1
|
Huang S, Shi C, Tao D, Yang C, Luo Y. Modulating reward and aversion: Insights into addiction from the paraventricular nucleus. CNS Neurosci Ther 2024; 30:e70046. [PMID: 39295107 PMCID: PMC11410887 DOI: 10.1111/cns.70046] [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: 06/16/2024] [Revised: 08/14/2024] [Accepted: 08/31/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND Drug addiction, characterized by compulsive drug use and high relapse rates, arises from complex interactions between reward and aversion systems in the brain. The paraventricular nucleus (PVN), located in the anterior hypothalamus, serves as a neuroendocrine center and is a key component of the hypothalamic-pituitary-adrenal axis. OBJECTIVE This review aimed to explore how the PVN impacts reward and aversion in drug addiction through stress responses and emotional regulation and to evaluate the potential of PVN as a therapeutic target for drug addiction. METHODS We review the current literature, focusing on three main neuron types in the PVN-corticotropin-releasing factor, oxytocin, and arginine vasopressin neurons-as well as other related neurons, to understand their roles in modulating addiction. RESULTS Existing studies highlight the PVN as a key mediator in addiction, playing a dual role in reward and aversion systems. These findings are crucial for understanding addiction mechanisms and developing targeted therapies. CONCLUSION The role of PVN in stress response and emotional regulation suggests its potential as a therapeutic target in drug addiction, offering new insights for addiction treatment.
Collapse
Affiliation(s)
- Shihao Huang
- Hunan Province People's HospitalThe First‐Affiliated Hospital of Hunan Normal UniversityChangshaChina
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence ResearchPeking UniversityBeijingChina
- Department of Neurobiology, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
| | - Cuijie Shi
- College of Forensic MedicineHebei Medical UniversityShijiazhuangChina
| | - Dan Tao
- School of MedicineHunan Normal UniversityChangshaChina
| | - Chang Yang
- School of MedicineHunan Normal UniversityChangshaChina
| | - Yixiao Luo
- Hunan Province People's HospitalThe First‐Affiliated Hospital of Hunan Normal UniversityChangshaChina
- Key Laboratory for Birth Defects Research and Prevention of the National Health CommissionHunan Provincial Maternal and Child Health Care HospitalChangshaChina
| |
Collapse
|
2
|
Jiang Z, He M, Young C, Cai J, Xu Y, Jiang Y, Li H, Yang M, Tong Q. Dopaminergic Neurons in Zona Incerta Drives Appetitive Self-Grooming. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308974. [PMID: 39099402 PMCID: PMC11422805 DOI: 10.1002/advs.202308974] [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: 11/21/2023] [Revised: 07/07/2024] [Indexed: 08/06/2024]
Abstract
Dopaminergic (DA) neurons are known to play a key role in controlling behaviors. While DA neurons in other brain regions are extensively characterized, those in zona incerta (ZITH or A13) receive much less attention and their function remains to be defined. Here it is shown that optogenetic stimulation of these neurons elicited intensive self-grooming behaviors and promoted place preference, which can be enhanced by training but cannot be converted into contextual memory. Interestingly, the same stimulation increased DA release to periaqueductal grey (PAG) neurons and local PAG antagonism of DA action reduced the elicited self-grooming. In addition, A13 neurons increased their activity in response to various external stimuli and during natural self-grooming episodes. Finally, monosynaptic retrograde tracing showed that the paraventricular hypothalamus represents one of the major upstream brain regions to A13 neurons. Taken together, these results reveal that A13 neurons are one of the brain sites that promote appetitive self-grooming involving DA release to the PAG.
Collapse
Affiliation(s)
- Zhiying Jiang
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Michelle He
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Summer Undergraduate Research Program, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Sargent College of Health and Rehabilitation Sciences, Boston University, Boston, MA, 02215, USA
| | - Claire Young
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jing Cai
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- MD Anderson Cancer Center & UTHealth Graduate School for Biomedical Sciences, University of Texas Health Science at Houston, Houston, TX, 77030, USA
| | - Yuanzhong Xu
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yanyan Jiang
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Hongli Li
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Maojie Yang
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Qingchun Tong
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- MD Anderson Cancer Center & UTHealth Graduate School for Biomedical Sciences, University of Texas Health Science at Houston, Houston, TX, 77030, USA
| |
Collapse
|
3
|
Ramirez-Plascencia OD, De Luca R, Machado NLS, Eghlidi D, Khanday MA, Bandaru SS, Raffin F, Vujovic N, Arrigoni E, Saper CB. A hypothalamic circuit for circadian regulation of corticosterone secretion. RESEARCH SQUARE 2024:rs.3.rs-4718850. [PMID: 39041039 PMCID: PMC11261983 DOI: 10.21203/rs.3.rs-4718850/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
The secretion of cortisol in humans and corticosterone (Cort) in rodents follows a daily rhythm which is important in readying the individual for the daily active cycle and is impaired in chronic depression. This rhythm is orchestrated by the suprachiasmatic nucleus (SCN) which governs the activity of neurons in the paraventricular nucleus of the hypothalamus that produce the corticotropin-releasing hormone (PVHCRH neurons). The dorsomedial nucleus of the hypothalamus (DMH) serves as a crucial intermediary, being innervated by the SCN both directly and via relays in the subparaventricular zone, and projecting axons to the PVH, thereby exerting influence over the cortisol/corticosterone rhythm. However, the role and synaptic mechanisms by which DMH neurons regulate the daily rhythm of Cort secretion has not been explored. We found that either ablating or acutely inhibiting the DMH glutamatergic (DMHVglut2) neurons resulted in a 40-70% reduction in the daily peak of Cort. Deletion of the Vglut2 gene within the DMH produced a similar effect, highlighting the indispensable role of glutamatergic signaling. Chemogenetic stimulation of DMHVglut2 neurons led to an increase of Cort levels, and optogenetic activation of their terminals in the PVH in hypothalamic slices directly activated PVHCRH neurons through glutamate release (the DMHVglut2 → PVHCRH pathway). Similarly, ablating, inhibiting, or disrupting GABA transmission by DMH GABAergic (DMHVgat) neurons diminished the circadian peak of Cort, particularly under constant darkness conditions. Chemogenetic stimulation of DMHVgat neurons increased Cort, although with a lower magnitude compared to DMHVglut2 neuron stimulation, suggesting a role in disinhibiting PVHCRH neurons. Supporting this hypothesis, we found that rostral DMHVgat neurons project directly to GABAergic neurons in the caudal ventral part of the PVH and adjacent peri-PVH area (cvPVH), which directly inhibit PVHCRH neurons, and that activating the DMHVgat terminals in the cvPVH in brain slices reduced GABAergic afferent input onto the PVHCRH neurons. Finally, ablation of cvPVHVgat neurons resulted in increased Cort release at the onset of the active phase, affirming the pivotal role of the DMHVgat → cvPVHVgat → PVHCRH pathway in Cort secretion. In summary, our study delineates two parallel pathways transmitting temporal information to PVHCRH neurons, collectively orchestrating the daily surge in Cort in anticipation of the active phase. These findings are crucial to understand the neural circuits regulating Cort secretion, shedding light on the mechanisms governing this physiological process and the coordinated interplay between SCN, DMH, and PVH.
Collapse
Affiliation(s)
- Oscar D. Ramirez-Plascencia
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Roberto De Luca
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Natalia L. S. Machado
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Dominique Eghlidi
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Mudasir A. Khanday
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Sathyajit S. Bandaru
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Francesca Raffin
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, PV 27100, Italy
| | - Nina Vujovic
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
- Departments of Medicine and Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Elda Arrigoni
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Clifford B. Saper
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
4
|
Zuloaga DG, Lafrican JJ, Zuloaga KL. Androgen regulation of behavioral stress responses and the hypothalamic-pituitary-adrenal axis. Horm Behav 2024; 162:105528. [PMID: 38503191 PMCID: PMC11144109 DOI: 10.1016/j.yhbeh.2024.105528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/02/2024] [Accepted: 03/06/2024] [Indexed: 03/21/2024]
Abstract
Testosterone is a powerful steroid hormone that can impact the brain and behavior in various ways, including regulating behavioral and neuroendocrine (hypothalamic-pituitary-adrenal (HPA) axis) stress responses. Early in life androgens can act to alter development of brain regions associated with stress regulation, which ultimately impacts the display of stress responses later in life. Adult circulating androgens can also influence the expression of distinct genes and proteins that regulate stress responses. These changes in the brain are hypothesized to underlie the potent effects of androgens in regulating behaviors related to stress and stress-induced activation of the HPA axis. Androgens can induce alterations in these functions through direct binding to the androgen receptor (AR) or following conversion to estrogens and subsequent binding to estrogen receptors including estrogen receptor alpha (ERα), beta (ERβ), and G protein-coupled estrogen receptor 1 (GPER1). In this review, we focus on the role of androgens in regulating behavioral and neuroendocrine stress responses at different stages of the lifespan and the sex hormone receptors involved in regulating these effects. We also review the specific brain regions and cell phenotypes upon which androgens are proposed to act to regulate stress responses with an emphasis on hypothalamic and extended amygdala subregions. This knowledge of androgen effects on these neural systems is critical for understanding how sex hormones regulate stress responses.
Collapse
Affiliation(s)
- Damian G Zuloaga
- Department of Psychology, University at Albany, Albany, NY, USA.
| | | | - Kristen L Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| |
Collapse
|
5
|
Petrucci AN, Jones AR, Kreitlow BL, Buchanan GF. Peri-ictal activation of dorsomedial dorsal raphe serotonin neurons reduces mortality associated with maximal electroshock seizures. Brain Commun 2024; 6:fcae052. [PMID: 38487550 PMCID: PMC10939444 DOI: 10.1093/braincomms/fcae052] [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: 01/23/2023] [Revised: 12/13/2023] [Accepted: 03/08/2024] [Indexed: 03/17/2024] Open
Abstract
Over one-third of patients with epilepsy will develop refractory epilepsy and continue to experience seizures despite medical treatment. These patients are at the greatest risk for sudden unexpected death in epilepsy. The precise mechanisms underlying sudden unexpected death in epilepsy are unknown, but cardiorespiratory dysfunction and arousal impairment have been implicated. Substantial circumstantial evidence suggests serotonin is relevant to sudden unexpected death in epilepsy as it modulates sleep/wake regulation, breathing and arousal. The dorsal raphe nucleus is a major serotonergic center and a component of the ascending arousal system. Seizures disrupt the firing of dorsal raphe neurons, which may contribute to reduced responsiveness. However, the relevance of the dorsal raphe nucleus and its subnuclei to sudden unexpected death in epilepsy remains unclear. The dorsomedial dorsal raphe may be a salient target due to its role in stress and its connections with structures implicated in sudden unexpected death in epilepsy. We hypothesized that optogenetic activation of dorsomedial dorsal raphe serotonin neurons in TPH2-ChR2-YFP (n = 26) mice and wild-type (n = 27) littermates before induction of a maximal electroshock seizure would reduce mortality. In this study, pre-seizure activation of dorsal raphe nucleus serotonin neurons reduced mortality in TPH2-ChR2-YFP mice with implants aimed at the dorsomedial dorsal raphe. These results implicate the dorsomedial dorsal raphe in this novel circuit influencing seizure-induced mortality. It is our hope that these results and future experiments will define circuit mechanisms that could ultimately reduce sudden unexpected death in epilepsy.
Collapse
Affiliation(s)
- Alexandra N Petrucci
- Interdisciplinary Graduate Program in Neuroscience, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Allysa R Jones
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Benjamin L Kreitlow
- Interdisciplinary Graduate Program in Neuroscience, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Gordon F Buchanan
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
6
|
Bowling DL. Biological principles for music and mental health. Transl Psychiatry 2023; 13:374. [PMID: 38049408 PMCID: PMC10695969 DOI: 10.1038/s41398-023-02671-4] [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: 10/20/2022] [Revised: 10/30/2023] [Accepted: 11/17/2023] [Indexed: 12/06/2023] Open
Abstract
Efforts to integrate music into healthcare systems and wellness practices are accelerating but the biological foundations supporting these initiatives remain underappreciated. As a result, music-based interventions are often sidelined in medicine. Here, I bring together advances in music research from neuroscience, psychology, and psychiatry to bridge music's specific foundations in human biology with its specific therapeutic applications. The framework I propose organizes the neurophysiological effects of music around four core elements of human musicality: tonality, rhythm, reward, and sociality. For each, I review key concepts, biological bases, and evidence of clinical benefits. Within this framework, I outline a strategy to increase music's impact on health based on standardizing treatments and their alignment with individual differences in responsivity to these musical elements. I propose that an integrated biological understanding of human musicality-describing each element's functional origins, development, phylogeny, and neural bases-is critical to advancing rational applications of music in mental health and wellness.
Collapse
Affiliation(s)
- Daniel L Bowling
- Department of Psychiatry and Behavioral Sciences, Stanford University, School of Medicine, Stanford, CA, USA.
- Center for Computer Research in Music and Acoustics (CCRMA), Stanford University, School of Humanities and Sciences, Stanford, CA, USA.
| |
Collapse
|
7
|
Yang X, Geng F. Corticotropin-releasing factor signaling and its potential role in the prefrontal cortex-dependent regulation of anxiety. J Neurosci Res 2023; 101:1781-1794. [PMID: 37592912 DOI: 10.1002/jnr.25238] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/08/2023] [Accepted: 08/06/2023] [Indexed: 08/19/2023]
Abstract
A large body of literature has highlighted the significance of the corticotropin-releasing factor (CRF) system in the regulation of neuropsychiatric diseases. Anxiety disorders are among the most common neuropsychiatric disorders. An increasing number of studies have demonstrated that the CRF family mediates and regulates the development and maintenance of anxiety. Thus, the CRF family is considered to be a potential target for the treatment of anxiety disorders. The prefrontal cortex (PFC) plays a role in the occurrence and development of anxiety, and both CRF and CRF-R1 are widely expressed in the PFC. This paper begins by reviewing CRF-related signaling pathways and their different roles in anxiety and related processes. Then, the role of the CRF system in other neuropsychiatric diseases is reviewed and the potential role of PFC CRF signaling in the regulation of anxiety disorders is discussed. Although other signaling pathways are potentially involved in the process of anxiety, CRF in the PFC primarily modulates anxiety disorders through the activation of corticotropin-releasing factor type1 receptors (CRF-R1) and the excitation of the cAMP/PKA signaling pathway. Moreover, the main signaling pathways of CRF involved in sex differentiation in the PFC appear to be different. In summary, this review suggests that the CRF system in the PFC plays a critical role in the occurrence of anxiety. Thus, CRF signaling is of great significance as a potential target for the treatment of stress-related disorders in the future.
Collapse
Affiliation(s)
- Xin Yang
- Department of Physiology, Shantou University Medical College, Shantou, China
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fei Geng
- Department of Physiology, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| |
Collapse
|
8
|
Petrie GN, Balsevich G, Füzesi T, Aukema RJ, Driever WPF, van der Stelt M, Bains JS, Hill MN. Disruption of tonic endocannabinoid signalling triggers cellular, behavioural and neuroendocrine responses consistent with a stress response. Br J Pharmacol 2023; 180:3146-3159. [PMID: 37482931 DOI: 10.1111/bph.16198] [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: 01/31/2023] [Revised: 06/11/2023] [Accepted: 07/03/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND AND PURPOSE Endocannabinoid (eCB) signalling gates many aspects of the stress response, including the hypothalamic-pituitary-adrenal (HPA) axis. The HPA axis is controlled by corticotropin releasing hormone (CRH) producing neurons in the paraventricular nucleus of the hypothalamus (PVN). Disruption of eCB signalling increases drive to the HPA axis, but the mechanisms subserving this process are poorly understood. EXPERIMENTAL APPROACH Using an array of cellular, endocrine and behavioural readouts associated with activation of CRH neurons in the PVN, we evaluated the contributions of tonic eCB signalling to the generation of a stress response. KEY RESULTS The CB1 receptor antagonist/inverse agonist AM251, neutral antagonist NESS243 and NAPE PLD inhibitor LEI401 all uniformly increased Fos in the PVN, unmasked stress-linked behaviours, such as grooming, and increased circulating CORT, recapitulating the effects of stress. Similar effects were also seen after direct administration of AM251 into the PVN, while optogenetic inhibition of PVN CRH neurons ameliorated stress-like behavioural changes produced by disruption of eCB signalling. CONCLUSIONS AND IMPLICATIONS These data indicate that under resting conditions, constitutive eCB signalling restricts activation of the HPA axis through local regulation of CRH neurons in the PVN.
Collapse
Affiliation(s)
- Gavin N Petrie
- Neuroscience Program, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada
| | - Georgia Balsevich
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada
| | - Tamás Füzesi
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Robert J Aukema
- Neuroscience Program, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada
| | - Wouter P F Driever
- Department of Molecular Physiology, LIC, Leiden University, Leiden, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Mario van der Stelt
- Department of Molecular Physiology, LIC, Leiden University, Leiden, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Jaideep S Bains
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Department of Psychiatry, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
9
|
Rybka KA, Lafrican JJ, Rosinger ZJ, Ariyibi DO, Brooks MR, Jacobskind JS, Zuloaga DG. Sex differences in androgen receptor, estrogen receptor alpha, and c-Fos co-expression with corticotropin releasing factor expressing neurons in restrained adult mice. Horm Behav 2023; 156:105448. [PMID: 38344954 PMCID: PMC10861933 DOI: 10.1016/j.yhbeh.2023.105448] [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: 08/24/2023] [Revised: 10/03/2023] [Accepted: 10/23/2023] [Indexed: 02/15/2024]
Abstract
Gonadal hormone actions through androgen receptor (AR) and estrogen receptor alpha (ERα) regulate sex differences in hypothalamic-pituitary-adrenal (HPA) axis responsivity and stress-related behaviors. Here we tested whether corticotropin releasing factor (CRF) expressing neurons, which are widely known to regulate neuroendocrine and behavioral stress responses, co-express AR and ERα as a potential mechanism for gonadal hormone regulation of these responses. Using Crh-IRES-Cre::Ai9 reporter mice we report high co-localization of AR in CRF neurons within the medial preoptic area (MPOA), bed nucleus of the stria terminalis (BST), medial amygdala (MeA), and ventromedial hypothalamus (VMH), moderate levels within the central amygdala (CeA) and low levels in the paraventricular hypothalamus (PVN). Sex differences in CRF/AR co-expression were found in the principal nucleus of the BST (BSTmpl), CeA, MeA, and VMH (males>females). CRF co-localization with ERα was generally lower relative to AR co-localization. However, high co-expression was found within the MPOA, AVPV, and VMH, with moderate co-expression in the arcuate nucleus (ARC), BST, and MeA and low levels in the PVN and CeA. Sex differences in CRF/ERα co-localization were found in the BSTmpl and PVN (males>females). Finally, we assessed neural activation of CRF neurons in restraint-stressed mice and found greater CRF/c-Fos co-expression in females in the BSTmpl and periaqueductal gray, while co-expression was higher in males within the ARC and dorsal CA1. Given the known role of CRF in regulating behavioral stress responses and the HPA axis, AR/ERα co-expression and sex-specific activation of CRF cell groups indicate potential mechanisms for modulating sex differences in these functions.
Collapse
Affiliation(s)
- Krystyna A Rybka
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States of America
| | - Jennifer J Lafrican
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States of America
| | - Zachary J Rosinger
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States of America
| | - Deborah O Ariyibi
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States of America
| | - Mecca R Brooks
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States of America
| | - Jason S Jacobskind
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States of America
| | - Damian G Zuloaga
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States of America.
| |
Collapse
|
10
|
Gao F, Yuan WH, Wu SB, Wang ZB, Zhu GQ, Zhou MQ. Electroacupuncture in the treatment of IBS in rats: investigation of the mechanisms of CRH + neurons in the paraventricular nucleus. J Neurophysiol 2023; 130:380-391. [PMID: 37435647 PMCID: PMC10625839 DOI: 10.1152/jn.00156.2023] [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: 04/18/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/13/2023] Open
Abstract
Electroacupuncture (EA) is well documented to treat irritable bowel syndrome (IBS). However, the mechanism of the central nervous system related to IBS and acupuncture stimulation is still not well known. In this study, a rat model of IBS was established by cold-restraint comprehensive stresses for 15 days, and it was found that the levels of corticotropin-releasing hormone (CRH), corticosterone (CORT), and adrenocorticotropic hormone (ACTH) in the peripheral serum were increased; the visceral sensitivity was enhanced; and the intestinal motility was accelerated, specifically, there was an enhancement in the discharge frequency of neurons in the paraventricular nucleus (PVN). EA treatment for 3 days, 20 min/day, alleviated the increase in the levels of CRH, CORT, and ACTH in the peripheral serum of rats, reduced the visceral sensitivity of IBS rats, and inhibited colon movement and discharge frequency of the neurons in the PVN. In addition, EA could reduce the excitability of CRH neurons and the expression of corticotropin-releasing hormone receptor 1 (CRHR1) and corticotropin-releasing hormone receptor 2 (CRHR2) in PVN. At the same time, the expression of CRH, CRHR1, and CRHR2 in the peripheral colon was decreased. Taken together, EA appears to regulate intestinal functional activity through the central CRH nervous system, revealing the central regulation mechanism of EA in IBS rats, and providing a scientific research basis for the correlation among the meridians, viscera, and brain.NEW & NOTEWORTHY The purpose of this research was to determine the central regulatory mechanism of electroacupuncture (EA) in rats with irritable bowel syndrome (IBS). Our results showed that combined with the serum changes in corticotropin-releasing hormone (CRH), corticosterone (CORT), and adrenocorticotropic hormone (ACTH), the improvement of IBS by EA was related to them. Furthermore, EA could regulate intestinal functional activity through the central CRH+ nervous system.
Collapse
Affiliation(s)
- Fang Gao
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Wei-Hua Yuan
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Sheng-Bing Wu
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | | | - Guo-Qi Zhu
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Mei-Qi Zhou
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
11
|
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
|
12
|
Ma H, Cui Z, Guo X, Zhao Q, Zhang Y, Guan Y, Yang P, Zhu H, Wang S, Zhang X, Zhang Y, Pan HL, Ma H. Corticotropin-releasing factor potentiates glutamatergic input and excitability of presympathetic neurons in the hypothalamus in spontaneously hypertensive rats. Neuropharmacology 2023; 230:109506. [PMID: 36924924 DOI: 10.1016/j.neuropharm.2023.109506] [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: 11/16/2022] [Revised: 02/28/2023] [Accepted: 03/10/2023] [Indexed: 03/15/2023]
Abstract
Hyperactivity of presympathetic neurons in the hypothalamic paraventricular nucleus (PVN) plays a key role in generating excess sympathetic output in hypertension. However, the mechanisms driving hyperactivity of PVN presympathetic neurons in hypertension are unclear. In this study, we determined the role of corticotropin-releasing factor (CRF) in the PVN in augmented glutamatergic input, neuronal excitability and sympathetic outflow in hypertension. The number of CRF or c-Fos immunoreactive neurons and CRF/c-Fos double-labeled neurons in the PVN was significantly greater in spontaneously hypertensive rats (SHRs) than in normotensive Wistar-Kyoto (WKY) rats. Blocking glutamatergic input reduced the CRF-potentiated excitability of spinally projecting PVN neurons. Furthermore, CRF knockdown via Crispr/Cas9 in the PVN decreased the frequencies of spontaneous firing and miniature excitatory postsynaptic currents (mEPSCs) in spinally projecting PVN neurons in SHRs. In addition, the mRNA and protein levels of CRFR1, but not CRFR2, in the PVN were significantly higher in SHRs than in WKY rats. Blocking CRFR1 with NBI-35965, but not blocking CRFR2 with Antisauvagine-30, reduced the frequencies of spontaneous firing and mEPSCs of spinally projecting PVN neurons in SHRs. Also, microinjection of NBI-35965 into the PVN significantly reduced arterial blood pressure (ABP) and renal sympathetic nerve activity (RSNA) in anesthetized SHRs, but not in WKY rats. However, microinjection of Antisauvagine-30 into the PVN had no effect on ABP or RSNA in WKY rats and SHRs. Our findings suggest that endogenous CRF in the PVN potentiates glutamatergic input and firing activity of PVN presympathetic neurons via CRFR1, resulting in augmented sympathetic outflow in hypertension.
Collapse
Affiliation(s)
- Hongyu Ma
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Ziye Cui
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Xinqi Guo
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Qiyue Zhao
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Ying Zhang
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Yue Guan
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China; Key Laboratory of Neurophysiology of Hebei Province, Shijiazhuang, 050017, Hebei, China
| | - Peiyun Yang
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Huaibing Zhu
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Sheng Wang
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China; Key Laboratory of Neurophysiology of Hebei Province, Shijiazhuang, 050017, Hebei, China
| | - Xiangjian Zhang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yi Zhang
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Hui-Lin Pan
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Huijie Ma
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China; Key Laboratory of Neurophysiology of Hebei Province, Shijiazhuang, 050017, Hebei, China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China.
| |
Collapse
|
13
|
Xu Y, Jiang Z, Li H, Cai J, Jiang Y, Otiz-Guzman J, Xu Y, Arenkiel BR, Tong Q. Lateral septum as a melanocortin downstream site in obesity development. Cell Rep 2023; 42:112502. [PMID: 37171957 DOI: 10.1016/j.celrep.2023.112502] [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: 11/08/2022] [Revised: 03/27/2023] [Accepted: 04/26/2023] [Indexed: 05/14/2023] Open
Abstract
The melanocortin pathway is well established to be critical for body-weight regulation in both rodents and humans. Despite extensive studies focusing on this pathway, the downstream brain sites that mediate its action are not clear. Here, we found that, among the known paraventricular hypothalamic (PVH) neuron groups, those expressing melanocortin receptors 4 (PVHMc4R) preferably project to the ventral part of the lateral septum (LSv), a brain region known to be involved in emotional behaviors. Photostimulation of PVHMc4R neuron terminals in the LSv reduces feeding and causes aversion, whereas deletion of Mc4Rs or disruption of glutamate release from LSv-projecting PVH neurons causes obesity. In addition, disruption of AMPA receptor function in PVH-projected LSv neurons causes obesity. Importantly, chronic inhibition of PVH- or PVHMc4R-projected LSv neurons causes obesity associated with reduced energy expenditure. Thus, the LSv functions as an important node in mediating melanocortin action on body-weight regulation.
Collapse
Affiliation(s)
- Yuanzhong Xu
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - Zhiying Jiang
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hongli Li
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jing Cai
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center & UTHealth Houston Graduate School for Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yanyan Jiang
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Joshua Otiz-Guzman
- Department of Molecular and Human Genetics and Department of Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics and Department of Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Qingchun Tong
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center & UTHealth Houston Graduate School for Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Neurobiology and Anatomy of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
14
|
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: 3] [Impact Index Per Article: 3.0] [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
|
15
|
De Guzman RM, Rosinger ZJ, Rybka KA, Jacobskind JS, Thrasher CA, Caballero AL, Sturm KL, Sharif MS, Abbas MS, Parra KE, Zuloaga KL, Justice NJ, Zuloaga DG. Changes in Corticotropin-Releasing Factor Receptor Type 1, Co-Expression with Tyrosine Hydroxylase and Oxytocin Neurons, and Anxiety-Like Behaviors across the Postpartum Period in Mice. Neuroendocrinology 2023; 113:795-810. [PMID: 36917957 PMCID: PMC10389801 DOI: 10.1159/000530156] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/07/2023] [Indexed: 03/15/2023]
Abstract
INTRODUCTION Corticotropin-releasing factor and its primary receptor (CRFR1) are critical regulators of behavioral and neuroendocrine stress responses. CRFR1 has also been associated with stress-related behavioral changes in postpartum mice. Our previous studies indicate dynamic changes in CRFR1 levels and coupling of CRFR1 with tyrosine hydroxylase (TH) and oxytocin (OT) neurons in postpartum mice. In this study, we aimed to determine the time course of these changes during the postpartum period. METHODS Using a CRFR1-GFP reporter mouse line, we compared postpartum mice at five time points with nulliparous mice. We performed immunohistochemistry to assess changes in CRFR1 levels and changes in co-expression of TH/CRFR1-GFP and OT/CRFR1-GFP across the postpartum period. Mice were also assessed for behavioral stress responses in the open field test. RESULTS Relative to nulliparous mice, CRFR1 levels were elevated in the anteroventral periventricular nucleus (AVPV/PeN) but were decreased in the medial preoptic area from postpartum day 1 (P1) through P28. In the paraventricular hypothalamus (PVN), there is a transient decline in CRFR1 mid-postpartum with a nadir at P7. Co-localization of CRFR1 with TH-expressing neurons was also altered with a transient decrease found in the AVPV/PeN at P7 and P14. Co-expression of CRFR1 and OT neurons of the PVN and supraoptic nucleus was dramatically altered with virtually no co-expression found in nulliparous mice, but levels increased shortly after parturition and peaked near P21. A transient decrease in open field center time was found at P7, indicating elevated anxiety-like behavior. CONCLUSION This study revealed various changes in CRFR1 across the postpartum period, which may contribute to stress-related behavior changes in postpartum mice.
Collapse
Affiliation(s)
| | | | | | | | - Christina A. Thrasher
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | | | | | | | | | | | - Kristen L. Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Nicholas J. Justice
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine of McGovern Medical School, University of Texas Health Sciences Center, Houston, TX, USA
| | | |
Collapse
|
16
|
Wu CY, Chen HH, Tao PL, Yuan ZF. Comparisons of stress-related neuronal activation induced by restraint in adult male rat offspring with prenatal exposure to buprenorphine, methadone, or morphine. CHINESE J PHYSIOL 2023; 66:65-72. [PMID: 37082994 DOI: 10.4103/cjop.cjop-d-23-00015] [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: 04/22/2023] Open
Abstract
Prenatal opioid exposure may impede the development of adaptive responses to environmental stimuli by altering the stress-sensitive brain circuitry located at the paraventricular nucleus of the hypothalamus (PVH) and locus coeruleus (LC). Corticotropin-releasing factor (CRF) released from neurons in the PVH has emerged as a key molecule to initiate and integrate the stress response. Methadone (Meth) and buprenorphine (Bu) are two major types of synthetic opioid agonists for first-line medication-assisted treatment of opioid (e.g., morphine, Mor) use disorder in pregnant women. No studies have compared the detrimental effects of prenatal exposure to Meth versus Bu on the stress response of their offspring upon reaching adulthood. In this study, we aimed to compare stress-related neuronal activation in the PVH and LC induced by restraint (RST) stress in adult male rat offspring with prenatal exposure to the vehicle (Veh), Bu, Meth, or Mor. CFos-immunoreactive cells were used as an indicator for neuronal activation. We found that RST induced less neuronal activation in the Meth or Mor exposure groups compared with that in the Bu or Veh groups; no significant difference was detected between the Bu and Veh exposure groups. RST-induced neuronal activation was completely prevented by central administration of a CRF receptor antagonist (α-helical CRF9-41, 10 μg/3 μL) in all exposure groups, suggesting the crucial role of CRF in this stress response. In offspring without RST, central administration of CRF (0.5 μg/3 μL)-induced neuronal activation in the PVH and LC. CRF-induced neuronal activation was lessened in the Meth or Mor exposure groups compared with that in the Bu or Veh groups; no significant difference was detected between the Bu and Veh exposure groups. Moreover, RST- or CRF-induced neuronal activation in the Meth exposure group was comparable with that in the Mor exposure group. Further immunohistochemical analysis revealed that the Meth and Mor exposure groups displayed less CRF neurons in the PVH of offspring with or without RST compared with the Bu or Veh groups. Thus, stress-induced neuronal activation in the PVH and LC was well preserved in adult male rat offspring with prenatal exposure to Bu, but it was substantially lessened in those with prenatal exposure to Meth or Mor. Lowered neuronal activation found in the Meth or Mor exposure groups may be, at least in part, due to the reduction in the density of CRF neurons in the PVH.
Collapse
Affiliation(s)
- Chia-Yen Wu
- Department of Physiology, Tzu Chi University, Hualien, Taiwan
| | - Hwei-Hsien Chen
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Pao-Luh Tao
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Zung Fan Yuan
- Department of Physiology, Tzu Chi University; Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
17
|
Baskerville R, McGrath T, Castell L. The effects of physical activity on glutamate neurotransmission in neuropsychiatric disorders. Front Sports Act Living 2023; 5:1147384. [PMID: 36949894 PMCID: PMC10025343 DOI: 10.3389/fspor.2023.1147384] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/17/2023] [Indexed: 03/08/2023] Open
Abstract
Physical activity (PA) is an effective way of increasing cognitive and emotional health and counteracting many psychiatric conditions. Numerous neurobiological models for depression have emerged in the past 30 years but many struggle to incorporate the effects of exercise. The hippocampus and pre-frontal cortex (PFC) containing predominantly glutamate neurotransmission, are the centres of changes seen in depression. There is therefore increasing interest in glutamatergic systems which offers new paradigms of understanding mechanisms connecting physical activity, stress, inflammation and depression, not explained by the serotonin theories of depression. Similar hippocampal glutamate dysfunction is observed in many other neuropsychiatric conditions. Excitatory glutamate neurones have high functionality, but also high ATP requirements and are therefore vulnerable to glucocorticoid or pro-inflammatory stress that causes mitochondrial dysfunction, with synaptic loss, culminating in depressed mood and cognition. Exercise improves mitochondrial function, angiogenesis and synaptogenesis. Within the glutamate hypothesis of depression, the mechanisms of stress and inflammation have been extensively researched, but PA as a mitigator is less understood. This review examines the glutamatergic mechanisms underlying depression and the evidence of physical activity interventions within this framework. A dynamic glutamate-based homeostatic model is suggested whereby stress, neuroinflammation and PA form counterbalancing influences on hippocampal cell functionality, which manifests as depression and other neuropsychiatric conditions when homeostasis is disrupted.
Collapse
Affiliation(s)
- Richard Baskerville
- Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, United Kingdom
- Correspondence: Richard Baskerville
| | | | | |
Collapse
|
18
|
Walker JJ, Romanò N. Fast dynamics in the HPA axis: Insight from mathematical and experimental studies. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 27:100403. [PMID: 36632146 PMCID: PMC9823091 DOI: 10.1016/j.coemr.2022.100403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The activity of the hypothalamic-pituitary-adrenal (HPA) axis is characterised by complex dynamics spanning several timescales. This ranges from slow circadian rhythms in blood hormone concentration to faster ultradian pulses of hormone secretion and even more rapid oscillations in electrical and calcium activity in neuroendocrine cells of the hypothalamus and pituitary gland. Here, we focus on the system's oscillations on the short timescale. We highlight some of the mathematical modelling and experimental work that has been carried out to characterise the mechanisms regulating this highly dynamic mode of neuroendocrine signalling and discuss some future directions that may be explored to enhance understanding of HPA function.
Collapse
Affiliation(s)
- Jamie J. Walker
- Department of Mathematics and Statistics, Faculty of Environment, Science and Economy, University of Exeter, UK,Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, UK,Corresponding author: Walker, Jamie J
| | - Nicola Romanò
- Centre for Discovery Brain Sciences, University of Edinburgh, UK,Corresponding author: Romanò, Nicola twitter icon
| |
Collapse
|
19
|
Ugartemendia L, De Guzman RM, Cai J, Rajamanickam S, Jiang Z, Tao J, Zuloaga DG, Justice NJ. A subpopulation of oxytocin neurons initiate expression of CRF receptor 1 (CRFR1) in females post parturition. Psychoneuroendocrinology 2022; 145:105918. [PMID: 36116320 PMCID: PMC9881188 DOI: 10.1016/j.psyneuen.2022.105918] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/02/2022] [Accepted: 09/04/2022] [Indexed: 02/06/2023]
Abstract
Oxytocin (OT) is essential for successful reproduction, particularly during parturition and lactation. During the postpartum period, OT also influences maternal behavior to promote bonding between mothers and their newborns, and increases stress resilience. However, the mechanism by which stress influences OT neuron activity and OT release has remained unclear. Here, we provide evidence that a subpopulation of OT neurons initiate expression of the receptor for the stress neuropeptide Corticotropin Releasing Factor (CRF), CRFR1, in reproductive females. OT neuron expression of CRFR1 begins at the first parturition and increases during the postpartum period until weaning. The percentage of OT neurons that express CRFR1 increases with successive breeding cycles until it reaches a plateau of 20-25% of OT neurons. OT neuron expression of CRFR1 in reproductive females is maintained after they are no longer actively breeding. CRFR1 expression leads to activation of OT neurons when animals are stressed. We propose a model in which direct CRF signaling to OT neurons selectively in reproductive females potentiates OT release to promote stress resilience in mothers.
Collapse
Affiliation(s)
- Lierni Ugartemendia
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Sciences Center, Houston, TX 77030, United States
| | - Rose M De Guzman
- Department of Psychology, University at Albany, Albany, NY 12222, United States
| | - Jing Cai
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Sciences Center, Houston, TX 77030, United States
| | - Shivakumar Rajamanickam
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Sciences Center, Houston, TX 77030, United States
| | - Zhiying Jiang
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Sciences Center, Houston, TX 77030, United States
| | - Jonathan Tao
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Sciences Center, Houston, TX 77030, United States
| | - Damian G Zuloaga
- Department of Psychology, University at Albany, Albany, NY 12222, United States.
| | - Nicholas J Justice
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Sciences Center, Houston, TX 77030, United States.
| |
Collapse
|
20
|
Maita I, Roepke TA, Samuels BA. Chronic stress-induced synaptic changes to corticotropin-releasing factor-signaling in the bed nucleus of the stria terminalis. Front Behav Neurosci 2022; 16:903782. [PMID: 35983475 PMCID: PMC9378865 DOI: 10.3389/fnbeh.2022.903782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/05/2022] [Indexed: 11/22/2022] Open
Abstract
The sexually dimorphic bed nucleus of the stria terminalis (BNST) is comprised of several distinct regions, some of which act as a hub for stress-induced changes in neural circuitry and behavior. In rodents, the anterodorsal BNST is especially affected by chronic exposure to stress, which results in alterations to the corticotropin-releasing factor (CRF)-signaling pathway, including CRF receptors and upstream regulators. Stress increases cellular excitability in BNST CRF+ neurons by potentiating miniature excitatory postsynaptic current (mEPSC) amplitude, altering the resting membrane potential, and diminishing M-currents (a voltage-gated K+ current that stabilizes membrane potential). Rodent anterodorsal and anterolateral BNST neurons are also critical regulators of behavior, including avoidance of aversive contexts and fear learning (especially that of sustained threats). These rodent behaviors are historically associated with anxiety. Furthermore, BNST is implicated in stress-related mood disorders, including anxiety and Post-Traumatic Stress Disorders in humans, and may be linked to sex differences found in mood disorders.
Collapse
Affiliation(s)
- Isabella Maita
- Samuels Laboratory, Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States,Neuroscience Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Troy A. Roepke
- Roepke Laboratory, Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Benjamin A. Samuels
- Samuels Laboratory, Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States,*Correspondence: Benjamin A. Samuels,
| |
Collapse
|
21
|
WNT/β-catenin Pathway: a Possible Link Between Hypertension and Alzheimer's Disease. Curr Hypertens Rep 2022; 24:465-475. [PMID: 35788966 DOI: 10.1007/s11906-022-01209-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW Recent research has shown that older people with high blood pressure (BP), or hypertension, are more likely to have biomarkers of Alzheimer's disease (AD). Essential hypertension represents the most common cardiovascular disease worldwide and is thought to be responsible for about 13% of all deaths. People with essential hypertension who regularly take prescribed BP medications are half as likely to develop AD as those who do not take them. What then is the connection? RECENT FINDINGS We know that high BP can damage small blood vessels in the brain, affecting those parts that are responsible for memory and thinking. However, the link between AD and hypertension remains unclear. Recent advances in the field of molecular and cellular biology have revealed a downregulation of the canonical WNT/β-catenin pathway in both hypertension and AD. In AD, the glutamate transport function is decreased, a decrease that is associated with a loss of synapse and neuronal death. β-catenin signaling appears to act as a major regulator of glutamate transporters (EAAT and GS) expression and can be harnessed to remove excess glutamate in AD. This review focuses on the possible link between hypertension and AD through the decreased WNT/β-catenin which interacts with the glutamatergic pathway.
Collapse
|
22
|
Ichiyama A, Mestern S, Benigno GB, Scott KE, Allman BL, Muller L, Inoue W. State-dependent activity dynamics of hypothalamic stress effector neurons. eLife 2022; 11:76832. [PMID: 35770968 PMCID: PMC9278954 DOI: 10.7554/elife.76832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 06/17/2022] [Indexed: 11/30/2022] Open
Abstract
The stress response necessitates an immediate boost in vital physiological functions from their homeostatic operation to an elevated emergency response. However, the neural mechanisms underlying this state-dependent change remain largely unknown. Using a combination of in vivo and ex vivo electrophysiology with computational modeling, we report that corticotropin releasing hormone (CRH) neurons in the paraventricular nucleus of the hypothalamus (PVN), the effector neurons of hormonal stress response, rapidly transition between distinct activity states through recurrent inhibition. Specifically, in vivo optrode recording shows that under non-stress conditions, CRHPVN neurons often fire with rhythmic brief bursts (RB), which, somewhat counterintuitively, constrains firing rate due to long (~2 s) interburst intervals. Stressful stimuli rapidly switch RB to continuous single spiking (SS), permitting a large increase in firing rate. A spiking network model shows that recurrent inhibition can control this activity-state switch, and more broadly the gain of spiking responses to excitatory inputs. In biological CRHPVN neurons ex vivo, the injection of whole-cell currents derived from our computational model recreates the in vivo-like switch between RB and SS, providing direct evidence that physiologically relevant network inputs enable state-dependent computation in single neurons. Together, we present a novel mechanism for state-dependent activity dynamics in CRHPVN neurons.
Collapse
|
23
|
Shao YQ, Fan L, Wu WY, Zhu YJ, Xu HT. A developmental switch between electrical and neuropeptide communication in the ventromedial hypothalamus. Curr Biol 2022; 32:3137-3145.e3. [PMID: 35659861 DOI: 10.1016/j.cub.2022.05.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/15/2022] [Accepted: 05/11/2022] [Indexed: 12/29/2022]
Abstract
Dissecting neural connectivity patterns within local brain regions is an essential step to understanding the function of the brain.1 Neural microcircuits in brain regions, such as the neocortex and the hippocampus, have been extensively studied.2 By contrast, the microcircuit in the hypothalamus remains largely uncharacterized. The hypothalamus is crucial for animals' survival and reproduction.3 Knowledge of how different hypothalamic nuclei coordinate with each other and outside brain regions for hypothalamus-related functions has been significantly advanced.4-9 Although there are limited studies on the neural microcircuit in the lateral hypothalamus (LHA)10,11 and the suprachiasmatic nucleus (SCN),12,13 the patterns of neural microcircuits in most of the given hypothalamic nuclei remain largely unknown. This study applied combinatory approaches to address the local neural circuit pattern in the ventromedial hypothalamus (VMH) and other hypothalamic nuclei. We discovered a unique neural circuit design in the VMH. Neurons in the VMH were electrically coupled at the early postnatal stage like ones in the neocortex.14 However, unlike neocortical neurons,14,15 they developed very few chemical synapses after the disappearance of electrical synapses. Instead, VMH neurons communicated with neuropeptides. The similar scarceness of synaptic connectivity found in other hypothalamic nuclei further indicated that the lack of synaptic connections is a unique feature for local neural circuits in most adult hypothalamic nuclei. Thus, our findings provide a solid synaptic basis at the cellular level to understand hypothalamic functions better.
Collapse
Affiliation(s)
- Yin-Qi Shao
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liu Fan
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wen-Yan Wu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi-Jun Zhu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hua-Tai Xu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China.
| |
Collapse
|
24
|
Weera MM, Agoglia AE, Douglass E, Jiang Z, Rajamanickam S, Shackett RS, Herman MA, Justice NJ, Gilpin NW. Generation of a CRF 1-Cre transgenic rat and the role of central amygdala CRF 1 cells in nociception and anxiety-like behavior. eLife 2022; 11:e67822. [PMID: 35389341 PMCID: PMC9033268 DOI: 10.7554/elife.67822] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
Corticotropin-releasing factor type-1 (CRF1) receptors are critical to stress responses because they allow neurons to respond to CRF released in response to stress. Our understanding of the role of CRF1-expressing neurons in CRF-mediated behaviors has been largely limited to mouse experiments due to the lack of genetic tools available to selectively visualize and manipulate CRF1+ cells in rats. Here, we describe the generation and validation of a transgenic CRF1-Cre-tdTomato rat. We report that Crhr1 and Cre mRNA expression are highly colocalized in both the central amygdala (CeA), composed of mostly GABAergic neurons, and in the basolateral amygdala (BLA), composed of mostly glutamatergic neurons. In the CeA, membrane properties, inhibitory synaptic transmission, and responses to CRF bath application in tdTomato+ neurons are similar to those previously reported in GFP+ cells in CRFR1-GFP mice. We show that stimulatory DREADD receptors can be targeted to CeA CRF1+ cells via virally delivered Cre-dependent transgenes, that transfected Cre/tdTomato+ cells are activated by clozapine-n-oxide in vitro and in vivo, and that activation of these cells in vivo increases anxiety-like and nocifensive behaviors. Outside the amygdala, we show that Cre-tdTomato is expressed in several brain areas across the brain, and that the expression pattern of Cre-tdTomato cells is similar to the known expression pattern of CRF1 cells. Given the accuracy of expression in the CRF1-Cre rat, modern genetic techniques used to investigate the anatomy, physiology, and behavioral function of CRF1+ neurons can now be performed in assays that require the use of rats as the model organism.
Collapse
Affiliation(s)
- Marcus M Weera
- Department of Physiology, Louisiana State University Health Sciences CenterNew OrleansUnited States
| | - Abigail E Agoglia
- Department of Pharmacology, University of North CarolinaChapel HillUnited States
| | - Eliza Douglass
- Department of Pharmacology, University of North CarolinaChapel HillUnited States
| | - Zhiying Jiang
- Institute of Molecular Medicine, University of Texas Health Sciences CenterHoustonUnited States
| | - Shivakumar Rajamanickam
- Institute of Molecular Medicine, University of Texas Health Sciences CenterHoustonUnited States
| | - Rosetta S Shackett
- Department of Physiology, Louisiana State University Health Sciences CenterNew OrleansUnited States
| | - Melissa A Herman
- Department of Pharmacology, University of North CarolinaChapel HillUnited States
- Bowles Center for Alcohol Studies, University of North CarolinaChapel HillUnited States
| | - Nicholas J Justice
- Institute of Molecular Medicine, University of Texas Health Sciences CenterHoustonUnited States
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UT HealthHoustonUnited States
| | - Nicholas W Gilpin
- Department of Physiology, Louisiana State University Health Sciences CenterNew OrleansUnited States
- Neuroscience Center of Excellence, Louisiana State University Health Sciences CenterNew OrleansUnited States
- Alcohol & Drug Abuse Center of Excellence, Louisiana State University Health Sciences CenterNew OrleansUnited States
- Southeast Louisiana VA Healthcare System (SLVHCS)New OrleansUnited States
| |
Collapse
|
25
|
Rybka KA, Sturm KL, De Guzman RM, Bah S, Jacobskind JS, Rosinger ZJ, Taroc EZM, Forni PE, Zuloaga DG. Androgen regulation of corticotropin releasing factor receptor 1 in the mouse brain. Neuroscience 2022; 491:185-199. [DOI: 10.1016/j.neuroscience.2022.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 12/19/2022]
|
26
|
McGrath T, Baskerville R, Rogero M, Castell L. Emerging Evidence for the Widespread Role of Glutamatergic Dysfunction in Neuropsychiatric Diseases. Nutrients 2022; 14:nu14050917. [PMID: 35267893 PMCID: PMC8912368 DOI: 10.3390/nu14050917] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/06/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
The monoamine model of depression has long formed the basis of drug development but fails to explain treatment resistance or associations with stress or inflammation. Recent animal research, clinical trials of ketamine (a glutamate receptor antagonist), neuroimaging research, and microbiome studies provide increasing evidence of glutamatergic dysfunction in depression and other disorders. Glutamatergic involvement across diverse neuropathologies including psychoses, neurodevelopmental, neurodegenerative conditions, and brain injury forms the rationale for this review. Glutamate is the brain's principal excitatory neurotransmitter (NT), a metabolic and synthesis substrate, and an immune mediator. These overlapping roles and multiple glutamate NT receptor types complicate research into glutamate neurotransmission. The glutamate microcircuit comprises excitatory glutamatergic neurons, astrocytes controlling synaptic space levels, through glutamate reuptake, and inhibitory GABA interneurons. Astroglia generate and respond to inflammatory mediators. Glutamatergic microcircuits also act at the brain/body interface via the microbiome, kynurenine pathway, and hypothalamus-pituitary-adrenal axis. Disruption of excitatory/inhibitory homeostasis causing neuro-excitotoxicity, with neuronal impairment, causes depression and cognition symptoms via limbic and prefrontal regions, respectively. Persistent dysfunction reduces neuronal plasticity and growth causing neuronal death and tissue atrophy in neurodegenerative diseases. A conceptual overview of brain glutamatergic activity and peripheral interfacing is presented, including the common mechanisms that diverse diseases share when glutamate homeostasis is disrupted.
Collapse
Affiliation(s)
- Thomas McGrath
- Green Templeton College, University of Oxford, Oxford OX2 6HG, UK; (T.M.); (L.C.)
| | - Richard Baskerville
- Faculty of Health and Life Sciences, Oxford Brookes University, Oxford OX3 0BP, UK
- Correspondence:
| | - Marcelo Rogero
- School of Public Health, University of Sao Paulo, Sao Paulo 01246-904, Brazil;
| | - Linda Castell
- Green Templeton College, University of Oxford, Oxford OX2 6HG, UK; (T.M.); (L.C.)
| |
Collapse
|
27
|
Almeida MM, Dias-Rocha CP, Calviño C, Trevenzoli IH. Lipid endocannabinoids in energy metabolism, stress and developmental programming. Mol Cell Endocrinol 2022; 542:111522. [PMID: 34843899 DOI: 10.1016/j.mce.2021.111522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 11/09/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022]
Abstract
The endocannabinoid system (ECS) regulates brain development and function, energy metabolism and stress in a sex-, age- and tissue-dependent manner. The ECS comprises mainly the bioactive lipid ligands anandamide (AEA) and 2-aracdonoylglycerol (2-AG), cannabinoid receptors 1 and 2 (CB1 and CB2), and several metabolizing enzymes. The endocannabinoid tonus is increased in obesity, stimulating food intake and a preference for fat, reward, and lipid accumulation in peripheral tissues, as well as favoring a positive energy balance. Energy balance and stress responses share adaptive mechanisms regulated by the ECS that seem to underlie the complex relationship between feeding and emotional behavior. The ECS is also a key regulator of development. Environmental insults (diet, toxicants, and stress) in critical periods of developmental plasticity, such as gestation, lactation and adolescence, alter the ECS and may predispose individuals to the development of chronic diseases and behavioral changes in the long term. This review is focused on the ECS and the developmental origins of health and disease (DOHaD).
Collapse
Affiliation(s)
- Mariana Macedo Almeida
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, RJ, Brazil
| | | | - Camila Calviño
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, RJ, Brazil
| | - Isis Hara Trevenzoli
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
28
|
Lichlyter DA, Krumm ZA, Golde TA, Doré S. Role of CRF and the hypothalamic-pituitary-adrenal axis in stroke: revisiting temporal considerations and targeting a new generation of therapeutics. FEBS J 2022; 290:1986-2010. [PMID: 35108458 DOI: 10.1111/febs.16380] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/10/2021] [Accepted: 01/31/2022] [Indexed: 12/13/2022]
Abstract
Ischaemic neurovascular stroke represents a leading cause of death in the developed world. Preclinical and human epidemiological evidence implicates the corticotropin-releasing factor (CRF) family of neuropeptides as mediators of acute neurovascular injury pathology. Preclinical investigations of the role of CRF, CRF receptors and CRF-dependent activation of the hypothalamic-pituitary-adrenal (HPA) axis have pointed toward a tissue-specific and temporal relationship between activation of these pathways and physiological outcomes. Based on the literature, the major phases of ischaemic stroke aetiology may be separated into an acute phase in which CRF and anti-inflammatory stress signalling are beneficial and a chronic phase in which these contribute to neural degeneration, toxicity and apoptotic signalling. Significant gaps in knowledge remain regarding the pathway, temporality and systemic impact of CRF signalling and stress biology in neurovascular injury progression. Heterogeneity among experimental designs poses a challenge to defining the apparent reciprocal relationship between neurological injury and stress metabolism. Despite these challenges, it is our opinion that the elucidated temporality may be best matched with an antibody against CRF with a half-life of days to weeks as opposed to minutes to hours as with small-molecule CRF receptor antagonists. This state-of-the-art review will take a multipronged approach to explore the expected potential benefit of a CRF antibody by modulating CRF and corticotropin-releasing factor receptor 1 signalling, glucocorticoids and autonomic nervous system activity. Additionally, this review compares the modulation of CRF and HPA axis activity in neuropsychiatric diseases and their counterpart outcomes post-stroke and assess lessons learned from antibody therapies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Daniel A Lichlyter
- Department of Anesthesiology, University of Florida College of Medicine, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Zachary A Krumm
- Department of Neuroscience, University of Florida College of Medicine, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Todd A Golde
- Department of Neuroscience, University of Florida College of Medicine, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Sylvain Doré
- Department of Anesthesiology, University of Florida College of Medicine, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.,Department of Neuroscience, University of Florida College of Medicine, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.,Departments of Neurology, Psychiatry, Pharmaceutics, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
29
|
Simmons S, Langlois LD, Oyola MG, Gouty S, Wu TJ, Nugent FS. Blast-Induced Mild Traumatic Brain Injury Alterations of Corticotropin-Releasing Factor Neuronal Activity in the Mouse Hypothalamic Paraventricular Nucleus. Front Synaptic Neurosci 2022; 13:804898. [PMID: 35153711 PMCID: PMC8828487 DOI: 10.3389/fnsyn.2021.804898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
Blast-induced mild traumatic brain injury (mbTBI) is the most common cause of TBI in US service members and veterans. Those exposed to TBI are at greater risk of developing neuropsychiatric disorders such as posttraumatic stress disorder, anxiety and depressive disorders, and substance use disorders following TBI. Previously, we have demonstrated that mbTBI increases anxiety-like behaviors in mice and dysregulates stress at the level of corticotropin-releasing factor (CRF) neurons in the paraventricular nucleus (PVN). To expand on how mTBI may dysregulate the stress axis centrally, here PVN CRF neuronal activity was evaluated using whole cell-patch clamp recordings in hypothalamic slices from sham and mbTBI adult male CRF:tdTomato mice 7 days post-injury. We found that mbTBI generally did not affect the neuronal excitability and intrinsic membrane properties of PVN CRF neurons; this injury selectively increased the frequency of spontaneous neuronal firing of PVN CRF neurons localized to the dorsal PVN (dPVN) but not ventral PVN (vPVN). Consistently, mbTBI-induced dPVN CRF hyperactivity was associated with pre- and post-synaptic depression of spontaneous GABAergic transmission onto dPVN CRF neurons suggesting that mbTBI-induced GABAergic synaptic dysfunction may underlie dPVN CRF neuronal hyperactivity and increases in dPVN CRF signaling. The present results provide the first evidence for mbTBI-induced alterations in PVN CRF neuronal activity and GABAergic synaptic function that could mediate hypothalamic CRF dysregulation following mbTBI contributing to stress psychopathology associated with blast injury.
Collapse
Affiliation(s)
- Sarah Simmons
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Ludovic D. Langlois
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Mario G. Oyola
- Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Shawn Gouty
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - T. John Wu
- Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- *Correspondence: Fereshteh S. Nugent T. John Wu
| | - Fereshteh S. Nugent
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- *Correspondence: Fereshteh S. Nugent T. John Wu
| |
Collapse
|
30
|
Sheng JA, Tan SML, Hale TM, Handa RJ. Androgens and Their Role in Regulating Sex Differences in the Hypothalamic/Pituitary/Adrenal Axis Stress Response and Stress-Related Behaviors. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2022; 2:261-274. [PMID: 35024695 PMCID: PMC8744007 DOI: 10.1089/andro.2021.0021] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/24/2021] [Indexed: 02/06/2023]
Abstract
Androgens play a pivotal role during development. These gonadal hormones and their receptors exert organizational actions that shape brain morphology in regions controlling the stress regulatory systems in a male-specific manner. Specifically, androgens drive sex differences in the hypothalamic/pituitary/adrenal (HPA) axis and corresponding hypothalamic neuropeptides. While studies have examined the role of estradiol and its receptors in sex differences in the HPA axis and associated behaviors, the role of androgens remains far less studied. Androgens are generally thought to modulate the HPA axis through the activation of androgen receptors (ARs). They can also impact the HPA axis through reduction to estrogenic metabolites that can bind estrogen receptors in the brain and periphery. Such regulation of the HPA axis stress response by androgens can often result in sex-biased risk factors for stress-related disorders, such as anxiety and depression. This review focuses on the biosynthesis pathways and molecular actions of androgens and their nuclear receptors. The impact of androgens on hypothalamic neuropeptide systems (corticotropin-releasing hormone, arginine vasopressin, oxytocin, dopamine, and serotonin) that control the stress response and stress-related disorders is discussed. Finally, this review discusses potential therapeutics involving androgens (androgen replacement therapies, selective AR modulator therapies) and ongoing clinical trials.
Collapse
Affiliation(s)
- Julietta A Sheng
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Sarah M L Tan
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Taben M Hale
- Department of Basic Medical Science, University of Arizona College of Medicine - Phoenix, Arizona, USA
| | - Robert J Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
31
|
Short AK, Thai CW, Chen Y, Kamei N, Pham AL, Birnie MT, Bolton JL, Mortazavi A, Baram TZ. Single-Cell Transcriptional Changes in Hypothalamic Corticotropin-Releasing Factor-Expressing Neurons After Early-Life Adversity Inform Enduring Alterations in Vulnerabilities to Stress. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2021; 3:99-109. [PMID: 36712559 PMCID: PMC9874075 DOI: 10.1016/j.bpsgos.2021.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/11/2021] [Accepted: 12/03/2021] [Indexed: 02/01/2023] Open
Abstract
Background Mental health and vulnerabilities to neuropsychiatric disorders involve the interplay of genes and environment, particularly during sensitive developmental periods. Early-life adversity (ELA) and stress promote vulnerabilities to stress-related affective disorders, yet it is unknown how transient ELA dictates lifelong neuroendocrine and behavioral reactions to stress. The population of hypothalamic corticotropin-releasing factor (CRF)-expressing neurons that regulate stress responses is a promising candidate to mediate the long-lasting influences of ELA on stress-related behavioral and hormonal responses via enduring transcriptional and epigenetic mechanisms. Methods Capitalizing on a well-characterized model of ELA, we examined ELA-induced changes in gene expression profiles of CRF-expressing neurons in the hypothalamic paraventricular nucleus of developing male mice. We used single-cell RNA sequencing on isolated CRF-expressing neurons. We determined the enduring functional consequences of transcriptional changes on stress reactivity in adult ELA mice, including hormonal responses to acute stress, adrenal weights as a measure of chronic stress, and behaviors in the looming shadow threat task. Results Single-cell transcriptomics identified distinct and novel CRF-expressing neuronal populations, characterized by both their gene expression repertoire and their neurotransmitter profiles. ELA-provoked expression changes were selective to specific subpopulations and affected genes involved in neuronal differentiation, synapse formation, energy metabolism, and cellular responses to stress and injury. Importantly, these expression changes were impactful, apparent from adrenal hypertrophy and augmented behavioral responses to stress in adulthood. Conclusions We uncover a novel repertoire of stress-regulating CRF cell types differentially affected by ELA and resulting in augmented stress vulnerability, with relevance to the origins of stress-related affective disorders.
Collapse
Affiliation(s)
- Annabel K. Short
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California
| | - Christina W. Thai
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California
| | - Yuncai Chen
- Department of Pediatrics, University of California Irvine, Irvine, California
| | - Noriko Kamei
- Department of Pediatrics, University of California Irvine, Irvine, California
| | - Aidan L. Pham
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California
| | - Matthew T. Birnie
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California
| | - Jessica L. Bolton
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California
| | - Tallie Z. Baram
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California,Department of Neurology, University of California Irvine, Irvine, California,Address correspondence to Tallie Z. Baram, M.D., Ph.D.
| |
Collapse
|
32
|
Jones JR, Chaturvedi S, Granados-Fuentes D, Herzog ED. Circadian neurons in the paraventricular nucleus entrain and sustain daily rhythms in glucocorticoids. Nat Commun 2021; 12:5763. [PMID: 34599158 PMCID: PMC8486846 DOI: 10.1038/s41467-021-25959-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 09/02/2021] [Indexed: 02/08/2023] Open
Abstract
Signals from the central circadian pacemaker, the suprachiasmatic nucleus (SCN), must be decoded to generate daily rhythms in hormone release. Here, we hypothesized that the SCN entrains rhythms in the paraventricular nucleus (PVN) to time the daily release of corticosterone. In vivo recording revealed a critical circuit from SCN vasoactive intestinal peptide (SCNVIP)-producing neurons to PVN corticotropin-releasing hormone (PVNCRH)-producing neurons. PVNCRH neurons peak in clock gene expression around midday and in calcium activity about three hours later. Loss of the clock gene Bmal1 in CRH neurons results in arrhythmic PVNCRH calcium activity and dramatically reduces the amplitude and precision of daily corticosterone release. SCNVIP activation reduces (and inactivation increases) corticosterone release and PVNCRH calcium activity, and daily SCNVIP activation entrains PVN clock gene rhythms by inhibiting PVNCRH neurons. We conclude that daily corticosterone release depends on coordinated clock gene and neuronal activity rhythms in both SCNVIP and PVNCRH neurons.
Collapse
Affiliation(s)
- Jeff R Jones
- Department of Biology, Washington University, St. Louis, St. Louis, MO, USA
- Department of Biology, Texas A&M University, College Station, College Station, TX, USA
| | - Sneha Chaturvedi
- Department of Biology, Washington University, St. Louis, St. Louis, MO, USA
| | | | - Erik D Herzog
- Department of Biology, Washington University, St. Louis, St. Louis, MO, USA.
| |
Collapse
|
33
|
Ge T, Yao X, Zhao H, Yang W, Zou X, Peng F, Li B, Cui R. Gut microbiota and neuropsychiatric disorders: Implications for neuroendocrine-immune regulation. Pharmacol Res 2021; 173:105909. [PMID: 34543739 DOI: 10.1016/j.phrs.2021.105909] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 12/17/2022]
Abstract
Recently, increasing evidence has shown gut microbiota dysbiosis might be implicated in the physiological mechanisms of neuropsychiatric disorders. Altered microbial community composition, diversity and distribution traits have been reported in neuropsychiatric disorders. However, the exact pathways by which the intestinal microbiota contribute to neuropsychiatric disorders remain largely unknown. Given that the onset and progression of neuropsychiatric disorders are characterized with complicated alterations of neuroendocrine and immunology, both of which can be continually affected by gut microbiota via "microbiome-gut-brain axis". Thus, we assess the complicated crosstalk between neuroendocrine and immunological regulation might underlie the mechanisms of gut microbiota associated with neuropsychiatric disorders. In this review, we summarized clinical and preclinical evidence on the role of the gut microbiota in neuropsychiatry disorders, especially in mood disorders and neurodevelopmental disorders. This review may elaborate the potential mechanisms of gut microbiota implicating in neuroendocrine-immune regulation and provide a comprehensive understanding of physiological mechanisms for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Tongtong Ge
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Xiaoxiao Yao
- Department of Hepatopancreatobiliary Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Haisheng Zhao
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Xiaohan Zou
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Fanzhen Peng
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
34
|
De Guzman RM, Rosinger ZJ, Parra KE, Jacobskind JS, Justice NJ, Zuloaga DG. Alterations in corticotropin-releasing factor receptor type 1 in the preoptic area and hypothalamus in mice during the postpartum period. Horm Behav 2021; 135:105044. [PMID: 34507241 PMCID: PMC8653990 DOI: 10.1016/j.yhbeh.2021.105044] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 07/16/2021] [Accepted: 08/06/2021] [Indexed: 01/24/2023]
Abstract
Corticotropin-releasing factor (CRF) signaling through CRF receptor 1 (CRFR1) regulates autonomic, endocrine, and behavioral responses to stress, as well as behavioral changes during the maternal period. Previous work in our lab reported higher levels of CRFR1 in female, compared to male, mice within the rostral anteroventral periventricular nucleus (AVPV/PeN), a brain region involved in maternal behaviors. In this study, we used CRFR1-GFP reporter mice to investigate whether the reproductive status (postpartum vs. nulliparous) of acutely stressed females affects levels of CRFR1 in the AVPV/PeN and other regions involved in maternal functions. Compared to nulliparous, postpartum day 14 females showed increased AVPV/PeN CRFR1-GFP immunoreactivity and an elevated number of restraint stress-activated AVPV/PeN CRFR1 cells as assessed by immunohistochemical co-localization of CRFR1-GFP and phosphorylated CREB (pCREB). The medial preoptic area (MPOA) and paraventricular hypothalamus (PVN) of postpartum mice showed modest decreases in CRFR1-GFP immunoreactivity, while increased CRFR1-GFP/pCREB co-expressing cells were found in the PVN following restraint stress relative to nulliparous mice. Tyrosine hydroxylase (TH) and CRFR1-GFP co-localization was also assessed in the AVPV/PeN and other regions and revealed a decrease in co-localized neurons in the AVPV/PeN and ventral tegmental area of postpartum mice. Corticosterone analysis of restrained mice revealed blunted peak, but elevated recovery, levels in postpartum compared to nulliparous mice. Finally, we investigated projection patterns of AVPV/PeN CRFR1 neurons using female CRFR1-Cre mice and revealed dense efferent projections to several preoptic, hypothalamic, and hindbrain regions known to control stress-associated and maternal functions. Together, these findings contribute to our understanding of the neurobiology that might underlie changes in stress-related functions during the postpartum period.
Collapse
Affiliation(s)
- Rose M De Guzman
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - Zachary J Rosinger
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - Katherine E Parra
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - Jason S Jacobskind
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - Nicholas J Justice
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Sciences Center, Houston, TX, United States
| | - Damian G Zuloaga
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States.
| |
Collapse
|
35
|
Olson B, Diba P, Korzun T, Marks DL. Neural Mechanisms of Cancer Cachexia. Cancers (Basel) 2021; 13:cancers13163990. [PMID: 34439145 PMCID: PMC8391721 DOI: 10.3390/cancers13163990] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/05/2021] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Cancer cachexia is a devastating wasting syndrome that occurs in many illnesses, with signs and symptoms including anorexia, weight loss, cognitive impairment and fatigue. The brain is capable of exerting overarching homeostatic control of whole-body metabolism and is increasingly being recognized as an important mediator of cancer cachexia. Given the increased recognition and discovery of neural mechanisms of cancer cachexia, we sought to provide an in-depth review and update of mechanisms by which the brain initiates and propagates cancer cachexia programs. Furthermore, recent work has identified new molecular mediators of cachexia that exert their effects through their direct interaction with the brain. Therefore, this review will summarize neural mechanisms of cachexia and discuss recently identified neural mediators of cancer cachexia. Abstract Nearly half of cancer patients suffer from cachexia, a metabolic syndrome characterized by progressive atrophy of fat and lean body mass. This state of excess catabolism decreases quality of life, ability to tolerate treatment and eventual survival, yet no effective therapies exist. Although the central nervous system (CNS) orchestrates several manifestations of cachexia, the precise mechanisms of neural dysfunction during cachexia are still being unveiled. Herein, we summarize the cellular and molecular mechanisms of CNS dysfunction during cancer cachexia with a focus on inflammatory, autonomic and neuroendocrine processes and end with a discussion of recently identified CNS mediators of cachexia, including GDF15, LCN2 and INSL3.
Collapse
Affiliation(s)
- Brennan Olson
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; (B.O.); (P.D.); (T.K.)
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Parham Diba
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; (B.O.); (P.D.); (T.K.)
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Tetiana Korzun
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; (B.O.); (P.D.); (T.K.)
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Daniel L. Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
- Correspondence:
| |
Collapse
|
36
|
Huang Y, Lin X, Lin S. Neuropeptide Y and Metabolism Syndrome: An Update on Perspectives of Clinical Therapeutic Intervention Strategies. Front Cell Dev Biol 2021; 9:695623. [PMID: 34307371 PMCID: PMC8299562 DOI: 10.3389/fcell.2021.695623] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/21/2021] [Indexed: 12/20/2022] Open
Abstract
Through the past decade of research, the pathogenic mechanisms underlying metabolic syndrome have been suggested to involve not only the peripheral tissues, but also central metabolic regulation imbalances. The hypothalamus, and the arcuate nucleus in particular, is the control center for metabolic homeostasis and energy balance. Neuropeptide Y neurons are particularly abundantly expressed in the arcuate of the hypothalamus, where the blood-brain barrier is weak, such as to critically integrate peripheral metabolic signals with the brain center. Herein, focusing on metabolic syndrome, this manuscript aims to provide an overview of the regulatory effects of Neuropeptide Y on metabolic syndrome and discuss clinical intervention strategy perspectives for neurometabolic disease.
Collapse
Affiliation(s)
- Yinqiong Huang
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xiahong Lin
- Department of Endocrinology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.,Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| |
Collapse
|
37
|
Daviu N, Bains JS. Should I Stay or Should I Go? CRHPVN Neurons Gate State Transitions in Stress-Related Behaviors. Endocrinology 2021; 162:6206556. [PMID: 33787875 DOI: 10.1210/endocr/bqab061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Indexed: 11/19/2022]
Abstract
Corticotropin-releasing hormone (CRH) neurons in the paraventricular nucleus of the hypothalamus are the canonical controllers of the endocrine response to stress. Here we propose a new role for these cells as a gate for state transitions that allow the organism to engage in stress-related behaviors. Specifically, we review evidence indicating that activation of these cells at critical times allows organisms to move to a state that is permissive for motor action. This is evident when the organism is under duress (defensive behavior), when the organism has successfully vanquished a threat (coping behavior), and when an organism initiates approach to a conspecific (social behavior). The motor behavior that follows from the activation of CRH neurons is not necessarily under the control of these cells but is determined by higher order circuits that discriminate more refined features of environmental context to execute the appropriate behavior.
Collapse
Affiliation(s)
- Nuria Daviu
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jaideep S Bains
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
38
|
Elsaafien K, Kirchner MK, Mohammed M, Eikenberry SA, West C, Scott KA, de Kloet AD, Stern JE, Krause EG. Identification of Novel Cross-Talk between the Neuroendocrine and Autonomic Stress Axes Controlling Blood Pressure. J Neurosci 2021; 41:4641-4657. [PMID: 33858944 PMCID: PMC8260250 DOI: 10.1523/jneurosci.0251-21.2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 11/21/2022] Open
Abstract
The hypothalamic paraventricular nucleus (PVN) controls neuroendocrine axes and the autonomic nervous system to mount responses that cope with the energetic burdens of psychological or physiological stress. Neurons in the PVN that express the angiotensin Type 1a receptor (PVNAgtr1a) are implicated in neuroendocrine and autonomic stress responses; however, the mechanism by which these neurons coordinate activation of neuroendocrine axes with sympathetic outflow remains unknown. Here, we use a multidisciplinary approach to investigate intra-PVN signaling mechanisms that couple the activity of neurons synthesizing corticotropin-releasing-hormone (CRH) to blood pressure. We used the Cre-Lox system in male mice with in vivo optogenetics and cardiovascular recordings to demonstrate that excitation of PVNAgtr1a promotes elevated blood pressure that is dependent on the sympathetic nervous system. Next, neuroanatomical experiments found that PVNAgtr1a synthesize CRH, and intriguingly, fibers originating from PVNAgtr1a make appositions onto neighboring neurons that send projections to the rostral ventrolateral medulla and express CRH type 1 receptor (CRHR1) mRNA. We then used an ex vivo preparation that combined optogenetics, patch-clamp electrophysiology, and Ca2+ imaging to discover that excitation of PVNAgtr1a drives the local, intra-PVN release of CRH, which activates rostral ventrolateral medulla-projecting neurons via stimulation of CRHR1(s). Finally, we returned to our in vivo preparation and found that CRH receptor antagonism specifically within the PVN lowered blood pressure basally and during optogenetic activation of PVNAgtr1a Collectively, these results demonstrate that angiotensin II acts on PVNAgtr1a to conjoin hypothalamic-pituitary-adrenal axis activity with sympathetically mediated vasoconstriction in male mice.SIGNIFICANCE STATEMENT The survival of an organism is dependent on meeting the energetic demands imposed by stressors. This critical function is accomplished by the CNS's ability to orchestrate simultaneous activities of neurosecretory and autonomic axes. Here, we unveil a novel signaling mechanism within the paraventricular nucleus of the hypothalamus that links excitation of neurons producing corticotropin-releasing-hormone with excitation of neurons controlling sympathetic nervous system activity and blood pressure. The implication is that chronic stress exposure may promote cardiometabolic disease by dysregulating the interneuronal cross-talk revealed by our experiments.
Collapse
Affiliation(s)
- Khalid Elsaafien
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32611
- Center of Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida 32611
| | - Matthew K Kirchner
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, Georgia 30302
| | - Mazher Mohammed
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32611
| | - Sophia A Eikenberry
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida 32611
| | - Chloe West
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, Georgia 30302
| | - Karen A Scott
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32611
- Center of Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida 32611
| | - Annette D de Kloet
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida 32611
- Center of Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida 32611
- McKnight Brain Institute, University of Florida, Gainesville, Florida 32611
| | - Javier E Stern
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, Georgia 30302
| | - Eric G Krause
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32611
- Center of Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida 32611
- McKnight Brain Institute, University of Florida, Gainesville, Florida 32611
| |
Collapse
|
39
|
Barbier M, González JA, Houdayer C, Burdakov D, Risold P, Croizier S. Projections from the dorsomedial division of the bed nucleus of the stria terminalis to hypothalamic nuclei in the mouse. J Comp Neurol 2021; 529:929-956. [PMID: 32678476 PMCID: PMC7891577 DOI: 10.1002/cne.24988] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/30/2020] [Accepted: 07/14/2020] [Indexed: 12/18/2022]
Abstract
As stressful environment is a potent modulator of feeding, we seek in the present work to decipher the neuroanatomical basis for an interplay between stress and feeding behaviors. For this, we combined anterograde and retrograde tracing with immunohistochemical approaches to investigate the patterns of projections between the dorsomedial division of the bed nucleus of the stria terminalis (BNST), well connected to the amygdala, and hypothalamic structures such as the paraventricular (PVH) and dorsomedial (DMH), the arcuate (ARH) nuclei and the lateral hypothalamic areas (LHA) known to control feeding and motivated behaviors. We particularly focused our study on afferences to proopiomelanocortin (POMC), agouti-related peptide (AgRP), melanin-concentrating-hormone (MCH) and orexin (ORX) neurons characteristics of the ARH and the LHA, respectively. We found light to intense innervation of all these hypothalamic nuclei. We particularly showed an innervation of POMC, AgRP, MCH and ORX neurons by the dorsomedial and dorsolateral divisions of the BNST. Therefore, these results lay the foundation for a better understanding of the neuroanatomical basis of the stress-related feeding behaviors.
Collapse
Affiliation(s)
- Marie Barbier
- EA481, Neurosciences Intégratives et Cliniques, UFR SantéUniversité Bourgogne Franche‐ComtéBesançonFrance
- Department of PsychiatrySeaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - J. Antonio González
- The Francis Crick InstituteLondonUK
- The Rowett Institute, School of MedicineMedical Sciences and Nutrition, University of AberdeenAberdeenUK
| | - Christophe Houdayer
- EA481, Neurosciences Intégratives et Cliniques, UFR SantéUniversité Bourgogne Franche‐ComtéBesançonFrance
| | - Denis Burdakov
- The Francis Crick InstituteLondonUK
- Neurobehavioural Dynamics Lab, Institute for Neuroscience, D‐HESTSwiss Federal Institute of Technology / ETH ZürichZürichSwitzerland
| | - Pierre‐Yves Risold
- EA481, Neurosciences Intégratives et Cliniques, UFR SantéUniversité Bourgogne Franche‐ComtéBesançonFrance
| | - Sophie Croizier
- University of LausanneCenter for Integrative GenomicsLausanneSwitzerland
| |
Collapse
|
40
|
Giardino WJ, Pomrenze MB. Extended Amygdala Neuropeptide Circuitry of Emotional Arousal: Waking Up on the Wrong Side of the Bed Nuclei of Stria Terminalis. Front Behav Neurosci 2021; 15:613025. [PMID: 33633549 PMCID: PMC7900561 DOI: 10.3389/fnbeh.2021.613025] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/15/2021] [Indexed: 12/25/2022] Open
Abstract
Sleep is fundamental to life, and poor sleep quality is linked to the suboptimal function of the neural circuits that process and respond to emotional stimuli. Wakefulness ("arousal") is chiefly regulated by circadian and homeostatic forces, but affective mood states also strongly impact the balance between sleep and wake. Considering the bidirectional relationships between sleep/wake changes and emotional dynamics, we use the term "emotional arousal" as a representative characteristic of the profound overlap between brain pathways that: (1) modulate wakefulness; (2) interpret emotional information; and (3) calibrate motivated behaviors. Interestingly, many emotional arousal circuits communicate using specialized signaling molecules called neuropeptides to broadly modify neural network activities. One major neuropeptide-enriched brain region that is critical for emotional processing and has been recently implicated in sleep regulation is the bed nuclei of stria terminalis (BNST), a core component of the extended amygdala (an anatomical term that also includes the central and medial amygdalae, nucleus accumbens shell, and transition zones betwixt). The BNST encompasses an astonishing diversity of cell types that differ across many features including spatial organization, molecular signature, biological sex and hormonal milieu, synaptic input, axonal output, neurophysiological communication mode, and functional role. Given this tremendous complexity, comprehensive elucidation of the BNST neuropeptide circuit mechanisms underlying emotional arousal presents an ambitious set of challenges. In this review, we describe how rigorous investigation of these unresolved questions may reveal key insights to enhancing psychiatric treatments and global psychological wellbeing.
Collapse
|
41
|
Tong WH, Abdulai-Saiku S, Vyas A. Medial Amygdala Arginine Vasopressin Neurons Regulate Innate Aversion to Cat Odors in Male Mice. Neuroendocrinology 2021; 111:505-520. [PMID: 32447337 DOI: 10.1159/000508862] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/22/2020] [Indexed: 11/19/2022]
Abstract
Aversion to environmental cues of predators is an integral part of defensive behaviors in many prey animals. It enhances their survival and probability of future reproduction. At the same time, animals cannot be maximally defended because imperatives of defense usually trade-off with behaviors required for sexual reproduction like display of dominance and production of sexual pheromones. Here, we approach this trade-off through the lens of arginine vasopressin (AVP) neurons within the posterodorsal medial amygdala (MePD) of mice. This neuronal population is known to be involved in sexual behaviors like approach to sexually salient cues. We show that chemogenetic partial ablation of this neuronal population increases aversion to predator odors. Moreover, overexpression of AVP within this population is sufficient to reduce aversion to predator odors. The loss of fear of the predator odor occurs in parallel with increased recruitment of AVP neurons within the MePD. These observations suggest that AVP neurons in the medial aspect of the extended amygdala are a proximate locus for the reduction in innate fear during life stages dominated by reproductive efforts.
Collapse
Affiliation(s)
- Wen Han Tong
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Samira Abdulai-Saiku
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Ajai Vyas
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore,
| |
Collapse
|
42
|
Landry T, Li P, Shookster D, Jiang Z, Li H, Laing BT, Bunner W, Langton T, Tong Q, Huang H. Centrally circulating α-klotho inversely correlates with human obesity and modulates arcuate cell populations in mice. Mol Metab 2020; 44:101136. [PMID: 33301986 PMCID: PMC7777546 DOI: 10.1016/j.molmet.2020.101136] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/20/2022] Open
Abstract
Objective Our laboratory recently identified the centrally circulating α-klotho protein as a novel hypothalamic regulator of food intake and glucose metabolism in mice. The current study aimed to investigate novel molecular effectors of central α-klotho in the arcuate nucleus of the hypothalamus (ARC), while further deciphering its role regulating energy balance in both humans and mice. Methods Cerebrospinal fluid (CSF) was collected from 22 adults undergoing lower limb orthopedic surgeries, and correlations between body weight and α-klotho were determined using an α-klotho enzyme-linked immunosorbent assay (ELISA) kit. To investigate the effects of α-klotho on energy expenditure (EE), 2-day intracerebroventricular (ICV) treatment was performed in diet-induced obesity (DIO) mice housed in TSE Phenomaster indirect calorimetry metabolic cages. Immunohistochemical staining for cFOS and patch clamp electrophysiology were used to determine the effects of central α-klotho on proopiomelanocortin (POMC) and tyrosine hydroxylase (TH) neurons. Additional stainings were performed to determine novel roles for central α-klotho to regulate non-neuronal cell populations in the ARC. Lastly, ICV pretreatment with fibroblast growth factor receptor (FGFR) or PI3kinase inhibitors was performed to determine the intracellular signaling involved in α-klotho-mediated regulation of ARC nuclei. Results Obese/overweight human subjects had significantly lower CSF α-klotho concentrations compared to lean counterparts (1,044 ± 251 vs. 1616 ± 218 pmol/L, respectively). Additionally, 2 days of ICV α-klotho treatment increased EE in DIO mice. α-Klotho had no effects on TH neuron activity but elicited varied responses in POMC neurons, with 44% experiencing excitatory and 56% experiencing inhibitory effects. Inhibitor experiments identified an α-klotho→FGFR→PI3kinase signaling mechanism in the regulation of ARC POMC and NPY/AgRP neurons. Acute ICV α-klotho treatment also increased phosphorylated ERK in ARC astrocytes via FGFR signaling. Conclusion Our human CSF data provide the first evidence that impaired central α-klotho function may be involved in the pathophysiology of obesity. Furthermore, results in mouse models identify ARC POMC neurons and astrocytes as novel molecular effectors of central α-klotho. Overall, the current study highlights prominent roles of α-klotho→FGFR→PI3kinase signaling in the homeostatic regulation of ARC neurons and whole-body energy balance. Human CSF α-klotho concentrations exhibit a strong, inverse correlation with body weight and BMI. ICV α-klotho treatment increases energy expenditure in DIO mice. α-Klotho.→FGFR→PI3kinase signaling modulates ARC NPY/AgRP and POMC neurons. α-Klotho.→FGFR→ERK signaling regulates ARC astrocytes.
Collapse
Affiliation(s)
- Taylor Landry
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA; Department of Kinesiology, East Carolina University, Greenville, NC, USA; Human Performance Laboratory, College of Human Performance and Health, East Carolina University, Greenville, NC, USA
| | - Peixin Li
- Department of Comprehensive Surgery, Medical and Health Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Daniel Shookster
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA; Department of Kinesiology, East Carolina University, Greenville, NC, USA; Human Performance Laboratory, College of Human Performance and Health, East Carolina University, Greenville, NC, USA
| | - Zhiying Jiang
- Brown Foundation Institute of Molecular Medicine of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hongli Li
- Brown Foundation Institute of Molecular Medicine of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Brenton Thomas Laing
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA; Department of Kinesiology, East Carolina University, Greenville, NC, USA; Human Performance Laboratory, College of Human Performance and Health, East Carolina University, Greenville, NC, USA
| | - Wyatt Bunner
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA; Department of Kinesiology, East Carolina University, Greenville, NC, USA; Human Performance Laboratory, College of Human Performance and Health, East Carolina University, Greenville, NC, USA
| | - Theodore Langton
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA; Department of Kinesiology, East Carolina University, Greenville, NC, USA; Human Performance Laboratory, College of Human Performance and Health, East Carolina University, Greenville, NC, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hu Huang
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA; Department of Kinesiology, East Carolina University, Greenville, NC, USA; Human Performance Laboratory, College of Human Performance and Health, East Carolina University, Greenville, NC, USA; Department of Physiology, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
43
|
Sex-Specific Vasopressin Signaling Buffers Stress-Dependent Synaptic Changes in Female Mice. J Neurosci 2020; 40:8842-8852. [PMID: 33051356 DOI: 10.1523/jneurosci.1026-20.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/21/2020] [Accepted: 09/27/2020] [Indexed: 12/30/2022] Open
Abstract
In many species, social networks provide benefit for both the individual and the collective. In addition to transmitting information to others, social networks provide an emotional buffer for distressed individuals. Our understanding about the cellular mechanisms that contribute to buffering is poor. Stress has consequences for the entire organism, including a robust change in synaptic plasticity at glutamate synapses onto corticotropin-releasing hormone (CRH) neurons in the paraventricular nucleus of the hypothalamus (PVN). In females, however, this stress-induced metaplasticity is buffered by the presence of a naive partner. This buffering may be because of discrete behavioral interactions, signals in the context in which the interaction occurs (i.e., olfactory cues), or it may be influenced by local signaling events in the PVN. Here, we show that local vasopressin (VP) signaling in PVN buffers the short-term potentiation (STP) at glutamate synapses after stress. This social buffering of metaplasticity, which requires the presence of another individual, was prevented by pharmacological inhibition of the VP 1a receptor (V1aR) in female mice. Exogenous VP mimicked the effects of social buffering and reduced STP in CRHPVN neurons from females but not males. These findings implicate VP as a potential mediator of social buffering in female mice.SIGNIFICANCE STATEMENT In many organisms, including rodents and humans, social groups are beneficial to overall health and well-being. Moreover, it is through these social interactions that the harmful effects of stress can be mitigated, a phenomenon known as social buffering. In the present study, we describe a critical role for the neuropeptide vasopressin (VP) in social buffering of synaptic metaplasticity in stress-responsive corticotropin-releasing hormone (CRH) neurons in female mice. These effects of VP do not extend to social buffering of stress behaviors, suggesting this is a very precise and local form of sex-specific neuropeptide signaling.
Collapse
|
44
|
Supraspinal Mechanisms of Intestinal Hypersensitivity. Cell Mol Neurobiol 2020; 42:389-417. [PMID: 33030712 DOI: 10.1007/s10571-020-00967-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022]
Abstract
Gut inflammation or injury causes intestinal hypersensitivity (IHS) and hyperalgesia, which can persist after the initiating pathology resolves, are often referred to somatic regions and exacerbated by psychological stress, anxiety or depression, suggesting the involvement of both the spinal cord and the brain. The supraspinal mechanisms of IHS remain to be fully elucidated, however, over the last decades the series of intestinal pathology-associated neuroplastic changes in the brain has been revealed, being potentially responsible for the phenomenon. This paper reviews current clinical and experimental data, including the authors' own findings, on these functional, structural, and neurochemical/molecular changes within cortical, subcortical and brainstem regions processing and modulating sensory signals from the gut. As concluded in the review, IHS can develop and maintain due to the bowel inflammation/injury-induced persistent hyperexcitability of viscerosensory brainstem and thalamic nuclei and sensitization of hypothalamic, amygdala, hippocampal, anterior insular, and anterior cingulate cortical areas implicated in the neuroendocrine, emotional and cognitive modulation of visceral sensation and pain. An additional contribution may come from the pathology-triggered dysfunction of the brainstem structures inhibiting nociception. The mechanism underlying IHS-associated regional hyperexcitability is enhanced NMDA-, AMPA- and group I metabotropic receptor-mediated glutamatergic neurotransmission in association with altered neuropeptide Y, corticotropin-releasing factor, and cannabinoid 1 receptor signaling. These alterations are at least partially mediated by brain microglia and local production of cytokines, especially tumor necrosis factor α. Studying the IHS-related brain neuroplasticity in greater depth may enable the development of new therapeutic approaches against chronic abdominal pain in inflammatory bowel disease.
Collapse
|
45
|
Chen C, Jiang Z, Fu X, Yu D, Huang H, Tasker JG. Astrocytes Amplify Neuronal Dendritic Volume Transmission Stimulated by Norepinephrine. Cell Rep 2020; 29:4349-4361.e4. [PMID: 31875545 PMCID: PMC7010232 DOI: 10.1016/j.celrep.2019.11.092] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/10/2019] [Accepted: 11/22/2019] [Indexed: 11/03/2022] Open
Abstract
In addition to their support role in neurotransmitter and ion buffering, astrocytes directly regulate neurotransmission at synapses via local bidirectional signaling with neurons. Here, we reveal a form of neuronal-astrocytic signaling that transmits retrograde dendritic signals to distal upstream neurons in order to activate recurrent synaptic circuits. Norepinephrine activates α1 adrenoreceptors in hypothalamic corticotropin-releasing hormone (CRH) neurons to stimulate dendritic release, which triggers an astrocytic calcium response and release of ATP; ATP stimulates action potentials in upstream glutamate and GABA neurons to activate recurrent excitatory and inhibitory synaptic circuits to the CRH neurons. Thus, norepinephrine activates a retrograde signaling mechanism in CRH neurons that engages astrocytes in order to extend dendritic volume transmission to reach distal presynaptic glutamate and GABA neurons, thereby amplifying volume transmission mediated by dendritic release.
Collapse
Affiliation(s)
- Chun Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - ZhiYing Jiang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Xin Fu
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Diankun Yu
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Hai Huang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Jeffrey G Tasker
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA.
| |
Collapse
|
46
|
Polymorphisms of stress pathway genes and emergence of suicidal ideation at antidepressant treatment onset. Transl Psychiatry 2020; 10:320. [PMID: 32952155 PMCID: PMC7502493 DOI: 10.1038/s41398-020-01003-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 01/17/2023] Open
Abstract
The prescription of antidepressant drugs is one of the most frequently used strategies to prevent suicide and suicidal behavior. However, some patients develop suicidal ideation at antidepressant treatment onset, a phenomenon known as treatment-emergent suicidal ideation (TESI). Few studies have explored TESI pharmacogenomics. As the Hypothalamic-Pituitary-Adrenal (HPA) axis might be implicated in suicidal behavior, we assessed the relationship between TESI and single nucleotide polymorphisms (SNPs) in the HPA axis-implicated NR3C1 (n = 7 SNPs), FKBP5 (n = 5 SNPs), AVPR1B (n = 1 SNPs), CRHR1 (n = 1 SNPs), and SKA2 (n = 1 SNPs) genes, in a sample of 3566 adult outpatients with depression for whom an antidepressant treatment was introduced. General practitioners and psychiatrists throughout France followed participants for 6 weeks after the initial prescription of tianeptine, an antidepressant molecule showing mu agonism. Suicidal ideation was assessed with item 10 of the Montgomery-Åsberg Depression Rating Scale (item dedicated to suicidal ideation) at baseline, and at week 2, 4, and 6 of treatment. Within the informative sample, 112 patients reported TESI and 384 did not. TESI was significantly associated with the TT genotype of the SNP rs6902321 in FKBP5 (OR = 1.76, 95% CI = [1.07; 2.90]; p-value = 0.03) and the GG/AG genotype of the SNP rs7208505 in SKA2 (OR = 1.85, 95% CI = [1.03;3.33]; p-value = 0.04). These associations were not significant after multiple test correction. Nevertheless, our results suggest a possible involvement of HPA axis elements in treatment-emergent suicidal ideation (TESI).
Collapse
|
47
|
Agoglia AE, Tella J, Herman MA. Sex differences in corticotropin releasing factor peptide regulation of inhibitory control and excitability in central amygdala corticotropin releasing factor receptor 1-neurons. Neuropharmacology 2020; 180:108296. [PMID: 32950560 DOI: 10.1016/j.neuropharm.2020.108296] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/26/2020] [Accepted: 08/30/2020] [Indexed: 12/28/2022]
Abstract
The central amygdala (CeA) is a critical regulator of emotional behavior that has been implicated in psychiatric illnesses, including anxiety disorders and addiction. The CeA corticotropin releasing factor receptor 1 (CRF1) system has been implicated in alcohol use disorder (AUD) and mood disorders, and has been shown to regulate anxiety-like behavior and alcohol consumption in rodents. However, the effects of CRF signaling within the CRF receptor 1-containing (CRF1+) population of the CeA remain unclear, and the effects of ethanol and CRF1 manipulations in female rodents have not been assessed. Here, we characterized inhibitory control and CRF1 signaling in male and female CRF1-GFP reporter mice. Male and female CRF1+ CeA neurons exhibited similar baseline GABAergic signaling and excitability and were comparably sensitive to CRF-induced increases in presynaptic GABA release. CRF1 antagonism reduced GABA release onto CRF1-containing neurons comparably in both males and females. Acute ethanol application reduced GABA release onto CRF1+ neurons from males, but female CRF1+ neurons were insensitive to ethanol. Exogenous CRF increased the firing rate of CRF1-containing neurons to a greater extent in male cells versus female cells, and CRF1 antagonism reduced firing in females but not males. Together, these findings indicate a critical sex-specific role for the CRF system in regulating inhibitory control and excitability of CRF1-containing neurons in the central amygdala. Sex differences in sensitivity of CRF/CRF1 signaling provide useful context for the sex differences in psychiatric illness reported in human patients, particularly AUD.
Collapse
Affiliation(s)
- Abigail E Agoglia
- Department of Pharmacology, United States; Bowles Center for Alcohol Studies, United States
| | | | - Melissa A Herman
- Department of Pharmacology, United States; Bowles Center for Alcohol Studies, United States.
| |
Collapse
|
48
|
Zuloaga DG, Heck AL, De Guzman RM, Handa RJ. Roles for androgens in mediating the sex differences of neuroendocrine and behavioral stress responses. Biol Sex Differ 2020; 11:44. [PMID: 32727567 PMCID: PMC7388454 DOI: 10.1186/s13293-020-00319-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 07/09/2020] [Indexed: 12/17/2022] Open
Abstract
Estradiol and testosterone are powerful steroid hormones that impact brain function in numerous ways. During development, these hormones can act to program the adult brain in a male or female direction. During adulthood, gonadal steroid hormones can activate or inhibit brain regions to modulate adult functions. Sex differences in behavioral and neuroendocrine (i.e., hypothalamic pituitary adrenal (HPA) axis) responses to stress arise as a result of these organizational and activational actions. The sex differences that are present in the HPA and behavioral responses to stress are particularly important considering their role in maintaining homeostasis. Furthermore, dysregulation of these systems can underlie the sex biases in risk for complex, stress-related diseases that are found in humans. Although many studies have explored the role of estrogen and estrogen receptors in mediating sex differences in stress-related behaviors and HPA function, much less consideration has been given to the role of androgens. While circulating androgens can act by binding and activating androgen receptors, they can also act by metabolism to estrogenic molecules to impact estrogen signaling in the brain and periphery. This review focuses on androgens as an important hormone for modulating the HPA axis and behaviors throughout life and for setting up sex differences in key stress regulatory systems that could impact risk for disease in adulthood. In particular, impacts of androgens on neuropeptide systems known to play key roles in HPA and behavioral responses to stress (corticotropin-releasing factor, vasopressin, and oxytocin) are discussed. A greater knowledge of androgen action in the brain is key to understanding the neurobiology of stress in both sexes.
Collapse
Affiliation(s)
| | - Ashley L Heck
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | | | - Robert J Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
49
|
Lecarpentier Y, Schussler O, Hébert JL, Vallée A. Molecular Mechanisms Underlying the Circadian Rhythm of Blood Pressure in Normotensive Subjects. Curr Hypertens Rep 2020; 22:50. [PMID: 32661611 PMCID: PMC7359176 DOI: 10.1007/s11906-020-01063-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Blood pressure (BP) follows a circadian rhythm (CR) in normotensive subjects. BP increases in the morning and decreases at night. This review aims at providing an up-to-date overview regarding the molecular mechanisms underlying the circadian regulation of BP. RECENT FINDINGS The suprachiasmatic nucleus (SCN) is the regulatory center for CRs. In SCN astrocytes, the phosphorylated glycogen synthase kinase-3β (pGSK-3β) also follows a CR and its expression reaches a maximum in the morning and decreases at night. pGSK-3β induces the β-catenin migration to the nucleus. During the daytime, the nuclear β-catenin increases the expression of the glutamate excitatory amino acid transporter 2 (EAAT2) and glutamine synthetase (GS). In SCN, EAAT2 removes glutamate from the synaptic cleft of glutamatergic neurons and transfers it to the astrocyte cytoplasm where GS converts glutamate into glutamine. Thus, glutamate decreases in the synaptic cleft. This decreases the stimulation of the glutamate receptors AMPA-R and NMDA-R located on glutamatergic post-synaptic neurons. Consequently, activation of NTS is decreased and BP increases. The opposite occurs at night. Despite several studies resulting from animal studies, the circadian regulation of BP appears largely controlled in normotensive subjects by the canonical WNT/β-catenin pathway involving the SCN, astrocytes, and glutamatergic neurons.
Collapse
Affiliation(s)
- Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien, 77104, Meaux, France.
| | - Olivier Schussler
- Department of Thoracic surgery, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- Department of Cardiovascular Surgery, Research Laboratory, Geneva University Hospital, Geneva, Switzerland
| | - Jean-Louis Hébert
- Cardiology Institute, Pitié-Salpétrière Hospital, AP-HP, Paris, France
| | - Alexandre Vallée
- Diagnosis and Therapeutic Center, Hypertension and Cardiovascular Prevention Unit, Paris-Descartes University, Hôtel-Dieu Hospital, AP-HP, Paris, France
| |
Collapse
|
50
|
Minokoshi Y, Nakajima KI, Okamoto S. Homeostatic versus hedonic control of carbohydrate selection. J Physiol 2020; 598:3831-3844. [PMID: 32643799 DOI: 10.1113/jp280066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/06/2020] [Indexed: 11/08/2022] Open
Abstract
Macronutrient intake is associated with cardiometabolic health, ageing and longevity, but the mechanisms underlying its regulation have remained unclear. Most rodents increase carbohydrate selection under certain physiological and pathological conditions such as fasting. When presented with a choice between a basally preferable high-fat diet (HFD) and a high-carbohydrate diet (HCD) such as a high-sucrose diet, fasted mice first eat the HFD and then switch to the HCD during the first few hours of refeeding and continue to eat the HCD up to 24 h in the two-diet choice approach. Such consumption of an HCD after fasting reverses the fasting-induced increase in the plasma concentration of ketone bodies more rapidly than does refeeding with an HFD alone. 5'-AMP-activated protein kinase (AMPK)-regulated neurons in the paraventricular nucleus of the hypothalamus (PVH) that express corticotropin-releasing hormone (CRH) are necessary and sufficient for the fasting-induced selection of carbohydrate over an HFD in mice. These neurons appear to contribute to a fasting-induced increase in the positive valence of carbohydrate without affecting the preference for more palatable and energy-dense diets such as an HFD. Identification of the neural circuits in which AMPK-regulated CRH neurons in the PVH of mice are embedded should shed new light on the physiological and molecular mechanisms responsible for macronutrient selection.
Collapse
Affiliation(s)
- Yasuhiko Minokoshi
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institute of Natural Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi, 444-8585, Japan.,School of Life Science, The Graduate University for Advanced Studies SOKENDAI, 38 Nishigonaka, Myodaiji, Okazaki, Aichi, 444-8585, Japan
| | - Ken-Ichiro Nakajima
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institute of Natural Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi, 444-8585, Japan.,School of Life Science, The Graduate University for Advanced Studies SOKENDAI, 38 Nishigonaka, Myodaiji, Okazaki, Aichi, 444-8585, Japan
| | - Shiki Okamoto
- Second Department of Internal Medicine (Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology), Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami-gun, Okinawa, 903-0215, Japan
| |
Collapse
|