexin receptors in paraventricular nucleus influence sexual behavior via regulating the sympathetic outflow in males. Andrology 2024; 12:198-210. [PMID: 37084406 DOI: 10.1111/andr.13444] [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: 09/06/2022] [Revised: 02/22/2023] [Accepted: 04/16/2023] [Indexed: 04/23/2023]
Abstract
BACKGROUND Orexins are hypothalamic neuropeptides associated with various neurophysiological activities such as sleep, arousal, and reward. However, there are few studies investigating the relationships between orexin receptors in the paraventricular nucleus and sexual behaviors. OBJECTIVES To explore the roles of orexin receptors in the paraventricular nucleus on sexual behaviors and uncover its potential mechanisms in males. MATERIALS AND METHODS Orexin A, orexin 1 receptor antagonist SB334867, and orexin 2 receptor antagonist TCS-OX2-29 were microinjected into the paraventricular nucleus to investigate the effects of orexin receptors on copulatory behavior testing of C57BL/6 mice. To explore if ejaculation could activate orexin 1 receptor-expressing neurons in the paraventricular nucleus, fluorescence immunohistochemical double staining was utilized. The levels of serum norepinephrine were measured and the lumbar sympathetic nerve activity was recorded to reflect the sympathetic nervous system activity. Moreover, the bulbospongiosus muscle-electromyogram was recorded and analyzed. To test whether perifornical/lateral hypothalamic area orexinergic neurons directly projected to the paraventricular nucleus, virus retrograde tracing technology was utilized. RESULTS Orexin A significantly enhanced sexual performance by shortening the intromission and ejaculation latencies, and increasing the mount and intromission frequencies, while the opposite outcomes appeared with SB334867. However, TCS-OX2-29 had no significant effects on sexual behaviors. Moreover, orexin A increased lumbar sympathetic nerve activity and the levels of serum norepinephrine, while SB334867 decreased lumbar sympathetic nerve activity and norepinephrine, which caused a significant decrease in sympathetic nervous system outflow. Meanwhile, a robust increase in the bulbospongiosus muscle-electromyogram activity was identified after microinjecting orexin A. Furthermore, cFos immunopositive cells were increased and double stained with orexin 1 receptor-expressing neurons in the mating group. Additionally, the retrograde tracing results demonstrated that orexinergic neurons in the perifornical/lateral hypothalamic area directly projected to the paraventricular nucleus. CONCLUSIONS Orexin 1 receptor in the paraventricular nucleus could influence the ejaculatory reflex via mediating the sympathetic nervous system activity, which might be of great importance in the treatment of premature ejaculation in the future.
Collapse
Affiliation(s)
- Jiao-Chen Luan
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi-Jie Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xuan Zhou
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiang Zhou
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi Gu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jia-Dong Xia
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ning-Hong Song
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Affiliated Kezhou People's Hospital of Nanjing Medical University, Kezhou, China
| |
Collapse
|
31
|
Van Loh BM, Yaw AM, Breuer JA, Jackson B, Nguyen D, Jang K, Ramos F, Ho EV, Cui LJ, Gillette DLM, Sempere LF, Gorman MR, Tonsfeldt KJ, Mellon PL, Hoffmann HM. The transcription factor VAX1 in VIP neurons of the suprachiasmatic nucleus impacts circadian rhythm generation, depressive-like behavior, and the reproductive axis in a sex-specific manner in mice. Front Endocrinol (Lausanne) 2023; 14:1269672. [PMID: 38205198 PMCID: PMC10777845 DOI: 10.3389/fendo.2023.1269672] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 11/28/2023] [Indexed: 01/12/2024] Open
Abstract
Background The suprachiasmatic nucleus (SCN) within the hypothalamus is a key brain structure required to relay light information to the body and synchronize cell and tissue level rhythms and hormone release. Specific subpopulations of SCN neurons, defined by their peptide expression, regulate defined SCN output. Here we focus on the vasoactive intestinal peptide (VIP) expressing neurons of the SCN. SCN VIP neurons are known to regulate circadian rhythms and reproductive function. Methods To specifically study SCN VIP neurons, we generated a novel knock out mouse line by conditionally deleting the SCN enriched transcription factor, Ventral Anterior Homeobox 1 (Vax1), in VIP neurons (Vax1Vip; Vax1fl/fl:VipCre). Results We found that Vax1Vip females presented with lengthened estrous cycles, reduced circulating estrogen, and increased depressive-like behavior. Further, Vax1Vip males and females presented with a shortened circadian period in locomotor activity and ex vivo SCN circadian period. On a molecular level, the shortening of the SCN period was driven, at least partially, by a direct regulatory role of VAX1 on the circadian clock genes Bmal1 and Per2. Interestingly, Vax1Vip females presented with increased expression of arginine vasopressin (Avp) in the paraventricular nucleus, which resulted in increased circulating corticosterone. SCN VIP and AVP neurons regulate the reproductive gonadotropin-releasing hormone (GnRH) and kisspeptin neurons. To determine how the reproductive neuroendocrine network was impacted in Vax1Vip mice, we assessed GnRH sensitivity to a kisspeptin challenge in vivo. We found that GnRH neurons in Vax1Vip females, but not males, had an increased sensitivity to kisspeptin, leading to increased luteinizing hormone release. Interestingly, Vax1Vip males showed a small, but significant increase in total sperm and a modest delay in pubertal onset. Both male and female Vax1Vip mice were fertile and generated litters comparable in size and frequency to controls. Conclusion Together, these data identify VAX1 in SCN VIP neurons as a neurological overlap between circadian timekeeping, female reproduction, and depressive-like symptoms in mice, and provide novel insight into the role of SCN VIP neurons.
Collapse
Affiliation(s)
- Brooke M. Van Loh
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, United States
| | - Alexandra M. Yaw
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, United States
| | - Joseph A. Breuer
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Brooke Jackson
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, United States
| | - Duong Nguyen
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, United States
| | - Krystal Jang
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, United States
| | - Fabiola Ramos
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, United States
| | - Emily V. Ho
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Laura J. Cui
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Dominique L. M. Gillette
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Lorenzo F. Sempere
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, United States
| | - Michael R. Gorman
- Department of Psychology, University of California, San Diego, La Jolla, CA, United States
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, United States
| | - Karen J. Tonsfeldt
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, United States
| | - Pamela L. Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, United States
| | - Hanne M. Hoffmann
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, United States
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Sun Q, Li L, Jin F, Liu Y, Yang B, Meng W, Zhang Z, Qi F. SARS-CoV-2 Spike Protein S1 Exposure Increases Susceptibility to Angiotensin II-Induced Hypertension in Rats by Promoting Central Neuroinflammation and Oxidative Stress. Neurochem Res 2023; 48:3016-3026. [PMID: 37269471 PMCID: PMC10239221 DOI: 10.1007/s11064-023-03949-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 06/05/2023]
Abstract
The SARS-CoV-2 spike S1 subunit (S1) can cross the blood-brain barrier and elicit neuroinflammatory response independent of viral infection. Here we examined whether S1 influences blood pressure (BP) and sensitizes the hypertensive response to angiotensin (ANG) II by enhancing neuroinflammation and oxidative stress in hypothalamic paraventricular nucleus (PVN), a key brain cardiovascular regulatory center. Rats received central S1 or vehicle (VEH) injection for 5 days. One week after injection, ANG II or saline (control) was subcutaneously delivered for 2 weeks. S1 injection induced greater increases in BP, PVN neuronal excitation and sympathetic drive in ANG II rats but had no effects in control rats. One week after S1 injection, mRNA for proinflammatory cytokines and oxidative stress marker were higher but mRNA of Nrf2, the master regulator of inducible antioxidant and anti-inflammatory responses, was lower in the PVN in S1-injected rats than in VEH-injected rats. Three weeks after S1 injection, mRNA for proinflammatory cytokines and oxidative stress marker, microglia activation and reactive oxygen species in the PVN were comparable between S1 and VEH treated control rats but were elevated in two groups of ANG II rats. Notably, ANG II-induced elevations in these parameters were exaggerated by S1. Interestingly, ANG II increased PVN Nrf2 mRNA in VEH-treated rats but not in S1-treated rats. These data suggest that S1 exposure has no effect on BP, but post-S1 exposure increases susceptibility to ANG II-induced hypertension by downregulating PVN Nrf2 to promote neuroinflammation and oxidative stress and augment sympathetic excitation.
Collapse
Affiliation(s)
- Qingmei Sun
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China
| | - Liang Li
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China
| | - Feihong Jin
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China
| | - Yu Liu
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China
| | - Bo Yang
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China
| | - Wanping Meng
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China
| | - Zibin Zhang
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China
| | - Feng Qi
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China.
| |
Collapse
|
35
|
Fernandez G, De Francesco PN, Cornejo MP, Cabral A, Aguggia JP, Duque VJ, Sayar N, Cantel S, Burgos JI, Fehrentz JA, Rorato R, Atasoy D, Mecawi AS, Perello M. Ghrelin Action in the PVH of Male Mice: Accessibility, Neuronal Targets, and CRH Neurons Activation. Endocrinology 2023; 164:bqad154. [PMID: 37823477 PMCID: PMC11491828 DOI: 10.1210/endocr/bqad154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/08/2023] [Accepted: 10/11/2023] [Indexed: 10/13/2023]
Abstract
The hormone ghrelin displays several well-characterized functions, including some with pharmaceutical interest. The receptor for ghrelin, the growth hormone secretagogue receptor (GHSR), is expressed in the hypothalamic paraventricular nucleus (PVH), a critical hub for the integration of metabolic, neuroendocrine, autonomic, and behavioral functions. Here, we performed a neuroanatomical and functional characterization of the neuronal types mediating ghrelin actions in the PVH of male mice. We found that fluorescent ghrelin mainly labels PVH neurons immunoreactive for nitric oxide synthase 1 (NOS1), which catalyze the production of nitric oxide [NO]). Centrally injected ghrelin increases c-Fos in NOS1 PVH neurons and NOS1 phosphorylation in the PVH. We also found that a high dose of systemically injected ghrelin increases the ghrelin level in the cerebrospinal fluid and in the periventricular PVH, and induces c-Fos in NOS1 PVH neurons. Such a high dose of systemically injected ghrelin activates a subset of NOS1 PVH neurons, which do not express oxytocin, via an arcuate nucleus-independent mechanism. Finally, we found that pharmacological inhibition of NO production fully abrogates ghrelin-induced increase of calcium concentration in corticotropin-releasing hormone neurons of the PVH whereas it partially impairs ghrelin-induced increase of plasma glucocorticoid levels. Thus, plasma ghrelin can directly target a subset of NO-producing neurons of the PVH that is involved in ghrelin-induced activation of the hypothalamic-pituitary-adrenal neuroendocrine axis.
Collapse
Affiliation(s)
- Gimena Fernandez
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Pablo N De Francesco
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - María P Cornejo
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Agustina Cabral
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Julieta P Aguggia
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Victor J Duque
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Nilufer Sayar
- Department of Neuroscience and Pharmacology, Carver College of Medicine, Iowa Neuroscience Institute and Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa, Iowa City, IA 52242, USA
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, Montpellier cedex 5 34293, France
| | - Juan I Burgos
- Centro de Investigaciones Cardiovasculares “Dr. Horacio Eugenio Cingolani” (CONICET and National University of La Plata), La Plata 1900, Buenos Aires, Argentina
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, Montpellier cedex 5 34293, France
| | - Rodrigo Rorato
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, Carver College of Medicine, Iowa Neuroscience Institute and Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa, Iowa City, IA 52242, USA
| | - André S Mecawi
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala 751 05, Sweden
| |
Collapse
|
36
|
Li C, Kühn NK, Alkislar I, Sans-Dublanc A, Zemmouri F, Paesmans S, Calzoni A, Ooms F, Reinhard K, Farrow K. Pathway-specific inputs to the superior colliculus support flexible responses to visual threat. SCIENCE ADVANCES 2023; 9:eade3874. [PMID: 37647395 PMCID: PMC10468139 DOI: 10.1126/sciadv.ade3874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 07/31/2023] [Indexed: 09/01/2023]
Abstract
Behavioral flexibility requires directing feedforward sensory information to appropriate targets. In the superior colliculus, divergent outputs orchestrate different responses to visual threats, but the circuit organization enabling the flexible routing of sensory information remains unknown. To determine this structure, we focused on inhibitory projection (Gad2) neurons. Trans-synaptic tracing and neuronal recordings revealed that Gad2 neurons projecting to the lateral geniculate nucleus (LGN) and the parabigeminal nucleus (PBG) form two separate populations, each receiving a different set of non-retinal inputs. Inhibiting the LGN- or PBG-projecting Gad2 neurons resulted in opposing effects on behavior; increasing freezing or escape probability to visual looming, respectively. Optogenetic activation of selected inputs to the LGN- and PBG-projecting Gad2 cells predictably regulated responses to visual threat. These data suggest that projection-specific sampling of brain-wide inputs provides a circuit design principle that enables visual inputs to be selectively routed to produce context-specific behavior.
Collapse
Affiliation(s)
- Chen Li
- Neuro-Electronics Research Flanders, VIB, Leuven, Belgium
- Department of Biology, KU Leuven, Leuven, Belgium
| | - Norma K. Kühn
- Neuro-Electronics Research Flanders, VIB, Leuven, Belgium
- Department of Biology, KU Leuven, Leuven, Belgium
| | - Ilayda Alkislar
- Neuro-Electronics Research Flanders, VIB, Leuven, Belgium
- Northeastern University, Boston, MA, USA
| | - Arnau Sans-Dublanc
- Neuro-Electronics Research Flanders, VIB, Leuven, Belgium
- Department of Biology, KU Leuven, Leuven, Belgium
| | - Firdaouss Zemmouri
- Neuro-Electronics Research Flanders, VIB, Leuven, Belgium
- Faculty of Pharmaceutical, Biomedical, and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Soraya Paesmans
- Neuro-Electronics Research Flanders, VIB, Leuven, Belgium
- Department of Biology, KU Leuven, Leuven, Belgium
| | - Alex Calzoni
- Neuro-Electronics Research Flanders, VIB, Leuven, Belgium
- Department of Biology, KU Leuven, Leuven, Belgium
| | - Frédérique Ooms
- Neuro-Electronics Research Flanders, VIB, Leuven, Belgium
- Imec, Leuven, Belgium
| | - Katja Reinhard
- Neuro-Electronics Research Flanders, VIB, Leuven, Belgium
- Department of Biology, KU Leuven, Leuven, Belgium
| | - Karl Farrow
- Neuro-Electronics Research Flanders, VIB, Leuven, Belgium
- Department of Biology, KU Leuven, Leuven, Belgium
- Imec, Leuven, Belgium
| |
Collapse
|
37
|
van de Poll Y, Cras Y, Ellender TJ. The neurophysiological basis of stress and anxiety - comparing neuronal diversity in the bed nucleus of the stria terminalis (BNST) across species. Front Cell Neurosci 2023; 17:1225758. [PMID: 37711509 PMCID: PMC10499361 DOI: 10.3389/fncel.2023.1225758] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/03/2023] [Indexed: 09/16/2023] Open
Abstract
The bed nucleus of the stria terminalis (BNST), as part of the extended amygdala, has become a region of increasing interest regarding its role in numerous human stress-related psychiatric diseases, including post-traumatic stress disorder and generalized anxiety disorder amongst others. The BNST is a sexually dimorphic and highly complex structure as already evident by its anatomy consisting of 11 to 18 distinct sub-nuclei in rodents. Located in the ventral forebrain, the BNST is anatomically and functionally connected to many other limbic structures, including the amygdala, hypothalamic nuclei, basal ganglia, and hippocampus. Given this extensive connectivity, the BNST is thought to play a central and critical role in the integration of information on hedonic-valence, mood, arousal states, processing emotional information, and in general shape motivated and stress/anxiety-related behavior. Regarding its role in regulating stress and anxiety behavior the anterolateral group of the BNST (BNSTALG) has been extensively studied and contains a wide variety of neurons that differ in their electrophysiological properties, morphology, spatial organization, neuropeptidergic content and input and output synaptic organization which shape their activity and function. In addition to this great diversity, further species-specific differences are evident on multiple levels. For example, classic studies performed in adult rat brain identified three distinct neuron types (Type I-III) based on their electrophysiological properties and ion channel expression. Whilst similar neurons have been identified in other animal species, such as mice and non-human primates such as macaques, cross-species comparisons have revealed intriguing differences such as their comparative prevalence in the BNSTALG as well as their electrophysiological and morphological properties, amongst other differences. Given this tremendous complexity on multiple levels, the comprehensive elucidation of the BNSTALG circuitry and its role in regulating stress/anxiety-related behavior is a major challenge. In the present Review we bring together and highlight the key differences in BNSTALG structure, functional connectivity, the electrophysiological and morphological properties, and neuropeptidergic profiles of BNSTALG neurons between species with the aim to facilitate future studies of this important nucleus in relation to human disease.
Collapse
Affiliation(s)
- Yana van de Poll
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Yasmin Cras
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Tommas J. Ellender
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
38
|
Arestakesyan H, Blackmore K, Smith HC, Popratiloff A, Young CN. Large-field-of-view scanning electron microscopy of the paraventricular nucleus of the hypothalamus during diet-induced obesity. J Neurophysiol 2023; 130:345-352. [PMID: 37435651 PMCID: PMC10396219 DOI: 10.1152/jn.00208.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/22/2023] [Accepted: 07/01/2023] [Indexed: 07/13/2023] Open
Abstract
Dysregulation in the paraventricular nucleus of the hypothalamus (PVN) is associated with a variety of diseases including those related to obesity. Although most investigations have focused on molecular changes, structural alterations in PVN neurons can reveal underlying functional disruptions. Although electron microscopy (EM) can provide nanometer resolution of brain structures, an inherent limitation of traditional transmission EM is the single field of view nature of data collection. To overcome this, we used large-field-of-view high-resolution backscatter scanning electron microscopy (bSEM) of the PVN. By stitching high-resolution bSEM images, taken from normal chow and high-fat diet mice, we achieved interactive, zoomable maps that allow for low-magnification screening of the entire PVN and high-resolution analyses of ultrastructure at the level of the smallest cellular organelle. Using this approach, quantitative analysis across the PVN revealed marked electron-dense regions within neuronal nucleoplasm following high-fat diet feeding, with an increase in kurtosis, indicative of a shift away from a normal distribution. Furthermore, measures of skewness indicated a shift toward darker clustered electron-dense regions, potentially indicative of heterochromatin clusters. We further demonstrate the utility to map out healthy and altered neurons throughout the PVN and the ability to remotely perform bSEM imaging in situations that require social distancing, such as the COVID-19 pandemic. Collectively, these findings present an approach that allows for the precise placement of PVN cells within an overall structural and functional map of the PVN. Moreover, they suggest that obesity may disrupt PVN neuronal chromatin structure.NEW & NOTEWORTHY Paraventricular nucleus of the hypothalamus (PVN) alterations are linked to obesity-related conditions, but limited knowledge exists about neuroanatomical changes in this region. A large-field-of-view backscatter scanning electron microscopy (bSEM) method was used, which allowed the identification of up to 40 PVN neurons in individual samples. During obesity in mice, bSEM revealed changes in PVN neuronal nucleoplasm, possibly indicating chromatin clustering. This microscopy advancement offers valuable insights into neuroanatomy in both healthy and disease conditions.
Collapse
Affiliation(s)
- Hovhannes Arestakesyan
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Katherine Blackmore
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Hannah C Smith
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Anastas Popratiloff
- Nanofabrication and Imaging Center, George Washington University, Washington, District of Columbia, United States
| | - Colin N Young
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| |
Collapse
|
39
|
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
|
40
|
Herget U, Ryu S, De Marco RJ. Altered glucocorticoid reactivity and behavioral phenotype in rx3-/- larval zebrafish. Front Endocrinol (Lausanne) 2023; 14:1187327. [PMID: 37484970 PMCID: PMC10358986 DOI: 10.3389/fendo.2023.1187327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction The transcription factor rx3 is important for the formation of the pituitary and parts of the hypothalamus. Mutant animals lacking rx3 function have been well characterized in developmental studies, but relatively little is known about their behavioral phenotypes. Methods We used cell type staining to reveal differences in stress axis architecture, and performed cortisol measurements and behavior analysis to study both hormonal and behavioral stress responses in rx3 mutants. Results and Discussion Consistent with the role of rx3 in hypothalamus and pituitary development, we show a distinct loss of corticotrope cells involved in stress regulation, severe reduction of pituitary innervation by hypothalamic cells, and lack of stress-induced cortisol release in rx3 mutants. Interestingly, despite these deficits, we report that rx3-/- larval zebrafish can still display nominal behavioral responses to both stressful and non-stressful stimuli. However, unlike wildtypes, mutants lacking proper pituitary-interrenal function do not show enhanced behavioral performance under moderate stress level, supporting the view that corticotroph cells are not required for behavioral responses to some types of stressful stimuli but modulate subtle behavioral adjustments under moderate stress.
Collapse
Affiliation(s)
- Ulrich Herget
- Research Group Developmental Genetics of the Nervous System, Max Planck Institute for Medical Research, Heidelberg, Germany
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Soojin Ryu
- Research Group Developmental Genetics of the Nervous System, Max Planck Institute for Medical Research, Heidelberg, Germany
- Living Systems Institute, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Rodrigo J. De Marco
- Research Group Developmental Genetics of the Nervous System, Max Planck Institute for Medical Research, Heidelberg, Germany
- School of Biological and Environmental Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
41
|
Kobori N, Moore AN, Redell JB, Dash PK. Caudal DMN neurons innervate the spleen and release CART peptide to regulate neuroimmune function. J Neuroinflammation 2023; 20:158. [PMID: 37403174 PMCID: PMC10318820 DOI: 10.1186/s12974-023-02838-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/19/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Inflammation is a fundamental biological response to injury and infection, which if unregulated can contribute to the pathophysiology of many diseases. The vagus nerve, which primarily originates from the dorsal motor nucleus (DMN), plays an important role in rapidly dampening inflammation by regulating splenic function. However, direct vagal innervation of the spleen, which houses the majority of immune and inflammatory cells, has not been established. As an alternative to direct innervation, an anti-inflammatory reflex pathway has been proposed which involves the vagus nerve, the sympathetic celiac ganglion, and the neurotransmitter norepinephrine. Although sympathetic regulation of inflammation has been shown, the interaction of the vagus nerve and the celiac ganglia requires a unique interaction of parasympathetic and sympathetic inputs, making this putative mechanism of brain-spleen interaction controversial. BODY: As neuropeptides can be expressed at relatively high levels in neurons, we reasoned that DMN neuropeptide immunoreactivity could be used to determine their target innervation. Employing immunohistochemistry, subdiaphragmatic vagotomy, viral tract tracing, CRISPR-mediated knock-down, and functional assays, we show that cocaine and amphetamine-regulated transcript (CART) peptide-expressing projection neurons in the caudal DMN directly innervate the spleen. In response to lipopolysaccharide (LPS) stimulation, CART acts to reduce inflammation, an effect that can be augmented by intrasplenic administration of a synthetic CART peptide. These in vivo effects could be recapitulated in cultured splenocytes, suggesting that these cells express the as yet unidentified CART receptor(s). CONCLUSION Our results provide evidence for direct connections between the caudal DMN and spleen. In addition to acetylcholine, these neurons express the neuropeptide CART that, once released, acts to suppress inflammation by acting directly upon splenocytes.
Collapse
Affiliation(s)
- Nobuhide Kobori
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| | - Anthony N Moore
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| | - John B Redell
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA.
| |
Collapse
|
42
|
Pan S, Worker CJ, Feng Earley Y. The hypothalamus as a key regulator of glucose homeostasis: emerging roles of the brain renin-angiotensin system. Am J Physiol Cell Physiol 2023; 325:C141-C154. [PMID: 37273237 PMCID: PMC10312332 DOI: 10.1152/ajpcell.00533.2022] [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: 11/29/2022] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/06/2023]
Abstract
The regulation of plasma glucose levels is a complex and multifactorial process involving a network of receptors and signaling pathways across numerous organs that act in concert to ensure homeostasis. However, much about the mechanisms and pathways by which the brain regulates glycemic homeostasis remains poorly understood. Understanding the precise mechanisms and circuits employed by the central nervous system to control glucose is critical to resolving the diabetes epidemic. The hypothalamus, a key integrative center within the central nervous system, has recently emerged as a critical site in the regulation of glucose homeostasis. Here, we review the current understanding of the role of the hypothalamus in regulating glucose homeostasis, with an emphasis on the paraventricular nucleus, the arcuate nucleus, the ventromedial hypothalamus, and lateral hypothalamus. In particular, we highlight the emerging role of the brain renin-angiotensin system in the hypothalamus in regulating energy expenditure and metabolic rate, as well as its potential importance in the regulation of glucose homeostasis.
Collapse
Affiliation(s)
- Shiyue Pan
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| | - Caleb J Worker
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| | - Yumei Feng Earley
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| |
Collapse
|
43
|
Garrett L, Trümbach D, Spielmann N, Wurst W, Fuchs H, Gailus-Durner V, Hrabě de Angelis M, Hölter SM. A rationale for considering heart/brain axis control in neuropsychiatric disease. Mamm Genome 2023; 34:331-350. [PMID: 36538124 PMCID: PMC10290621 DOI: 10.1007/s00335-022-09974-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
Neuropsychiatric diseases (NPD) represent a significant global disease burden necessitating innovative approaches to pathogenic understanding, biomarker identification and therapeutic strategy. Emerging evidence implicates heart/brain axis malfunction in NPD etiology, particularly via the autonomic nervous system (ANS) and brain central autonomic network (CAN) interaction. This heart/brain inter-relationship harbors potentially novel NPD diagnosis and treatment avenues. Nevertheless, the lack of multidisciplinary clinical approaches as well as a limited appreciation of molecular underpinnings has stymied progress. Large-scale preclinical multi-systemic functional data can therefore provide supplementary insight into CAN and ANS interaction. We here present an overview of the heart/brain axis in NPD and establish a unique rationale for utilizing a preclinical cardiovascular disease risk gene set to glean insights into heart/brain axis control in NPD. With a top-down approach focusing on genes influencing electrocardiogram ANS function, we combined hierarchical clustering of corresponding regional CAN expression data and functional enrichment analysis to reveal known and novel molecular insights into CAN and NPD. Through 'support vector machine' inquiries for classification and literature validation, we further pinpointed the top 32 genes highly expressed in CAN brain structures altering both heart rate/heart rate variability (HRV) and behavior. Our observations underscore the potential of HRV/hyperactivity behavior as endophenotypes for multimodal disease biomarker identification to index aberrant executive brain functioning with relevance for NPD. This work heralds the potential of large-scale preclinical functional genetic data for understanding CAN/ANS control and introduces a stepwise design leveraging preclinical data to unearth novel heart/brain axis control genes in NPD.
Collapse
Affiliation(s)
- Lillian Garrett
- German Research Center for Environmental Health, Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
- German Research Center for Environmental Health, Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Dietrich Trümbach
- German Research Center for Environmental Health, Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- German Research Center for Environmental Health, Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Nadine Spielmann
- German Research Center for Environmental Health, Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Wolfgang Wurst
- German Research Center for Environmental Health, Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Developmental Genetics, TUM School of Life Sciences, Technische Universität München, Freising-Weihenstephan, Germany
- Deutsches Institut Für Neurodegenerative Erkrankungen (DZNE) Site Munich, Feodor-Lynen-Str. 17, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 17, 81377, Munich, Germany
| | - Helmut Fuchs
- German Research Center for Environmental Health, Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Valerie Gailus-Durner
- German Research Center for Environmental Health, Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Research Center for Environmental Health, Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Alte Akademie 8, 85354, Freising, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Sabine M Hölter
- German Research Center for Environmental Health, Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany.
- German Research Center for Environmental Health, Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany.
- Technische Universität München, Freising-Weihenstephan, Germany.
- Helmholtz Center Munich, Institute of Developmental Genetics, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany.
| |
Collapse
|
44
|
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
|
45
|
Sharvin BL, Aburto MR, Cryan JF. Decoding the neurocircuitry of gut feelings: Region-specific microbiome-mediated brain alterations. Neurobiol Dis 2023; 179:106033. [PMID: 36758820 DOI: 10.1016/j.nbd.2023.106033] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Research in the last decade has unveiled a crucial role for the trillions of microorganisms that reside in the gut in influencing host neurodevelopment across the lifespan via the microbiota-gut-brain axis. Studies have linked alterations in the composition, complexity, and diversity of the gut microbiota to changes in behaviour including abnormal social interactions, cognitive deficits, and anxiety- and depressive-like phenotypes. Moreover, the microbiota has been linked with neurodevelopmental, neuropsychiatric, and neurodegenerative disorders. Interestingly, there appears to be specific brain regions governing the neurocircuitry driving higher cognitive function that are susceptible to influence from manipulations to the host microbiome. This review will aim to elucidate the region-specific effects mediated by the gut microbiota, with a focus on translational animal models and some existing human neuroimaging data. Compelling preclinical evidence suggests disruption to normal microbiota-gut-brain signalling can have detrimental effects on the prefrontal cortex, amygdala, hippocampus, hypothalamus, and striatum. Furthermore, human neuroimaging studies have unveiled a role for the microbiota in mediating functional connectivity and structure of specific brain regions that can be traced back to neurocognition and behavioural output. Understanding these microbiota-mediated changes will aid in identifying unique therapeutic targets for treating neurological disorders associated with these regions.
Collapse
Affiliation(s)
- Brendan L Sharvin
- APC Microbiome, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Maria Rodriguez Aburto
- APC Microbiome, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
46
|
Wang F, Chen Y, Lin Y, Wang X, Li K, Han Y, Wu J, Shi X, Zhu Z, Long C, Hu X, Duan S, Gao Z. A parabrachial to hypothalamic pathway mediates defensive behavior. eLife 2023; 12:85450. [PMID: 36930206 PMCID: PMC10023160 DOI: 10.7554/elife.85450] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/13/2023] [Indexed: 03/18/2023] Open
Abstract
Defensive behaviors are critical for animal's survival. Both the paraventricular nucleus of the hypothalamus (PVN) and the parabrachial nucleus (PBN) have been shown to be involved in defensive behaviors. However, whether there are direct connections between them to mediate defensive behaviors remains unclear. Here, by retrograde and anterograde tracing, we uncover that cholecystokinin (CCK)-expressing neurons in the lateral PBN (LPBCCK) directly project to the PVN. By in vivo fiber photometry recording, we find that LPBCCK neurons actively respond to various threat stimuli. Selective photoactivation of LPBCCK neurons promotes aversion and defensive behaviors. Conversely, photoinhibition of LPBCCK neurons attenuates rat or looming stimuli-induced flight responses. Optogenetic activation of LPBCCK axon terminals within the PVN or PVN glutamatergic neurons promotes defensive behaviors. Whereas chemogenetic and pharmacological inhibition of local PVN neurons prevent LPBCCK-PVN pathway activation-driven flight responses. These data suggest that LPBCCK neurons recruit downstream PVN neurons to actively engage in flight responses. Our study identifies a previously unrecognized role for the LPBCCK-PVN pathway in controlling defensive behaviors.
Collapse
Affiliation(s)
- Fan Wang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Yuge Chen
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Yuxin Lin
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Xuze Wang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Kaiyuan Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Yong Han
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Jintao Wu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Xingyi Shi
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Zhenggang Zhu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Chaoying Long
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Xiaojun Hu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang UniversityHangzhouChina
| | - Shumin Duan
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang UniversityHangzhouChina
- Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan UniversityShanghaiChina
- The Institute of Brain and Cognitive Sciences, Zhejiang University City CollegeHangzhouChina
- Chuanqi Research and Development Center of Zhejiang UniversityHangzhouChina
| | - Zhihua Gao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang UniversityHangzhouChina
| |
Collapse
|
47
|
Lauritano D, Mastrangelo F, D’Ovidio C, Ronconi G, Caraffa A, Gallenga CE, Frydas I, Kritas SK, Trimarchi M, Carinci F, Conti P. Activation of Mast Cells by Neuropeptides: The Role of Pro-Inflammatory and Anti-Inflammatory Cytokines. Int J Mol Sci 2023; 24:ijms24054811. [PMID: 36902240 PMCID: PMC10002992 DOI: 10.3390/ijms24054811] [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: 02/02/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Mast cells (MCs) are tissue cells that are derived from bone marrow stem cells that contribute to allergic reactions, inflammatory diseases, innate and adaptive immunity, autoimmunity, and mental disorders. MCs located near the meninges communicate with microglia through the production of mediators such as histamine and tryptase, but also through the secretion of IL-1, IL-6 and TNF, which can create pathological effects in the brain. Preformed chemical mediators of inflammation and tumor necrosis factor (TNF) are rapidly released from the granules of MCs, the only immune cells capable of storing the cytokine TNF, although it can also be produced later through mRNA. The role of MCs in nervous system diseases has been extensively studied and reported in the scientific literature; it is of great clinical interest. However, many of the published articles concern studies on animals (mainly rats or mice) and not on humans. MCs are known to interact with neuropeptides that mediate endothelial cell activation, resulting in central nervous system (CNS) inflammatory disorders. In the brain, MCs interact with neurons causing neuronal excitation with the production of neuropeptides and the release of inflammatory mediators such as cytokines and chemokines. This article explores the current understanding of MC activation by neuropeptide substance P (SP), corticotropin-releasing hormone (CRH), and neurotensin, and the role of pro-inflammatory cytokines, suggesting a therapeutic effect of the anti-inflammatory cytokines IL-37 and IL-38.
Collapse
Affiliation(s)
- Dorina Lauritano
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Filiberto Mastrangelo
- Department of Clinical and Experimental Medicine, School of Dentistry, University of Foggia, 71100 Foggia, Italy
| | - Cristian D’Ovidio
- Section of Legal Medicine, Department of Medicine and Aging Sciences, G. d’Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Gianpaolo Ronconi
- Clinica dei Pazienti del Territorio, Fondazione Policlinico Gemelli, 00185 Rome, Italy
| | | | - Carla E. Gallenga
- Section of Ophthalmology, Department of Biomedical Sciences and Specialist Surgery, University of Ferrara, 44121 Ferrara, Italy
| | - Ilias Frydas
- Department of Parasitology, Aristotle University, 54124 Thessaloniki, Greece
| | - Spyros K. Kritas
- Department of Microbiology and Infectious Diseases, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Macedonia, Greece
| | - Matteo Trimarchi
- Centre of Neuroscience of Milan, Department of Medicine and Surgery, University of Milan, 20122 Milano, Italy
| | - Francesco Carinci
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Pio Conti
- Immunology Division, Postgraduate Medical School, University of Chieti, 66100 Chieti, Italy
- Correspondence:
| |
Collapse
|
48
|
Jammoul M, Naddour J, Madi A, Reslan MA, Hatoum F, Zeineddine J, Abou-Kheir W, Lawand N. Investigating the possible mechanisms of autonomic dysfunction post-COVID-19. Auton Neurosci 2023; 245:103071. [PMID: 36580747 PMCID: PMC9789535 DOI: 10.1016/j.autneu.2022.103071] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 12/10/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022]
Abstract
Patients with long COVID suffer from many neurological manifestations that persist for 3 months following infection by SARS-CoV-2. Autonomic dysfunction (AD) or dysautonomia is one complication of long COVID that causes patients to experience fatigue, dizziness, syncope, dyspnea, orthostatic intolerance, nausea, vomiting, and heart palpitations. The pathophysiology behind AD onset post-COVID is largely unknown. As such, this review aims to highlight the potential mechanisms by which AD occurs in patients with long COVID. The first proposed mechanism includes the direct invasion of the hypothalamus or the medulla by SARS-CoV-2. Entry to these autonomic centers may occur through the neuronal or hematogenous routes. However, evidence so far indicates that neurological manifestations such as AD are caused indirectly. Another mechanism is autoimmunity whereby autoantibodies against different receptors and glycoproteins expressed on cellular membranes are produced. Additionally, persistent inflammation and hypoxia can work separately or together to promote sympathetic overactivation in a bidirectional interaction. Renin-angiotensin system imbalance can also drive AD in long COVID through the downregulation of relevant receptors and formation of autoantibodies. Understanding the pathophysiology of AD post-COVID-19 may help provide early diagnosis and better therapy for patients.
Collapse
Affiliation(s)
- Maya Jammoul
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Lebanon
| | - Judith Naddour
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Lebanon
| | - Amir Madi
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy
| | - Mohammad Amine Reslan
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Firas Hatoum
- Faculty of Medicine, American University of Beirut, Lebanon
| | | | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Lebanon
| | - Nada Lawand
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Lebanon; Department of Neurology, Faculty of Medicine, American University of Beirut, Lebanon.
| |
Collapse
|
49
|
Wang XY, Chen XQ, Wang GQ, Cai RL, Wang H, Wang HT, Peng XQ, Zhang MT, Huang S, Shen GM. A neural circuit for gastric motility disorders driven by gastric dilation in mice. Front Neurosci 2023; 17:1069198. [PMID: 36908796 PMCID: PMC9992744 DOI: 10.3389/fnins.2023.1069198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023] Open
Abstract
INTRODUCTION Symptoms of gastric motility disorders are common clinical manifestations of functional gastrointestinal disorders (FGIDs), and are triggered and exacerbated by stress, but the neural pathways underpinning them remain unclear. METHODS We set-up a mouse model by gastric dilation (GD) in which the gastric dynamics were assessed by installing strain gauges on the surface of the stomach. The neural pathway associated with gastric motility disorders was investigated by behavioral tests, electrophysiology, neural circuit tracing, and optogenetics and chemogenetics involving projections of the corticotropin-releasing hormone (CRH) from the paraventricular nucleus of the hypothalamus (PVN) to acetylcholine (ChAT) neurons in the dorsal motor nucleus of the vagus (DMV). RESULTS We found that GD induced gastric motility disorders were accompanied by activation of PVN CRH neurons, which could be alleviated by strategies that inhibits the activity of PVN CRH neurons. In addition, we identified a neural pathway in which PVN CRH neurons project into DMV ChAT neurons, modulated activity of the PVN CRH →DMV ChAT pathway to alleviate gastric motility disorders induced by GD. DISCUSSION These findings indicate that the PVN CRH →DMV ChAT pathway may mediate at least some aspects of GD related gastric motility, and provide new insights into the mechanisms by which somatic stimulation modulates the physiological functions of internal organs and systems.
Collapse
Affiliation(s)
- Xi-yang Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xiao-qi Chen
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Guo-quan Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Rong-lin Cai
- Research Institute of Acupuncture and Moxibustion, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hao Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hai-tao Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xiao-qi Peng
- School of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Meng-ting Zhang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Shun Huang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Guo-ming Shen
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Institute of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
50
|
Lamptey RNL, Sun C, Layek B, Singh J. Neurogenic Hypertension, the Blood-Brain Barrier, and the Potential Role of Targeted Nanotherapeutics. Int J Mol Sci 2023; 24:2213. [PMID: 36768536 PMCID: PMC9916775 DOI: 10.3390/ijms24032213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023] Open
Abstract
Hypertension is a major health concern globally. Elevated blood pressure, initiated and maintained by the brain, is defined as neurogenic hypertension (NH), which accounts for nearly half of all hypertension cases. A significant increase in angiotensin II-mediated sympathetic nervous system activity within the brain is known to be the key driving force behind NH. Blood pressure control in NH has been demonstrated through intracerebrovascular injection of agents that reduce the sympathetic influence on cardiac functions. However, traditional antihypertensive agents lack effective brain permeation, making NH management extremely challenging. Therefore, developing strategies that allow brain-targeted delivery of antihypertensives at the therapeutic level is crucial. Targeting nanotherapeutics have become popular in delivering therapeutics to hard-to-reach regions of the body, including the brain. Despite the frequent use of nanotherapeutics in other pathological conditions such as cancer, their use in hypertension has received very little attention. This review discusses the underlying pathophysiology and current management strategies for NH, as well as the potential role of targeted therapeutics in improving current treatment strategies.
Collapse
Affiliation(s)
| | | | - Buddhadev Layek
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND 58105, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND 58105, USA
| |
Collapse
|