1
|
Kim HR, Tabiatnejad P, Arestakesyan H, Young CN. Modulation of liver lipid metabolic pathways by central nervous system ER stress. Am J Physiol Endocrinol Metab 2025; 328:E833-E844. [PMID: 40261717 PMCID: PMC12147869 DOI: 10.1152/ajpendo.00392.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/11/2024] [Accepted: 04/01/2025] [Indexed: 04/24/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), considered as the hepatic manifestation of metabolic syndrome, can increase the risk for cardiometabolic diseases. Accumulating reports have implicated the central nervous system in MASLD pathogenesis, specifically endoplasmic reticulum (ER) stress in subfornical organ (SFO) to hypothalamic paraventricular nucleus (PVN) projecting neurons (SFO→PVN). Here, we investigated how ER stress in this neural circuit influences hepatic lipid regulatory pathways that may contribute to MASLD development during obesity. Hepatic steatosis was elicited by feeding C57BL/6J male mice a high-fat diet for 11 wk. Intersectional viral targeting was used to inhibit ER stress in SFO→PVN neurons to examine the contribution of ER stress in this circuit to hepatic lipid acquisition and disposal genes during obesity. Inhibition of ER stress in SFO→PVN neurons of obese mice resulted in a reduction in hepatic triglycerides and lipid acquisition genes that was paralleled by a reduction in liver tyrosine hydroxylase, the rate-limiting enzyme in catecholamine synthesis. Moreover, hepatic tyrosine hydroxylase expression was positively correlated with lipid acquisition but not disposal pathways. These results indicate that ER stress in SFO→PVN neurons may contribute to MASLD through sympathetic nervous system influences, primarily on hepatic lipid acquisition.NEW & NOTEWORTHY Endoplasmic reticulum stress in SFO→PVN neurons modulates hepatic lipid acquisition and disposal pathways during obesity-induced hepatic steatosis. Hepatic tyrosine hydroxylase levels are positively correlated with liver triglyceride levels and lipid acquisition pathway-related genes in diet-induced obese animals.
Collapse
Affiliation(s)
- Han Rae Kim
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Parisa Tabiatnejad
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Hovhannes Arestakesyan
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Colin N. Young
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| |
Collapse
|
2
|
Martins Dos Santos K, Saunders SE, Antunes VR, Boychuk CR. Insulin activates parasympathetic hepatic-related neurons of the paraventricular nucleus of the hypothalamus through mTOR signaling. J Neurophysiol 2025; 133:320-332. [PMID: 39665212 PMCID: PMC11918334 DOI: 10.1152/jn.00284.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/05/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024] Open
Abstract
Integration of autonomic and metabolic regulation, including hepatic function, is a critical role played by the brain's hypothalamic region. Specifically, the paraventricular nucleus of the hypothalamus (PVN) regulates autonomic functions related to metabolism, such as hepatic glucose production. Although insulin can act directly on hepatic tissue to inhibit hepatic glucose production, recent evidence implicates that central actions of insulin within PVN also regulate glucose metabolism. However, specific central circuits responsible for insulin signaling with relation to hepatic regulation are poorly understood. As a heterogeneous nucleus essential to controlling parasympathetic motor output with notable expression of insulin receptors, PVN is an appealing target for insulin-dependent modulation of parasympathetic activity. Here, we tested the hypothesis that insulin activates hepatic-related PVN (PVNhepatic) neurons through a parasympathetic pathway. Using transsynaptic retrograde tracing, labeling within PVN was first identified 24 h after its expression in the dorsal motor nucleus of the vagus (DMV) and 72 h after hepatic injection. Critically, nearly all labeling in medial PVN was abolished after a left vagotomy, indicating that PVNhepatic neurons in this region are part of a central circuit innervating parasympathetic motor neurons. Insulin also significantly increased the firing frequency of PVNhepatic neurons in this subregion. Mechanistically, rapamycin pretreatment inhibited insulin-dependent activation of PVNhepatic neurons. Therefore, central insulin signaling can activate a subset of PVNhepatic neurons that are part of a unique parasympathetic network in control of hepatic function. Taken together, PVNhepatic neurons related to parasympathetic output regulation could serve as a key central network in insulin's ability to control hepatic functions.NEW & NOTEWORTHY Increased peripheral insulin concentrations are known to decrease hepatic glucose production through both direct actions on hepatocytes and central autonomic networks. Despite this understanding, how (and in which brain regions) insulin exerts its action is still obscure. Here, we demonstrate that insulin activates parasympathetic hepatic-related PVN neurons (PVNhepatic) and that this effect relies on mammalian target of rapamycin (mTOR) signaling, suggesting that insulin modulates hepatic function through autonomic pathways involving insulin receptor intracellular signaling cascades.
Collapse
Affiliation(s)
- Karoline Martins Dos Santos
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, United States
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sandy E Saunders
- Dalton Cardiovascular Research Center, Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
| | - Vagner R Antunes
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carie R Boychuk
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, United States
- Dalton Cardiovascular Research Center, Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
3
|
Chen X, Yan X, Yu C, Chen QH, Bi L, Shan Z. PTSD Increases Risk for Hypertension Development Through PVN Activation and Vascular Dysfunction in Sprague Dawley Rats. Antioxidants (Basel) 2024; 13:1423. [PMID: 39594564 PMCID: PMC11590931 DOI: 10.3390/antiox13111423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
This study investigates the impact of single prolonged stress (SPS), a model of post-traumatic stress disorder (PTSD), on cardiovascular responses, hypothalamic paraventricular nucleus (PVN) activity, and vascular function to elucidate the mechanisms linking traumatic stress to hypertension. Although SPS did not directly cause chronic hypertension in male Sprague Dawley (SD) rats, it induced acute but transient increases in blood pressure and heart rate and significantly altered the expression of hypertension-associated genes, such as vasopressin, angiotensin II type 1 receptor (AT1R), and FOSL1 in the PVN. Notably, mitochondrial reactive oxygen species (mtROS) were predominantly elevated in the pre-autonomic regions of the PVN, colocalizing with AT1R- and FOSL1-expressing cells, suggesting that oxidative stress may amplify sympathetic activation and stress responses. SPS also increased mRNA levels of pro-inflammatory cytokines (TNFα and IL1β) and inducible nitric oxide synthase (iNOS) in the aorta, and impaired vascular reactivity to vasoconstrictor and vasodilator stimuli, reflecting compromised vascular function. These findings suggest that SPS-sensitize neuroendocrine, autonomic, and vascular pathways create a state of cardiovascular vulnerability that could predispose individuals to hypertension when exposed to additional stressors. Understanding these mechanisms provides critical insights into the pathophysiology of stress-related cardiovascular disorders and underscores the need for targeted therapeutic interventions that address oxidative stress and modulate altered PVN pathways to mitigate the cardiovascular impact of PTSD and related conditions.
Collapse
Affiliation(s)
- Xinqian Chen
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, MI 49931, USA; (X.C.); (Q.-h.C.)
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA; (X.Y.); (C.Y.)
| | - Xin Yan
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA; (X.Y.); (C.Y.)
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA
| | - Chunxiu Yu
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA; (X.Y.); (C.Y.)
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Qing-hui Chen
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, MI 49931, USA; (X.C.); (Q.-h.C.)
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA; (X.Y.); (C.Y.)
| | - Lanrong Bi
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA; (X.Y.); (C.Y.)
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA
| | - Zhiying Shan
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, MI 49931, USA; (X.C.); (Q.-h.C.)
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA; (X.Y.); (C.Y.)
| |
Collapse
|
4
|
Hasebe Y, Yokota S, Fukushi I, Takeda K, Yoshizawa M, Onimaru H, Kono Y, Sugama S, Uchiyama M, Koizumi K, Horiuchi J, Kakinuma Y, Pokorski M, Toda T, Izumizaki M, Mori Y, Sugita K, Okada Y. Persistence of post-stress blood pressure elevation requires activation of astrocytes. Sci Rep 2024; 14:22984. [PMID: 39363030 PMCID: PMC11450218 DOI: 10.1038/s41598-024-73345-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 09/16/2024] [Indexed: 10/05/2024] Open
Abstract
The reflexive excitation of the sympathetic nervous system in response to psychological stress leads to elevated blood pressure, a condition that persists even after the stress has been alleviated. This sustained increase in blood pressure, which may contribute to the pathophysiology of hypertension, could be linked to neural plasticity in sympathetic nervous activity. Given the critical role of astrocytes in various forms of neural plasticity, we investigated their involvement in maintaining elevated blood pressure during the post-stress phase. Specifically, we examined the effects of arundic acid, an astrocytic inhibitor, on blood pressure and heart rate responses to air-jet stress. First, we confirmed that the inhibitory effect of arundic acid is specific to astrocytes. Using c-Fos immunohistology, we then observed that psychological stress activates neurons in cardiovascular brain regions, and that this stress-induced neuronal activation was suppressed by arundic acid pre-treatment in rats. By evaluating astrocytic process thickness, we also confirmed that astrocytes in the cardiovascular brain regions were activated by stress, and this activation was blocked by arundic acid pre-treatment. Next, we conducted blood pressure measurements on unanesthetized, unrestrained rats. Air-jet stress elevated blood pressure, which remained high for a significant period during the post-stress phase. However, pre-treatment with arundic acid, which inhibited astrocytic activation, suppressed stress-induced blood pressure elevation both during and after stress. In contrast, arundic acid had no significant impact on heart rate. These findings suggest that both neurons and astrocytes play integral roles in stress-induced blood pressure elevation and its persistence after stress, offering new insights into the pathophysiological mechanisms underlying hypertension.
Collapse
Affiliation(s)
- Yohei Hasebe
- Department of Pediatrics, School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
- Clinical Research Center, Murayama Medical Center, 2-37-1 Gakuen, Musashimurayama, Tokyo, 208-0011, Japan
| | - Shigefumi Yokota
- Department of Anatomy and Morphological Neuroscience, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Isato Fukushi
- Clinical Research Center, Murayama Medical Center, 2-37-1 Gakuen, Musashimurayama, Tokyo, 208-0011, Japan
- Graduate School of Health Sciences, Aomori University of Health and Welfare, Aomori, Japan
| | - Kotaro Takeda
- Clinical Research Center, Murayama Medical Center, 2-37-1 Gakuen, Musashimurayama, Tokyo, 208-0011, Japan
- Faculty of Rehabilitation, School of Health Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Masashi Yoshizawa
- Department of Pediatrics, School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
- Clinical Research Center, Murayama Medical Center, 2-37-1 Gakuen, Musashimurayama, Tokyo, 208-0011, Japan
| | - Hiroshi Onimaru
- Department of Physiology, Showa University, School of Medicine, Tokyo, Japan
| | - Yosuke Kono
- Department of Pediatrics, School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
- Clinical Research Center, Murayama Medical Center, 2-37-1 Gakuen, Musashimurayama, Tokyo, 208-0011, Japan
| | - Shuei Sugama
- Center for Medical Sciences, International University of Health and Welfare, Otawara, Tochigi, Japan
| | - Makoto Uchiyama
- Department of Synthetic Chemistry and Biological Chemistry Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Keiichi Koizumi
- Department of Pediatrics, School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Jouji Horiuchi
- Department of Biomedical Engineering, Graduate School of Science and Engineering, Toyo University, Saitama, Japan
| | | | | | - Takako Toda
- Department of Pediatrics, School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Masahiko Izumizaki
- Department of Physiology, Showa University, School of Medicine, Tokyo, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Kanji Sugita
- Department of Pediatrics, School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Yasumasa Okada
- Clinical Research Center, Murayama Medical Center, 2-37-1 Gakuen, Musashimurayama, Tokyo, 208-0011, Japan.
| |
Collapse
|
5
|
Salam SNA, Kamaludin NF, Awang N, Ithnin A, Nata DHMS, Mohd Saat NZ. Occupational noise exposure and its effects among mill workers: A narrative review. Noise Health 2024; 26:461-473. [PMID: 39787546 PMCID: PMC11813248 DOI: 10.4103/nah.nah_43_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 05/03/2024] [Accepted: 08/21/2024] [Indexed: 01/12/2025] Open
Abstract
There are many possibilities for noise exposure in industrial workplaces, including sectors that extensively use heavy machinery in processing each product. Various studies indicate a causal relationship between noise exposure and auditory/nonauditory effects among workers. Noise exposure poses risks to health and workers' hearing acuity and safety. This review paper aims to identify and summarize occupational noise exposure by examining the auditory and nonauditory effects among mill workers. The mills included in the study are palm oil, lumber, textiles, rice, sugarcane, flour, paper, and sawmills. A two-step methodology was used to achieve the aims of this narrative review. The literature search was conducted by gathering PubMed and Google Scholar papers, focusing on occupational noise in different occupational mill environments. Grey's literature was conducted by screening the worldwide national standard for the laws, guidelines, and other related information based on this topic. A total of 22 articles published have been included. The content of each publication was subsequently summarized. Most of the machine areas within the mills emitted high level of noise. The auditory effects of occupational noise exposure among mill workers were noise-induced hearing loss and hearing impairment. Hypertension, stress, communication disturbance, cardiovascular disease, and headache were the nonauditory effects experienced by workers. The recommendations provided in this review for mitigating noise exposure are derived from the hierarchy of control. Overall, most mill workers are exposed to a high noise level exceeding the standard noise exposure limit. Preventive actions should be taken to address and reduce work-related injuries among mill workers worldwide.
Collapse
Affiliation(s)
- Siti Najihah Abdul Salam
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Nurul Farahana Kamaludin
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Normah Awang
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Anuar Ithnin
- Center for Community Health Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Dayana Hazwani Mohd Suadi Nata
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Nur Zakiah Mohd Saat
- Center for Community Health Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Ikeda S, Shinohara K, Kashihara S, Matsumoto S, Yoshida D, Nakashima R, Ono Y, Matsushima S, Tsutsui H, Kinugawa S. Esaxerenone: blood pressure reduction and cardiorenal protection without reflex sympathetic activation in salt-loaded stroke-prone spontaneously hypertensive rats. Hypertens Res 2024; 47:2133-2143. [PMID: 38802501 DOI: 10.1038/s41440-024-01733-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/18/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024]
Abstract
Mineralocorticoid receptor (MR) is involved in the mechanisms of blood pressure elevation, organ fibrosis, and inflammation. MR antagonists have been used in patients with hypertension, heart failure, or chronic kidney disease. Esaxerenone, a recently approved MR blocker with a nonsteroidal structure, has demonstrated a strong blood pressure-lowering effect. However, blood pressure reduction may lead to sympathetic activation through the baroreflex. The effect of esaxerenone on the sympathetic nervous system remains unclear. We investigated the effect of esaxerenone on organ damage and the sympathetic nervous system in salt-loaded stroke-prone spontaneously hypertensive rats (SHRSP), a well-established model of essential hypertension with sympathoexcitation and organ damage. Three-week administration of esaxerenone or hydralazine successfully attenuated the blood pressure elevation. Both esaxerenone and hydralazine comparably suppressed left ventricular hypertrophy and urinary albumin excretion. However, renal fibrosis and glomerular sclerosis were suppressed by esaxerenone but not hydralazine. Furthermore, plasma norepinephrine level, a parameter of systemic sympathetic activity, was significantly increased by hydralazine but not by esaxerenone. Consistent with these findings, the activity of the control centers of sympathetic nervous system, the parvocellular region of the paraventricular nucleus in the hypothalamus and the rostral ventrolateral medulla, was enhanced by hydralazine but remained unaffected by esaxerenone. These results suggest that esaxerenone effectively lowers blood pressure without inducing reflex sympathetic nervous system activation. Moreover, the organ-protective effects of esaxerenone appear to be partially independent of its blood pressure-lowering effect. In conclusion, esaxerenone demonstrates a blood pressure-lowering effect without concurrent sympathetic activation and exerts organ-protective effects in salt-loaded SHRSP. Esaxerenone has antihypertensive and cardiorenal protective effects without reflex sympathetic activation in salt-loaded stroke-prone spontaneously hypertensive rats.
Collapse
Affiliation(s)
- Shota Ikeda
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keisuke Shinohara
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Soichiro Kashihara
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sho Matsumoto
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daisuke Yoshida
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryosuke Nakashima
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshiyasu Ono
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- School of Medicine and Graduate School, International University of Health and Welfare, Fukuoka, Japan
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
7
|
Savani R, Park E, Busannagari N, Lu Y, Kwon H, Wang L, Pang Z. Metabolic and behavioral alterations associated with viral vector-mediated toxicity in the paraventricular hypothalamic nucleus. Biosci Rep 2024; 44:BSR20231846. [PMID: 38227343 PMCID: PMC10830444 DOI: 10.1042/bsr20231846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 01/17/2024] Open
Abstract
OBJECTIVE Combining adeno-associated virus (AAV)-mediated expression of Cre recombinase with genetically modified floxed animals is a powerful approach for assaying the functional role of genes in regulating behavior and metabolism. Extensive research in diverse cell types and tissues using AAV-Cre has shown it can save time and avoid developmental compensation as compared to using Cre driver mouse line crossings. We initially sought to study the impact of ablation of corticotropin-releasing hormone (CRH) in the paraventricular hypothalamic nucleus (PVN) using intracranial AAV-Cre injection in adult animals. METHODS In this study, we stereotactically injected AAV8-hSyn-Cre or a control AAV8-hSyn-GFP both Crh-floxed and wild-type mouse PVN to assess behavioral and metabolic impacts. We then used immunohistochemical markers to systematically evaluate the density of hypothalamic peptidergic neurons and glial cells. RESULTS We found that delivery of one specific preparation of AAV8-hSyn-Cre in the PVN led to the development of obesity, hyperphagia, and anxiety-like behaviors. This effect occurred independent of sex and in both floxed and wild-type mice. We subsequently found that AAV8-hSyn-Cre led to neuronal cell death and gliosis at the site of viral vector injections. These behavioral and metabolic deficits were dependent on injection into the PVN. An alternatively sourced AAV-Cre did not reproduce the same results. CONCLUSIONS Our findings reveal that delivery of a specific batch of AAV-Cre could lead to cellular toxicity and lesions in the PVN that cause robust metabolic and behavioral impacts. These alterations can complicate the interpretation of Cre-mediated gene knockout and highlight the need for rigorous controls.
Collapse
Affiliation(s)
- Rohan Savani
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
- Department of Cell Biology and Neuroscience, Undergraduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
| | - Erin Park
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
- Department of Cell Biology and Neuroscience, Undergraduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
| | - Nidhi Busannagari
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
- Department of Cell Biology and Neuroscience, Undergraduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
| | - Yi Lu
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
| | - Hyokjoon Kwon
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, U.S.A
| | - Le Wang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
| | - Zhiping P. Pang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, U.S.A
- Department of Neuroscience and Cell Biology, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, U.S.A
- Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, U.S.A
| |
Collapse
|
8
|
Ferraro S, Nigri A, Bruzzone MG, Medina Carrion JP, Fedeli D, Demichelis G, Chiapparini L, Ciullo G, Gonzalez AA, Proietti Cecchini A, Giani L, Becker B, Leone M. Involvement of the ipsilateral-to-the-pain anterior-superior hypothalamic subunit in chronic cluster headache. J Headache Pain 2024; 25:7. [PMID: 38212704 PMCID: PMC10782620 DOI: 10.1186/s10194-023-01711-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/27/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Despite hypothalamus has long being considered to be involved in the pathophysiology of cluster headache, the inconsistencies of previous neuroimaging studies and a limited understanding of the hypothalamic areas involved, impede a comprehensive interpretation of its involvement in this condition. METHODS We used an automated algorithm to extract hypothalamic subunit volumes from 105 cluster headache patients (57 chronic and 48 episodic) and 59 healthy individuals; after correcting the measures for the respective intracranial volumes, we performed the relevant comparisons employing logist regression models. Only for subunits that emerged as abnormal, we calculated their correlation with the years of illness and the number of headache attacks per day, and the effects of lithium treatment. As a post-hoc approach, using the 7 T resting-state fMRI dataset from the Human Connectome Project, we investigated whether the observed abnormal subunit, comprising the paraventricular nucleus and preoptic area, shows robust functional connectivity with the mesocorticolimbic system, which is known to be modulated by oxytocin neurons in the paraventricular nucleus and that is is abnormal in chronic cluster headache patients. RESULTS Patients with chronic (but not episodic) cluster headache, compared to control participants, present an increased volume of the anterior-superior hypothalamic subunit ipsilateral to the pain, which, remarkably, also correlates significantly with the number of daily attacks. The post-hoc approach showed that this hypothalamic area presents robust functional connectivity with the mesocorticolimbic system under physiological conditions. No evidence of the effects of lithium treatment on this abnormal subunit was found. CONCLUSIONS We identified the ipsilateral-to-the-pain antero-superior subunit, where the paraventricular nucleus and preoptic area are located, as the key hypothalamic region of the pathophysiology of chronic cluster headache. The significant correlation between the volume of this area and the number of daily attacks crucially reinforces this interpretation. The well-known roles of the paraventricular nucleus in coordinating autonomic and neuroendocrine flow in stress adaptation and modulation of trigeminovascular mechanisms offer important insights into the understanding of the pathophysiology of cluster headache.
Collapse
Affiliation(s)
- Stefania Ferraro
- School of Life Science and Technology, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
- Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, Italy
| | - Anna Nigri
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, Italy.
| | - Maria Grazia Bruzzone
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, Italy
| | - Jean Paul Medina Carrion
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, Italy
| | - Davide Fedeli
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, Italy
| | - Greta Demichelis
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, Italy
| | - Luisa Chiapparini
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, Italy
- Radiology Unit, Fodazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giuseppe Ciullo
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Ariosky Areces Gonzalez
- Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Faculty of Technical Sciences, University of Pinar del Río "Hermanos Saiz Montes de Oca", Pinar del Río, Cuba
| | | | - Luca Giani
- Department of Neurology, Fondazione Maugeri, IRCCS, Milan, Italy
| | - Benjamin Becker
- School of Life Science and Technology, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
- Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
- Department of Psychology, The University of Hong Kong, Hong Kong, China
| | - Massimo Leone
- Department of Neuroalgology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
9
|
Savani R, Park E, Busannagari N, Lu Y, Kwon H, Wang L, Pang ZP. Metabolic and behavioral alterations associated with viral vector-mediated toxicity in the paraventricular hypothalamic nucleus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.26.564009. [PMID: 37961695 PMCID: PMC10634907 DOI: 10.1101/2023.10.26.564009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Objective Combining adeno-associated virus (AAV)-mediated expression of Cre recombinase with genetically modified floxed animals is a powerful approach for assaying the functional role of genes in regulating behavior and metabolism. Extensive research in diverse cell types and tissues using AAV-Cre has shown it can save time and avoid developmental compensation as compared to using Cre driver mouse line crossings. We initially sought to study the impact of ablation of corticotropin-releasing hormone (CRH) in the paraventricular hypothalamic nucleus (PVN) using intracranial AAV-Cre injection in adult animals. Methods In this study, we stereotactically injected AAV8-hSyn-Cre or a control AAV8-hSyn-GFP both Crh-floxed and wild-type mouse PVN to assess behavioral and metabolic impacts. We then used immunohistochemical markers to systematically evaluate the density of hypothalamic peptidergic neurons and glial cells. Results We found that delivery of one specific preparation of AAV8-hSyn-Cre in the PVN led to the development of obesity, hyperphagia, and anxiety-like behaviors. This effect occurred independent of sex and in both floxed and wild-type mice. We subsequently found that AAV8-hSyn-Cre led to neuronal cell death and gliosis at the site of viral vector injections. These behavioral and metabolic deficits were dependent on injection into the PVN. An alternatively sourced AAV-Cre did not reproduce the same results. Conclusions Our findings reveal that delivery of a specific batch of AAV-Cre could lead to cellular toxicity and lesions in the PVN that cause robust metabolic and behavioral impacts. These alterations can complicate the interpretation of Cre-mediated gene knockout and highlight the need for rigorous controls.
Collapse
Affiliation(s)
- Rohan Savani
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Department of Cell Biology and Neuroscience, Undergraduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Erin Park
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Department of Cell Biology and Neuroscience, Undergraduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Nidhi Busannagari
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Department of Cell Biology and Neuroscience, Undergraduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Yi Lu
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Hyokjoon Kwon
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Le Wang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Zhiping P. Pang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
10
|
Van Loh BM, Yaw AM, Breuer JA, Jackson B, Nguyen D, Jang K, Ramos F, Ho EV, Cui LJ, Gillette DLM, Sempere LF, Gorman MR, Tonsfeldt KJ, Mellon PL, Hoffmann HM. The transcription factor VAX1 in VIP neurons of the suprachiasmatic nucleus impacts circadian rhythm generation, depressive-like behavior, and the reproductive axis in a sex-specific manner in mice. Front Endocrinol (Lausanne) 2023; 14:1269672. [PMID: 38205198 PMCID: PMC10777845 DOI: 10.3389/fendo.2023.1269672] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 11/28/2023] [Indexed: 01/12/2024] Open
Abstract
Background The suprachiasmatic nucleus (SCN) within the hypothalamus is a key brain structure required to relay light information to the body and synchronize cell and tissue level rhythms and hormone release. Specific subpopulations of SCN neurons, defined by their peptide expression, regulate defined SCN output. Here we focus on the vasoactive intestinal peptide (VIP) expressing neurons of the SCN. SCN VIP neurons are known to regulate circadian rhythms and reproductive function. Methods To specifically study SCN VIP neurons, we generated a novel knock out mouse line by conditionally deleting the SCN enriched transcription factor, Ventral Anterior Homeobox 1 (Vax1), in VIP neurons (Vax1Vip; Vax1fl/fl:VipCre). Results We found that Vax1Vip females presented with lengthened estrous cycles, reduced circulating estrogen, and increased depressive-like behavior. Further, Vax1Vip males and females presented with a shortened circadian period in locomotor activity and ex vivo SCN circadian period. On a molecular level, the shortening of the SCN period was driven, at least partially, by a direct regulatory role of VAX1 on the circadian clock genes Bmal1 and Per2. Interestingly, Vax1Vip females presented with increased expression of arginine vasopressin (Avp) in the paraventricular nucleus, which resulted in increased circulating corticosterone. SCN VIP and AVP neurons regulate the reproductive gonadotropin-releasing hormone (GnRH) and kisspeptin neurons. To determine how the reproductive neuroendocrine network was impacted in Vax1Vip mice, we assessed GnRH sensitivity to a kisspeptin challenge in vivo. We found that GnRH neurons in Vax1Vip females, but not males, had an increased sensitivity to kisspeptin, leading to increased luteinizing hormone release. Interestingly, Vax1Vip males showed a small, but significant increase in total sperm and a modest delay in pubertal onset. Both male and female Vax1Vip mice were fertile and generated litters comparable in size and frequency to controls. Conclusion Together, these data identify VAX1 in SCN VIP neurons as a neurological overlap between circadian timekeeping, female reproduction, and depressive-like symptoms in mice, and provide novel insight into the role of SCN VIP neurons.
Collapse
Affiliation(s)
- Brooke M. Van Loh
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, United States
| | - Alexandra M. Yaw
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, United States
| | - Joseph A. Breuer
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Brooke Jackson
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, United States
| | - Duong Nguyen
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, United States
| | - Krystal Jang
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, United States
| | - Fabiola Ramos
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, United States
| | - Emily V. Ho
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Laura J. Cui
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Dominique L. M. Gillette
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Lorenzo F. Sempere
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, United States
| | - Michael R. Gorman
- Department of Psychology, University of California, San Diego, La Jolla, CA, United States
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, United States
| | - Karen J. Tonsfeldt
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, United States
| | - Pamela L. Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, United States
| | - Hanne M. Hoffmann
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, United States
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
11
|
Walther LM, Wirtz PH. Physiological reactivity to acute mental stress in essential hypertension-a systematic review. Front Cardiovasc Med 2023; 10:1215710. [PMID: 37636310 PMCID: PMC10450926 DOI: 10.3389/fcvm.2023.1215710] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
Objective Exaggerated physiological reactions to acute mental stress (AMS) are associated with hypertension (development) and have been proposed to play an important role in mediating the cardiovascular disease risk with hypertension. A variety of studies compared physiological reactivity to AMS between essential hypertensive (HT) and normotensive (NT) individuals. However, a systematic review of studies across stress-reactive physiological systems including intermediate biological risk factors for cardiovascular diseases is lacking. Methods We conducted a systematic literature search (PubMed) for original articles and short reports, published in English language in peer-reviewed journals in November and December 2022. We targeted studies comparing the reactivity between essential HT and NT to AMS in terms of cognitive tasks, public speaking tasks, or the combination of both, in at least one of the predefined stress-reactive physiological systems. Results We included a total of 58 publications. The majority of studies investigated physiological reactivity to mental stressors of mild or moderate intensity. Whereas HT seem to exhibit increased reactivity in response to mild or moderate AMS only under certain conditions (i.e., in response to mild mental stressors with specific characteristics, in an early hyperkinetic stage of HT, or with respect to certain stress systems), increased physiological reactivity in HT as compared to NT to AMS of strong intensity was observed across all investigated stress-reactive physiological systems. Conclusion Overall, this systematic review supports the proposed and expected generalized physiological hyperreactivity to AMS with essential hypertension, in particular to strong mental stress. Moreover, we discuss potential underlying mechanisms and highlight open questions for future research of importance for the comprehensive understanding of the observed hyperreactivity to AMS in essential hypertension.
Collapse
Affiliation(s)
- Lisa-Marie Walther
- Biological Work and Health Psychology, University of Konstanz, Konstanz, Germany
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, Konstanz, Germany
| | - Petra H. Wirtz
- Biological Work and Health Psychology, University of Konstanz, Konstanz, Germany
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, Konstanz, Germany
| |
Collapse
|
12
|
Martins Sá RW, Theparambil SM, Dos Santos KM, Christie IN, Marina N, Cardoso BV, Hosford PS, Antunes VR. Salt-loading promotes extracellular ATP release mediated by glial cells in the hypothalamic paraventricular nucleus of rats. Mol Cell Neurosci 2023; 124:103806. [PMID: 36592801 DOI: 10.1016/j.mcn.2022.103806] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/01/2023] Open
Abstract
Previously, we have shown that purinergic signalling is involved in the control of hyperosmotic-induced sympathoexcitation at the level of the PVN, via activation of P2X receptors. However, the source(s) of ATP that drives osmotically-induced increases in sympathetic outflow remained undetermined. Here, we tested the two competing hypotheses that either (1) higher extracellular ATP in PVN during salt loading (SL) is a result of a failure of ectonucleotidases to metabolize ATP; and/or (2) SL can stimulate PVN astrocytes to release ATP. Rats were salt loaded with a 2 % NaCl solution replacing drinking water up to 4 days, an experimental model known to cause a gradual increase in blood pressure and plasma osmolarity. Immunohistochemical assessment of glial-fibrillary acidic protein (GFAP) revealed increased glial cell reactivity in the PVN of rats after 4 days of high salt exposure. ATP and adenosine release measurements via biosensors in hypothalamic slices showed that baseline ATP release was increased 17-fold in the PVN while adenosine remained unchanged. Disruption of Ca2+-dependent vesicular release mechanisms in PVN astrocytes by virally-driven expression of a dominant-negative SNARE protein decreased the release of ATP. The activity of ectonucleotidases quantified in vitro by production of adenosine from ATP was increased in SL group. Our results showed that SL stimulates the release of ATP in the PVN, at least in part, from glial cells by a vesicle-mediated route and likely contributes to the neural control of circulation during osmotic challenges.
Collapse
Affiliation(s)
- Renato W Martins Sá
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Shefeeq M Theparambil
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Karoline Martins Dos Santos
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Isabel N Christie
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Nephtali Marina
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Barbara V Cardoso
- Department of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Patrick S Hosford
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, UK.
| | - Vagner R Antunes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
13
|
Wang XY, Chen XQ, Wang GQ, Cai RL, Wang H, Wang HT, Peng XQ, Zhang MT, Huang S, Shen GM. A neural circuit for gastric motility disorders driven by gastric dilation in mice. Front Neurosci 2023; 17:1069198. [PMID: 36908796 PMCID: PMC9992744 DOI: 10.3389/fnins.2023.1069198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023] Open
Abstract
INTRODUCTION Symptoms of gastric motility disorders are common clinical manifestations of functional gastrointestinal disorders (FGIDs), and are triggered and exacerbated by stress, but the neural pathways underpinning them remain unclear. METHODS We set-up a mouse model by gastric dilation (GD) in which the gastric dynamics were assessed by installing strain gauges on the surface of the stomach. The neural pathway associated with gastric motility disorders was investigated by behavioral tests, electrophysiology, neural circuit tracing, and optogenetics and chemogenetics involving projections of the corticotropin-releasing hormone (CRH) from the paraventricular nucleus of the hypothalamus (PVN) to acetylcholine (ChAT) neurons in the dorsal motor nucleus of the vagus (DMV). RESULTS We found that GD induced gastric motility disorders were accompanied by activation of PVN CRH neurons, which could be alleviated by strategies that inhibits the activity of PVN CRH neurons. In addition, we identified a neural pathway in which PVN CRH neurons project into DMV ChAT neurons, modulated activity of the PVN CRH →DMV ChAT pathway to alleviate gastric motility disorders induced by GD. DISCUSSION These findings indicate that the PVN CRH →DMV ChAT pathway may mediate at least some aspects of GD related gastric motility, and provide new insights into the mechanisms by which somatic stimulation modulates the physiological functions of internal organs and systems.
Collapse
Affiliation(s)
- Xi-yang Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xiao-qi Chen
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Guo-quan Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Rong-lin Cai
- Research Institute of Acupuncture and Moxibustion, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hao Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hai-tao Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xiao-qi Peng
- School of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Meng-ting Zhang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Shun Huang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Guo-ming Shen
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Institute of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
14
|
Emerging mechanisms involving brain Kv7 channel in the pathogenesis of hypertension. Biochem Pharmacol 2022; 206:115318. [PMID: 36283445 DOI: 10.1016/j.bcp.2022.115318] [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: 08/24/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 12/14/2022]
Abstract
Hypertension is a prevalent health problem inducing many organ damages. The pathogenesis of hypertension involves a complex integration of different organ systems including the brain. The elevated sympathetic nerve activity is closely related to the etiology of hypertension. Ion channels are critical regulators of neuronal excitability. Several mechanisms have been proposed to contribute to hypothalamic-driven elevated sympathetic activity, including altered ion channel function. Recent findings indicate one of the voltage-gated potassium channels, Kv7 channels (M channels), plays a vital role in regulating cardiovascular-related neurons activity, and the expression of Kv7 channels is downregulated in hypertension. This review highlights recent findings that the Kv7 channels in the brain, blood vessels, and kidneys are emerging targets involved in the pathogenesis of hypertension, suggesting new therapeutic targets for treating drug-resistant, neurogenic hypertension.
Collapse
|
15
|
Lu Q, Kim JY. Mammalian circadian networks mediated by the suprachiasmatic nucleus. FEBS J 2022; 289:6589-6604. [PMID: 34657394 DOI: 10.1111/febs.16233] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/09/2021] [Accepted: 10/15/2021] [Indexed: 12/28/2022]
Abstract
The brain has a complex structure composed of hundreds of regions, forming networks to cooperate body functions. Therefore, understanding how various brain regions communicate with each other and with peripheral organs is important to understand human physiology. The suprachiasmatic nucleus (SCN) in the brain is the circadian pacemaker. The SCN receives photic information from the environment and conveys this to other parts of the brain and body to synchronize all circadian clocks. The circadian clock is an endogenous oscillator that generates daily rhythms in metabolism and physiology in almost all cells via a conserved transcriptional-translational negative feedback loop. So, the information flow from the environment to the SCN to other tissues synchronizes locally distributed circadian clocks to maintain homeostasis. Thus, understanding the circadian networks and how they adjust to environmental changes will better understand human physiology. This review will focus on circadian networks mediated by the SCN to understand how the environment, brain, and peripheral tissues form networks for cooperation.
Collapse
Affiliation(s)
- Qingqing Lu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Jin Young Kim
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Tung Foundation Biomedical Sciences Centre, Hong Kong, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| |
Collapse
|
16
|
Savić B, Murphy D, Japundžić-Žigon N. The Paraventricular Nucleus of the Hypothalamus in Control of Blood Pressure and Blood Pressure Variability. Front Physiol 2022; 13:858941. [PMID: 35370790 PMCID: PMC8966844 DOI: 10.3389/fphys.2022.858941] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/15/2022] [Indexed: 12/26/2022] Open
Abstract
The paraventricular nucleus (PVN) is a highly organized structure of the hypothalamus that has a key role in regulating cardiovascular and osmotic homeostasis. Functionally, the PVN is divided into autonomic and neuroendocrine (neurosecretory) compartments, both equally important for maintaining blood pressure (BP) and body fluids in the physiological range. Neurosecretory magnocellular neurons (MCNs) of the PVN are the main source of the hormones vasopressin (VP), responsible for water conservation and hydromineral balance, and oxytocin (OT), involved in parturition and milk ejection during lactation. Further, neurosecretory parvocellular neurons (PCNs) take part in modulation of the hypothalamic–pituitary–adrenal axis and stress responses. Additionally, the PVN takes central place in autonomic adjustment of BP to environmental challenges and contributes to its variability (BPV), underpinning the PVN as an autonomic master controller of cardiovascular function. Autonomic PCNs of the PVN modulate sympathetic outflow toward heart, blood vessels and kidneys. These pre-autonomic neurons send projections to the vasomotor nucleus of rostral ventrolateral medulla and to intermediolateral column of the spinal cord, where postganglionic fibers toward target organs arise. Also, PVN PCNs synapse with NTS neurons which are the end-point of baroreceptor primary afferents, thus, enabling the PVN to modify the function of baroreflex. Neuroendocrine and autonomic parts of the PVN are segregated morphologically but they work in concert when the organism is exposed to environmental challenges via somatodendritically released VP and OT by MCNs. The purpose of this overview is to address both neuroendocrine and autonomic PVN roles in BP and BPV regulation.
Collapse
Affiliation(s)
- Bojana Savić
- Laboratory for Cardiovascular Pharmacology and Toxicology, Faculty of Medicine, Institute of Pharmacology, Clinical Pharmacology and Toxicology, University of Belgrade, Belgrade, Serbia
| | - David Murphy
- Molecular Neuroendocrinology Research Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Nina Japundžić-Žigon
- Laboratory for Cardiovascular Pharmacology and Toxicology, Faculty of Medicine, Institute of Pharmacology, Clinical Pharmacology and Toxicology, University of Belgrade, Belgrade, Serbia
- *Correspondence: Nina Japundžić-Žigon,
| |
Collapse
|
17
|
Norcliffe-Kaufmann L. Stress and the baroreflex. Auton Neurosci 2022; 238:102946. [PMID: 35086020 DOI: 10.1016/j.autneu.2022.102946] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/16/2021] [Accepted: 01/16/2022] [Indexed: 11/27/2022]
Abstract
The stress response to emotions elicits the release of glucocorticoids from the adrenal cortex, epinephrine from the adrenal medulla, and norepinephrine from the sympathetic nerves. The baroreflex adapts to buffer these responses to ensure that perfusion to the organs meets the demands while maintaining blood pressure within a within a narrow range. While stressor-evoked autonomic cardiovascular responses may be adaptive for the short-term, the recurrent exaggerated cardiovascular stress reactions can be maladaptive in the long-term. Prolonged stress or loss of the baroreflex's buffering capacity can predispose episodes of heightened sympathetic activity during stress leading to hypertension, tachycardia, and ventricular wall motion abnormalities. This review discusses 1) how the baroreflex responds to acute and chronic stressors, 2) how lesions in the neuronal pathways of the baroreflex alter the ability to respond or counteract the stress response, and 3) the techniques to assess baroreflex sensitivity and stress responses. Evidence suggests that loss of baroreflex sensitivity may predispose heightened autonomic responses to stress and at least in part explain the association between stress, mortality and cardiovascular diseases.
Collapse
|
18
|
Pittman QJ. Vasopressin and central control of the cardiovascular system: A 40-year retrospective. J Neuroendocrinol 2021; 33:e13011. [PMID: 34235812 DOI: 10.1111/jne.13011] [Citation(s) in RCA: 5] [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: 05/26/2021] [Accepted: 06/25/2021] [Indexed: 01/24/2023]
Abstract
In the 40 years since vasopressin (AVP) was reported to have a central action with respect to raising blood pressure, the finding has been repeatedly replicated using a variety of complimentary approaches. The role of AVP as a central neurotransmitter involved in control of the cardiovascular system is now textbook material. However, it is evident that brain AVP plays, at best, a minor role in regulation of normal blood pressure. However, it appears to be an important player in a several cardiovascular-associated pathologies, ranging from hypertension to neural changes associated with heart failure. There are many interventions that have been shown to affect neural function, many of which are associated with alterations in behaviour. Possible alterations in neuronal AVP actions relevant to cardiovascular control in the setting of chronic inflammatory disease, early-life stress and inflammation are suggested areas for future research.
Collapse
Affiliation(s)
- Quentin J Pittman
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
19
|
Huang S, Li G, Pan Y, Liu J, Zhao J, Zhang X, Lu W, Wan X, Krebs CJ, Wang Z, Han W, Zhang Z. Population variation alters aggression-associated oxytocin and vasopressin expressions in brains of Brandt's voles in field conditions. Front Zool 2021; 18:56. [PMID: 34717666 PMCID: PMC8557550 DOI: 10.1186/s12983-021-00441-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 10/12/2021] [Indexed: 11/29/2022] Open
Abstract
Density-dependent change in aggressive behavior contributes to the population regulation of many small rodents, but the underlying neurological mechanisms have not been examined in field conditions. We hypothesized that crowding stress and aggression-associated oxytocin (OT) and arginine vasopressin (AVP) in specific regions of the brain may be closely related to aggressive behaviors and population changes of small rodents. We analyzed the association of OT and AVP expression, aggressive behavior, and population density of Brandt’s voles in 24 large semi-natural enclosures (0.48 ha each) in Inner Mongolia grassland. We tested the effects of population density on the OT/AVP system and aggressive behavior by experimentally manipulating populations of Brandt’s voles in the grassland enclosures. High density was positively and significantly associated with more aggressive behavior, and increased expression of mRNA and protein of AVP and its receptor, but decreased expression of mRNA and protein of OT and its receptor in specific brain regions of the voles. Our study suggests that changes in OT/AVP expression are likely a result of the increased psychosocial stress that these voles experience during overcrowding, and thus the OT/AVP system can be used as indicators of density-dependent stressors in Brandt’s voles.
Collapse
Affiliation(s)
- Shuli Huang
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guoliang Li
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yongliang Pan
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,School of Medicine, Huzhou University, Huzhou, 313000, China
| | - Jing Liu
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jidong Zhao
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xin Zhang
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Lu
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinrong Wan
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Charles J Krebs
- Department of Zoology, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Zuoxin Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306-1270, USA
| | - Wenxuan Han
- College of Resources and Environmental Sciences, China Agricultural University, Beijing, 100193, China
| | - Zhibin Zhang
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China. .,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
20
|
Nasimi A, Haddad F, Mirzaei-Damabi N, Rostami B, Hatam M. Another controller system for arterial pressure. AngII-vasopressin neural network of the parvocellular paraventricular nucleus may regulate arterial pressure during hypotension. Brain Res 2021; 1769:147618. [PMID: 34400123 DOI: 10.1016/j.brainres.2021.147618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023]
Abstract
Angiotensin II (AngII) immunoreactive cells, fibers and receptors, were found in the parvocelluar region of paraventricular nucleus (PVNp) and AngII receptors are present on vasopressinergic neurons. However, the mechanism by which vasopressin (AVP) and AngII may interact to regulate arterial pressure is not known. Thus, we tested the cardiovascular effects of blockade of the AngII receptors on AVP neurons and blockade of vasopressin V1a receptors on AngII neurons. We also explored whether the PVNp vasopressin plays a regulatory role during hypotension in anesthetized rat or not. Hypovolemic-hypotension was induced by gradual bleeding from femoral venous catheter. Either AngII or AVP injected into the PVNp produced pressor and tachycardia responses. The responses to AngII were blocked by V1a receptor antagonist. The responses to AVP were partially attenuated by AT1 antagonist and greatly attenuated by AT2 antagonist. Hemorrhage augmented the pressor response to AVP, indicating that during hemorrhage, sensitivity of PVNp to vasopressin was increased. By hemorrhagic-hypotension and bilateral blockade of V1a receptors of the PVNp, we found that vasopressinergic neurons of the PVNp regulate arterial pressure towards normal during hypotension. Taken together these findings and our previous findings about angII (Khanmoradi and Nasimi, 2017a) for the first time, we found that a mutual cooperative system of angiotensinergic and vasopressinergic neurons in the PVNp is a major regulatory controller of the cardiovascular system during hypotension.
Collapse
Affiliation(s)
- Ali Nasimi
- Dept. of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Haddad
- Dept. of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nafiseh Mirzaei-Damabi
- Dept. of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran; Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahar Rostami
- Dept. of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoumeh Hatam
- Dept. of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
21
|
Makrygianni EA, Chrousos GP. From Brain Organoids to Networking Assembloids: Implications for Neuroendocrinology and Stress Medicine. Front Physiol 2021; 12:621970. [PMID: 34177605 PMCID: PMC8222922 DOI: 10.3389/fphys.2021.621970] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 04/19/2021] [Indexed: 12/13/2022] Open
Abstract
Brain organoids are three-dimensional cultures that contain multiple types of cells and cytoarchitectures, and resemble fetal human brain structurally and functionally. These organoids are being used increasingly to model brain development and disorders, however, they only partially recapitulate such processes, because of several limitations, including inability to mimic the distinct cortical layers, lack of functional neuronal circuitry as well as non-neural cells and gyrification, and increased cellular stress. Efforts to create improved brain organoid culture systems have led to region-specific organoids, vascularized organoids, glia-containing organoids, assembloids, sliced organoids and polarized organoids. Assembloids are fused region-specific organoids, which attempt to recapitulate inter-regional and inter-cellular interactions as well as neural circuitry development by combining multiple brain regions and/or cell lineages. As a result, assembloids can be used to model subtle functional aberrations that reflect complex neurodevelopmental, neuropsychiatric and neurodegenerative disorders. Mammalian organisms possess a highly complex neuroendocrine system, the stress system, whose main task is the preservation of systemic homeostasis, when the latter is threatened by adverse forces, the stressors. The main central parts of the stress system are the paraventricular nucleus of the hypothalamus and the locus caeruleus/norepinephrine-autonomic nervous system nuclei in the brainstem; these centers innervate each other and interact reciprocally as well as with various other CNS structures. Chronic dysregulation of the stress system has been implicated in major pathologies, the so-called chronic non-communicable diseases, including neuropsychiatric, neurodegenerative, cardiometabolic and autoimmune disorders, which lead to significant population morbidity and mortality. We speculate that brain organoids and/or assembloids could be used to model the development, regulation and dysregulation of the stress system and to better understand stress-related disorders. Novel brain organoid technologies, combined with high-throughput single-cell omics and gene editing, could, thus, have major implications for precision medicine.
Collapse
Affiliation(s)
- Evanthia A Makrygianni
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Center for Adolescent Medicine and UNESCO Chair on Adolescent Health Care, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, Athens, Greece
| |
Collapse
|
22
|
Li Y, Yu XJ, Xiao T, Chi HL, Zhu GQ, Kang YM. Nrf1 Knock-Down in the Hypothalamic Paraventricular Nucleus Alleviates Hypertension Through Intervention of Superoxide Production-Removal Balance and Mitochondrial Function. Cardiovasc Toxicol 2021; 21:472-489. [PMID: 33582931 DOI: 10.1007/s12012-021-09641-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 01/28/2021] [Indexed: 12/19/2022]
Abstract
Oxidative stress in the hypothalamic paraventricular nucleus (PVN) contributes greatly to the development of hypertension. The recombinant nuclear respiratory factor 1 (Nrf1) regulates the transcription of several genes related to mitochondrial respiratory chain function or antioxidant expression, and thus may be involved in the pathogenesis of hypertension. Here we show that in the two-kidney, one-clip (2K1C) hypertensive rats the transcription level of Nrf1 was elevated comparing to the normotensive controls. Knocking down of Nrf1 in the PVN of 2K1C rats can significantly reduce their blood pressure and level of plasma norepinephrine (NE). Analysis revealed significant reduction of superoxide production level in both whole cell and mitochondria, along with up-regulation of superoxide dismutase 1 (Cu/Zn-SOD), NAD(P)H: quinone oxidoreductase 1 (NQO1), thioredoxin-dependent peroxiredoxin 3 (Prdx3), cytochrome c (Cyt-c) and glutathione synthesis rate-limiting enzyme (glutamyl-cysteine ligase catalytic subunit (Gclc) and modifier subunit (Gclm)), and down-regulation of cytochrome c oxidase subunit VI c (Cox6c) transcription after Nrf1 knock-down. In addition, the reduced ATP production and elevated mitochondrial membrane potential in the PVN of 2K1C rats were reinstated with Nrf1 knock-down, together with restored expression of peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α), mitochondrial transcription factor A (Tfam), coiled-coil myosin-like BCL2-interacting protein (Beclin1), and Mitofusin 1 (Mfn1), which are related to the mitochondrial biogenesis, fusion, and autophagy. Together, the results indicate that the PVN Nrf1 is associated with the development of 2K1C-induced hypertension, and Nrf1 knock-down in the PVN can alleviate hypertension through intervention of mitochondrial function and restorement of the production-removal balance of superoxide.
Collapse
Affiliation(s)
- Ying Li
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Shaanxi Engineering and Research Center of Vaccine, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, 710061, China
| | - Xiao-Jing Yu
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Shaanxi Engineering and Research Center of Vaccine, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, 710061, China
| | - Tong Xiao
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Shaanxi Engineering and Research Center of Vaccine, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, 710061, China
| | - Hong-Li Chi
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Shaanxi Engineering and Research Center of Vaccine, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, 710061, China
| | - Guo-Qing Zhu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, 210029, China
| | - Yu-Ming Kang
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Shaanxi Engineering and Research Center of Vaccine, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, 710061, China.
| |
Collapse
|
23
|
Jackson MG, Lightman SL, Gilmour G, Marston H, Robinson ESJ. Evidence for deficits in behavioural and physiological responses in aged mice relevant to the psychiatric symptom of apathy. Brain Neurosci Adv 2021; 5:23982128211015110. [PMID: 34104800 PMCID: PMC8161852 DOI: 10.1177/23982128211015110] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/13/2021] [Indexed: 11/30/2022] Open
Abstract
Apathy is widely reported in patients with neurological disorders or post viral infection but is also seen in otherwise-healthy aged individuals. This study investigated whether aged male mice express behavioural and physiological changes relevant to an apathy phenotype. Using measures of motivation to work for reward, we found deficits in the progressive ratio task related to rate of responding. In an effort-related decision-making task, aged mice were less willing to exert effort for high value reward. Aged mice exhibited reduced reward sensitivity but also lower measures of anxiety in the novelty supressed feeding test and an attenuated response to restraint stress with lower corticosterone and reduced paraventricular nucleus c-fos activation. This profile of affective changes did not align with those observed in models of depression but suggested emotional blunting. In a test of cognition (novel object recognition), aged mice showed no impairments, but activity was lower in a measure of exploration in a novel environment. Together, these data suggest aged mice show changes across the domains of motivated behaviour, reward sensitivity and emotional reactivity and may be a suitable model for the pre-clinical study of the psychiatric symptom of apathy.
Collapse
Affiliation(s)
- Megan G Jackson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Stafford L Lightman
- Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
| | | | | | - Emma S J Robinson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
24
|
Dos Santos KM, Moraes DJDA, da Silva MP, Antunes VR. Exercise training rescues the electrical activity of liver-projecting DMNV neurones in response to oxytocin in spontaneously hypertensive rats. J Neuroendocrinol 2021; 33:e12977. [PMID: 33942389 DOI: 10.1111/jne.12977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 12/15/2022]
Abstract
A neural circuit between the paraventricular nucleus of the hypothalamus (PVN) and the dorsal motor nucleus of the vagus (DMNV) constitutes part of an important parasympathetic autonomic pathway that controls hepatic glucose production. Intracerebroventricular injection of insulin activates oxytocinergic neurones in the PVN and elicits the release of oxytocin into the circulation, which plays an important role in the metabolism of glucose. Moreover, the central action of insulin can reduce the concentration of glucose in blood taken from the hepatic vein of Wistar rats via activation of vagal efferent nerves to the liver. This mechanism is impaired in sedentary spontaneously hypertensive rats (SHR). Because aerobic exercise increases vagal tone, partly mediated by increasing the oxytocinergic connections between the PVN and DMNV, we hypothesised that oxytocin (OT) might alter the excitability of liver-projecting DMNV neurones. Thus, we investigated the effects of OT on electrical properties of the liver-projecting DMNV neurones from Wistar, SHR subjected to 4 weeks of exercise training, as well sedentary controls, using whole cell patch-clamping. The results show that OT increased the resting membrane potential of DMNV neurones in Wistar rats, as well as the firing frequency of these cells, but not in sedentary SHR. However, in SHR subjected to 4 weeks of exercise training, the effects of OT on liver-projecting DMNV neurones of were similar to those seen in Wistar rats. These findings show that OT elicits similar changes in the electrophysiological properties of liver-projecting DMNV neurones of Wistar and exercise-trained but not sedentary SHR. These results indicate that exercise training can restore the sensitivity of liver-projecting DMNV neurones of exercise-trained SHR to OT.
Collapse
Affiliation(s)
- Karoline Martins Dos Santos
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Davi José de Almeida Moraes
- Department of Physiology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Melina Pires da Silva
- Department of Physiology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Vagner Roberto Antunes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
25
|
Genaro K, Prado WA. The role of the anterior pretectal nucleus in pain modulation: A comprehensive review. Eur J Neurosci 2021; 54:4358-4380. [PMID: 33909941 DOI: 10.1111/ejn.15255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/16/2021] [Accepted: 04/18/2021] [Indexed: 11/27/2022]
Abstract
Descending pain modulation involves multiple encephalic sites and pathways that range from the cerebral cortex to the spinal cord. Behavioral studies conducted in the 1980s revealed that electrical stimulation of the pretectal area causes antinociception dissociation from aversive responses. Anatomical and physiological studies identified the anterior pretectal nucleus and its descending projections to several midbrain, pontine, and medullary structures. The anterior pretectal nucleus is morphologically divided into a dorsal part that contains a dense neuron population (pars compacta) and a ventral part that contains a dense fiber band network (pars reticulata). Connections of the two anterior pretectal nucleus parts are broad and include prominent projections to and from major encephalic systems associated with somatosensory processes. Since the first observation that acute or chronic noxious stimuli activate the anterior pretectal nucleus, it has been established that numerous mediators participate in this response through distinct pathways. Recent studies have confirmed that at least two pain inhibitory pathways are activated from the anterior pretectal nucleus. This review focuses on rodent anatomical, behavioral, molecular, and neurochemical data that have helped to identify mediators of the anterior pretectal nucleus and pathways related to its role in pain modulation.
Collapse
Affiliation(s)
- Karina Genaro
- Department of Anesthesiology, University of California, Irvine, CA, USA
| | - Wiliam A Prado
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Burke SL, Barzel B, Jackson KL, Gueguen C, Young MJ, Head GA. Role of Mineralocorticoid and Angiotensin Type 1 Receptors in the Paraventricular Nucleus in Angiotensin-Induced Hypertension. Front Physiol 2021; 12:640373. [PMID: 33762970 PMCID: PMC7982587 DOI: 10.3389/fphys.2021.640373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/16/2021] [Indexed: 11/25/2022] Open
Abstract
The hypothalamic paraventricular nucleus (PVN) is an important site where an interaction between circulating angiotensin (Ang) and mineralocorticoid receptor (MR) activity may modify sympathetic nerve activity (SNA) to influence long-term elevation of blood pressure. We examined in conscious Ang II-treated rabbits, the effects on blood pressure and tonic and reflex renal SNA (RSNA) of microinjecting into the PVN either RU28318 to block MR, losartan to block Ang (AT1) receptors or muscimol to inhibit GABA A receptor agonist actions. Male rabbits received a moderate dose of Ang II (24 ng/kg/min subcutaneously) for 3 months (n = 13) or sham treatment (n = 13). At 3 months, blood pressure increased by +19% in the Ang II group compared to 10% in the sham (P = 0.022) but RSNA was similar. RU28318 lowered blood pressure in both Ang II and shams but had a greater effect on RSNA and heart rate in the Ang II-treated group (P < 0.05). Losartan also lowered RSNA, while muscimol produced sympatho-excitation in both groups. In Ang II-treated rabbits, RU28318 attenuated the blood pressure increase following chemoreceptor stimulation but did not affect responses to air jet stress. In contrast losartan and muscimol reduced blood pressure and RSNA responses to both hypoxia and air jet. While neither RU28318 nor losartan changed the RSNA baroreflex, RU28318 augmented the range of the heart rate baroreflex by 10% in Ang II-treated rabbits. Muscimol, however, augmented the RSNA baroreflex by 11% in sham animals and none of the treatments altered baroreflex sensitivity. In conclusion, 3 months of moderate Ang II treatment promotes activation of reflex RSNA principally via MR activation in the PVN, rather than via activation of AT1 receptors. However, the onset of hypertension is independent of both. Interestingly, the sympatho-excitatory effects of muscimol in both groups suggest that overall, the PVN regulates a tonic sympatho-inhibitory influence on blood pressure control.
Collapse
Affiliation(s)
- Sandra L. Burke
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Benjamin Barzel
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Kristy L. Jackson
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Cindy Gueguen
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Morag J. Young
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Geoffrey A. Head
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
27
|
Komnenov D, Quaal H, Rossi NF. V 1a and V 1b vasopressin receptors within the paraventricular nucleus contribute to hypertension in male rats exposed to chronic mild unpredictable stress. Am J Physiol Regul Integr Comp Physiol 2021; 320:R213-R225. [PMID: 33264070 DOI: 10.1152/ajpregu.00245.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/11/2020] [Accepted: 11/29/2020] [Indexed: 01/06/2023]
Abstract
Depression is an independent nontraditional risk factor for cardiovascular disease and mortality. The chronic unpredictable mild stress (CMS) rat model is a validated model of depression. Within the paraventricular nucleus (PVN), vasopressin (VP) via V1aR and V1bR have been implicated in stress and neurocardiovascular dysregulation. We hypothesized that in conscious, unrestrained CMS rats versus control, unstressed rats, PVN VP results in elevated arterial pressure (MAP), heart rate, and renal sympathetic nerve activity (RSNA) via activation of V1aR and/or V1bR. Male rats underwent 4 wk of CMS or control conditions. They were then equipped with hemodynamic telemetry transmitters, PVN cannula, and left renal nerve electrode. V1aR or V1bR antagonism dose-dependently inhibited MAP after VP injection. V1aR or V1bR blockers at their ED50 doses did not alter baseline parameters in either control or CMS rats but attenuated the pressor response to VP microinjected into PVN by ∼50%. Combined V1aR and V1bR inhibition completely blocked the pressor response to PVN VP in control but not CMS rats. CMS rats required combined maximally inhibitory doses to block either endogenous VP within the PVN or responses to microinjected VP. Compared with unstressed control rats, CMS rats had higher plasma VP levels and greater abundance of V1aR and V1bR transcripts within PVN. Thus, the CMS rat model of depression results in higher resting MAP, heart rate, and RSNA, which can be mitigated by inhibiting vasopressinergic mechanisms involving both V1aR and V1bR within the PVN. Circulating VP may also play a role in the pressor response.
Collapse
Affiliation(s)
- Dragana Komnenov
- Department of Internal Medicine, Wayne State University School of Medicine, Detroit, Michigan
| | - Harrison Quaal
- Department of Internal Medicine, Wayne State University School of Medicine, Detroit, Michigan
| | - Noreen F Rossi
- John D. Dingell Veterans Affairs Medical Center, Detroit, Michigan
- Department of Internal Medicine, Wayne State University School of Medicine, Detroit, Michigan
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
28
|
Gerlach DA, Manuel J, Hoff A, Kronsbein H, Hoffmann F, Heusser K, Ehmke H, Jordan J, Tank J, Beissner F. Medullary and Hypothalamic Functional Magnetic Imaging During Acute Hypoxia in Tracing Human Peripheral Chemoreflex Responses. Hypertension 2021; 77:1372-1382. [PMID: 33641354 DOI: 10.1161/hypertensionaha.120.16385] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Darius A Gerlach
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany (D.A.G., J.M., A.H., H.K., F.H., K.H., J.J., J.T.)
| | - Jorge Manuel
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany (D.A.G., J.M., A.H., H.K., F.H., K.H., J.J., J.T.).,Institute for Neuroradiology, Hannover Medical School, Germany (J.M., F.B.)
| | - Alex Hoff
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany (D.A.G., J.M., A.H., H.K., F.H., K.H., J.J., J.T.)
| | - Hendrik Kronsbein
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany (D.A.G., J.M., A.H., H.K., F.H., K.H., J.J., J.T.).,Institute of Cellular and Integrative Physiology, University Medical Center Eppendorf, Hamburg, Germany (H.K., H.E.)
| | - Fabian Hoffmann
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany (D.A.G., J.M., A.H., H.K., F.H., K.H., J.J., J.T.)
| | - Karsten Heusser
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany (D.A.G., J.M., A.H., H.K., F.H., K.H., J.J., J.T.)
| | - Heimo Ehmke
- Institute of Cellular and Integrative Physiology, University Medical Center Eppendorf, Hamburg, Germany (H.K., H.E.)
| | - Jens Jordan
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany (D.A.G., J.M., A.H., H.K., F.H., K.H., J.J., J.T.).,Chair of Aerospace Medicine, University of Cologne, Germany (J.J.)
| | - Jens Tank
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany (D.A.G., J.M., A.H., H.K., F.H., K.H., J.J., J.T.)
| | - Florian Beissner
- Institute for Neuroradiology, Hannover Medical School, Germany (J.M., F.B.)
| |
Collapse
|
29
|
Contoreggi NH, Mazid S, Goldstein LB, Park J, Ovalles AC, Waters EM, Glass MJ, Milner TA. Sex and age influence gonadal steroid hormone receptor distributions relative to estrogen receptor β-containing neurons in the mouse hypothalamic paraventricular nucleus. J Comp Neurol 2021; 529:2283-2310. [PMID: 33341960 DOI: 10.1002/cne.25093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/17/2022]
Abstract
Within the hypothalamic paraventricular nucleus (PVN), estrogen receptor (ER) β and other gonadal hormone receptors play a role in central cardiovascular processes. However, the influence of sex and age on the cellular and subcellular relationships of ERβ with ERα, G-protein ER (GPER1), as well as progestin and androgen receptors (PR and AR) in the PVN is uncertain. In young (2- to 3-month-old) females and males, ERβ-enhanced green fluorescent protein (EGFP) containing neurons were approximately four times greater than ERα-labeled and PR-labeled nuclei in the PVN. In subdivisions of the PVN, young females, compared to males, had: (1) more ERβ-EGFP neurons in neuroendocrine rostral regions; (2) fewer ERα-labeled nuclei in neuroendocrine and autonomic projecting medial subregions; and (3) more ERα-labeled nuclei in an autonomic projecting caudal region. In contrast, young males, compared to females, had approximately 20 times more AR-labeled nuclei, which often colocalized with ERβ-EGFP in neuroendocrine (approximately 70%) and autonomic (approximately 50%) projecting subregions. Ultrastructurally, in soma and dendrites, PVN ERβ-EGFP colocalized primarily with extranuclear AR (approximately 85% soma) and GPER1 (approximately 70% soma). Aged (12- to 24-month-old) males had more ERβ-EGFP neurons in a rostral neuroendocrine subregion compared to aged females and females with accelerated ovarian failure (AOF) and in a caudal autonomic subregion compared to post-AOF females. Late-aged (18- to 24-month-old) females compared to early-aged (12- to 14-month-old) females and AOF females had fewer AR-labeled nuclei in neuroendrocrine and autonomic projecting subregions. These findings indicate that gonadal steroids may directly and indirectly influence PVN neurons via nuclear and extranuclear gonadal hormone receptors in a sex-specific manner.
Collapse
Affiliation(s)
| | - Sanoara Mazid
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Lily B Goldstein
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - John Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Astrid C Ovalles
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Elizabeth M Waters
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, NY
| | - Michael J Glass
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY.,Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, NY
| |
Collapse
|
30
|
De Pablo-Fernández E, Warner TT. Hypothalamic α-synuclein and its relation to autonomic symptoms and neuroendocrine abnormalities in Parkinson disease. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:223-233. [PMID: 34266594 DOI: 10.1016/b978-0-12-819973-2.00015-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder presenting with defining motor features and a variable combination of nonmotor symptoms. There is growing evidence suggesting that hypothalamic involvement in PD may contribute to the pathogenesis of nonmotor symptoms. Initial neuropathologic studies demonstrated histologic involvement of hypothalamic nuclei by Lewy pathology, i.e., neuronal aggregates including Lewy bodies (round eosinophilic inclusions with a halo found in the neuronal perikarya) and other inclusions in neuronal processes such as Lewy neurites. Recent studies using more sensitive immunohistochemistry have shown that synuclein deposition is common in all hypothalamic nuclei and can happen at preclinical stages of the disease. Several neuropathologic changes, including synuclein deposition, neuronal loss, and adaptative morphologic changes, have been described in neurochemically defined specific hypothalamic cell populations with a potential role in the pathogenesis of nonmotor symptoms such as autonomic dysfunction, blood pressure control, circadian rhythms, sleep, and body weight regulation. The clinical implications of these hypothalamic neuropathologic changes are not fully understood and a direct clinical correlation may be challenging due to the multifactorial pathogenesis of the symptomatology and the additional involvement of other peripheral regulatory mechanisms. Future neuropathologic research using histological and functional assessments should establish the potential role of hypothalamic dysfunction on clinical burden, symptomatic therapies, and disease biomarkers in PD.
Collapse
Affiliation(s)
- Eduardo De Pablo-Fernández
- Reta Lila Weston Institute and Queen Square Brain Bank, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Movement and Clinical Neuroscience, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Thomas T Warner
- Reta Lila Weston Institute and Queen Square Brain Bank, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Movement and Clinical Neuroscience, UCL Queen Square Institute of Neurology, London, United Kingdom.
| |
Collapse
|
31
|
Chaudhary P, Wainford RD. Neuroanatomical characterization of Gαi 2-expressing neurons in the hypothalamic paraventricular nucleus of male and female Sprague-Dawley rats. Physiol Genomics 2021; 53:12-21. [PMID: 33252993 PMCID: PMC7847047 DOI: 10.1152/physiolgenomics.00097.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/26/2020] [Accepted: 11/30/2020] [Indexed: 11/22/2022] Open
Abstract
Hypertension is a global health burden. The hypothalamic paraventricular nucleus (PVN) is an essential component of the neuronal network that regulates sodium homeostasis and blood pressure (BP). Previously, we have shown PVN-specific G protein-coupled receptor-coupled Gαi2 subunit proteins are essential to counter the development of salt-sensitive hypertension by mediating the sympathoinhibitory and natriuretic responses to increased dietary sodium intake to maintain sodium homeostasis and normotension. However, the cellular localization and identity of PVN Gαi2-expressing neurons are currently unknown. In this study using in situ hybridization, we determined the neuroanatomical characterization of Gαi2-expressing PVN neurons in 3-mo-old male and female Sprague-Dawley rats. We observed that Gαi2-expressing neurons containing Gnai2 mRNA are highly localized in the parvocellular region of the hypothalamic PVN. At level 2 of the hypothalamic PVN, Gnai2 mRNA colocalized with ∼ 85% of GABA-expressing neurons and ∼28% of glutamatergic neurons. Additionally, within level 2 Gnai2 mRNA colocalized with ∼75% of corticotrophin-releasing hormone PVN neurons. Gnai2 neurons had lower colocalization with tyrosine hydroxylase (∼33%)-, oxytocin (∼6%)-, and arginine vasopressin (∼10%)-expressing parvocellular neurons in level 2 PVN. Colocalization was similar among male and female rats. The high colocalization of Gnai2 mRNA with GABAergic neurons, in conjunction with our previous findings that PVN Gαi2 proteins mediate sympathoinhibition, suggests that Gαi2 proteins potentially modulate GABAergic signaling to impact sympathetic outflow and BP.
Collapse
Affiliation(s)
- Parul Chaudhary
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
- The Whitaker Cardiovascular Institute, Boston University, Boston, Massachusetts
| | - Richard D Wainford
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
- The Whitaker Cardiovascular Institute, Boston University, Boston, Massachusetts
- Department of Health Sciences, Boston University Sargent College, Boston, Massachusetts
| |
Collapse
|
32
|
Sheppard A, Ralli M, Gilardi A, Salvi R. Occupational Noise: Auditory and Non-Auditory Consequences. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E8963. [PMID: 33276507 PMCID: PMC7729999 DOI: 10.3390/ijerph17238963] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/20/2020] [Accepted: 11/22/2020] [Indexed: 12/13/2022]
Abstract
Occupational noise exposure accounts for approximately 16% of all disabling hearing losses, but the true value and societal costs may be grossly underestimated because current regulations only identify hearing impairments in the workplace if exposures result in audiometric threshold shifts within a limited frequency region. Research over the past several decades indicates that occupational noise exposures can cause other serious auditory deficits such as tinnitus, hyperacusis, extended high-frequency hearing loss, and poor speech perception in noise. Beyond the audiogram, there is growing awareness that hearing loss is a significant risk factor for other debilitating and potentially life-threatening disorders such as cardiovascular disease and dementia. This review discusses some of the shortcomings and limitations of current noise regulations in the United States and Europe.
Collapse
Affiliation(s)
- Adam Sheppard
- Department of Communicative Disorders and Sciences and Center for Hearing and Deafness, University at Buffalo, Buffalo, NY 14221, USA;
| | - Massimo Ralli
- Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy; (M.R.); (A.G.)
| | - Antonio Gilardi
- Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy; (M.R.); (A.G.)
| | - Richard Salvi
- Department of Communicative Disorders and Sciences and Center for Hearing and Deafness, University at Buffalo, Buffalo, NY 14221, USA;
| |
Collapse
|
33
|
Hoshi RA, Santos IS, Dantas EM, Andreão RV, Mill JG, Duncan BB, Schmidt MI, Lotufo PA, Bensenor I. Diabetes and subclinical hypothyroidism on heart rate variability. Eur J Clin Invest 2020; 50:e13349. [PMID: 32654127 DOI: 10.1111/eci.13349] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND We aimed to analyse if the effects of coexistent diabetes and subclinical hypothyroidism extend to the cardio autonomic nervous system, using heart rate variability baseline data from the Brazilian Longitudinal Study of Adult Health. MATERIALS AND METHODS Heart rate variability analyses were performed by linear time and frequency domains in 5-minute time series collected in the supine position. The associations of diabetes and subclinical hypothyroidism with the lowest quartile group for heart rate and the highest quartile group for each heart rate variability parameter were analysed using additive and multiplicative terms in logistic models. For the first approach, the subsample was categorized into four groups: subjects without diabetes and normal thyroid function (controls); subjects without diabetes and subclinical hypothyroidism; patients with diabetes and normal thyroid function; and patients with diabetes and subclinical hypothyroidism. For the interaction alnalysis, diabetes and subclinical hypothyroidism diagnoses were included in separate, along with a multiplicative interaction term between them. RESULTS Point odds ratio estimates for the 4th quartiles of heart rate, and 1st quartiles of all heart rate variability measurements were higher for subjects with combined diabetes and subclinical hypothyroidism than for diabetes only, independently of main sociodemographic and clinical variables (HR: 8.33 vs 2.63; SDNN: 2.59 vs 1.61; RMSSD: 2.37 vs 1.42; LF: 2.83 vs 1.71; HF: 3.06 vs 1.39), but not independently of HbA1c and TSH. Only the interaction term for the association with heart rate, adjusted for sociodemographic and clinical variables, had borderline statistical significance. CONCLUSION Diabetes and subclinical hypothyroidism exert a potential joint impact on cardiac autonomic control, showed by additive effects between diabetes and subclinical hypothyroidism, as well as a significant interaction term for the association with heart rate.
Collapse
Affiliation(s)
- Rosangela A Hoshi
- Center for Clinical and Epidemiological Research of University Hospital, University of Sao Paulo, Sao Paulo, Brazil
| | - Itamar S Santos
- Center for Clinical and Epidemiological Research of University Hospital, University of Sao Paulo, Sao Paulo, Brazil
| | - Eduardo M Dantas
- Department of Biological Sciences, Federal University of Vale do Sao Francisco, Petrolina, Brazil
| | - Rodrigo V Andreão
- Department of Electrical Engineering, Federal Institute of Espirito Santo, Vitória, Brazil
| | - José G Mill
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Brazil
| | - Bruce B Duncan
- Department of Social Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Maria I Schmidt
- Postgraduate Programme in Epidemiology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Paulo A Lotufo
- Center for Clinical and Epidemiological Research of University Hospital, University of Sao Paulo, Sao Paulo, Brazil
| | - Isabela Bensenor
- Center for Clinical and Epidemiological Research of University Hospital, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
34
|
Levenberg K, Hajnal A, George DR, Saunders EFH. Prolonged functional cerebral asymmetry as a consequence of dysfunctional parvocellular paraventricular hypothalamic nucleus signaling: An integrative model for the pathophysiology of bipolar disorder. Med Hypotheses 2020; 146:110433. [PMID: 33317848 DOI: 10.1016/j.mehy.2020.110433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/14/2020] [Accepted: 11/24/2020] [Indexed: 01/09/2023]
Abstract
Approximately 45 million people worldwide are diagnosed with bipolar disorder (BD). While there are many known risk factors and models of the pathologic processes influencing BD, the exact neurologic underpinnings of BD are unknown. We attempt to integrate the existing literature and create a unifying hypothesis regarding the pathophysiology of BD with the hope that a concrete model may potentially facilitate more specific diagnosis, prevention, and treatment of BD in the future. We hypothesize that dysfunctional signaling from the parvocellular neurons of the paraventricular hypothalamic nucleus (PVN) results in the clinical presentation of BD. Functional damage to this nucleus and its signaling pathways may be mediated by myriad factors (e.g. immune dysregulation and auto-immune processes, polygenetic variation, dysfunctional interhemispheric connections, and impaired or overactivated hypothalamic axes) which could help explain the wide variety of clinical presentations along the BD spectrum. The neurons of the PVN regulate ultradian rhythms, which are observed in cyclic variations in healthy individuals, and mediate changes in functional hemispheric lateralization. Theoretically, dysfunctional PVN signaling results in prolonged functional hemispheric dominance. In this model, prolonged right hemispheric dominance leads to depressive symptoms, whereas left hemispheric dominance correlated to the clinical picture of mania. Subsequently, physiologic processes that increase signaling through the PVN (hypothalamic-pituitaryadrenal axis, hypothalamic- pituitary-gonadal axis, and hypothalamic-pituitary-thyroid axis activity, suprachiasmatic nucleus pathways) as well as, neuro-endocrine induced excito-toxicity, auto-immune and inflammatory flairs may induce mood episodes in susceptible individuals. Potentially, ultradian rhythms slowing with age, in combination with changes in hypothalamic axes and maturation of neural circuitry, accounts for BD clinically presenting more frequently in young adulthood than later in life.
Collapse
Affiliation(s)
- Kate Levenberg
- College of Medicine, Penn State University College of Medicine, State College, USA.
| | - Andras Hajnal
- Neural & Behavioral Sciences, Penn State University College of Medicine, State College, USA
| | - Daniel R George
- Department of Humanities, Penn State University College of Medicine, Hershey, USA
| | - Erika F H Saunders
- Psychiatry and Behavioral Health, Penn State University College of Medicine, State College, USA
| |
Collapse
|
35
|
Naguib YW, Yu Y, Wei SG, Morris A, Givens BE, Mekkawy AI, Weiss RM, Felder RB, Salem AK. An Injectable Microparticle Formulation Provides Long-Term Inhibition of Hypothalamic ERK1/2 Activity and Sympathetic Excitation in Rats with Heart Failure. Mol Pharm 2020; 17:3643-3648. [PMID: 32786958 DOI: 10.1021/acs.molpharmaceut.0c00501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Sympathetic excitation contributes to clinical deterioration in systolic heart failure (HF). Significant inhibition of hypothalamic paraventricular nucleus (PVN) ERK1/2 signaling and a subsequent reduction of plasma norepinephrine (NE) levels in HF rats were achieved 2 weeks after a single subcutaneous injection of PD98059-loaded polymeric microparticles, without apparent adverse events, while blank microparticles had no effect. Similar reductions in plasma NE, a general indicator of sympathetic excitation, were previously achieved in HF rats by intracerebroventricular infusion of PD98059 or genetic knockdown of PVN ERK1/2 expression. This study presents a clinically feasible therapeutic approach to the central abnormalities contributing to HF progression.
Collapse
Affiliation(s)
- Youssef W Naguib
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa 52242, United States.,Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Yang Yu
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States
| | - Shun-Guang Wei
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States.,Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States.,Francois M. Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States
| | - Angie Morris
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa 52242, United States
| | - Brittany E Givens
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa 52242, United States.,Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Aml I Mekkawy
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa 52242, United States.,Department of Pharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Sohag University, Sohag 82524, Egypt
| | - Robert M Weiss
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States
| | - Robert B Felder
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States.,Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States.,Francois M. Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States.,Research Service, Veterans Affairs Medical Center, Iowa City, Iowa 52242, United States
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa 52242, United States.,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242, United States
| |
Collapse
|
36
|
Chelban V, Catereniuc D, Aftene D, Gasnas A, Vichayanrat E, Iodice V, Groppa S, Houlden H. An update on MSA: premotor and non-motor features open a window of opportunities for early diagnosis and intervention. J Neurol 2020; 267:2754-2770. [PMID: 32436100 PMCID: PMC7419367 DOI: 10.1007/s00415-020-09881-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 01/27/2023]
Abstract
In this review, we describe the wide clinical spectrum of features that can be seen in multiple system atrophy (MSA) with a focus on the premotor phase and the non-motor symptoms providing an up-to-date overview of the current understanding in this fast-growing field. First, we highlight the non-motor features at disease onset when MSA can be indistinguishable from pure autonomic failure or other chronic neurodegenerative conditions. We describe the progression of clinical features to aid the diagnosis of MSA early in the disease course. We go on to describe the levels of diagnostic certainty and we discuss MSA subtypes that do not fit into the current diagnostic criteria, highlighting the complexity of the disease as well as the need for revised diagnostic tools. Second, we describe the pathology, clinical description, and investigations of cardiovascular autonomic failure, urogenital and sexual dysfunction, orthostatic hypotension, and respiratory and REM-sleep behavior disorders, which may precede the motor presentation by months or years. Their presence at presentation, even in the absence of ataxia and parkinsonism, should be regarded as highly suggestive of the premotor phase of MSA. Finally, we discuss how the recognition of the broader spectrum of clinical features of MSA and especially the non-motor features at disease onset represent a window of opportunity for disease-modifying interventions.
Collapse
Affiliation(s)
- Viorica Chelban
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
- Neurobiology and Medical Genetics Laboratory, "Nicolae Testemitanu" State University of Medicine and Pharmacy, 165, Stefan cel Mare si Sfant Boulevard, 2004, Chişinău, Republic of Moldova.
| | - Daniela Catereniuc
- Neurobiology and Medical Genetics Laboratory, "Nicolae Testemitanu" State University of Medicine and Pharmacy, 165, Stefan cel Mare si Sfant Boulevard, 2004, Chişinău, Republic of Moldova
- Department of Neurology, Epileptology and Internal Diseases, Institute of Emergency Medicine, 1, Toma Ciorba Street, 2004, Chişinău, Republic of Moldova
- Department of Neurology nr. 2, Nicolae Testemitanu" State University of Medicine and Pharmacy, 165, Stefan cel Mare si Sfant Boulevard, 2004, Chişinău, Republic of Moldova
| | - Daniela Aftene
- Department of Neurology, Epileptology and Internal Diseases, Institute of Emergency Medicine, 1, Toma Ciorba Street, 2004, Chişinău, Republic of Moldova
- Department of Neurology nr. 2, Nicolae Testemitanu" State University of Medicine and Pharmacy, 165, Stefan cel Mare si Sfant Boulevard, 2004, Chişinău, Republic of Moldova
| | - Alexandru Gasnas
- Department of Neurology, Epileptology and Internal Diseases, Institute of Emergency Medicine, 1, Toma Ciorba Street, 2004, Chişinău, Republic of Moldova
- Department of Neurology nr. 2, Nicolae Testemitanu" State University of Medicine and Pharmacy, 165, Stefan cel Mare si Sfant Boulevard, 2004, Chişinău, Republic of Moldova
- Cerebrovascular Diseases and Epilepsy Laboratory, Institute of Emergency Medicine, 1, Toma Ciorba Street, 2004, Chişinău, Republic of Moldova
| | - Ekawat Vichayanrat
- Autonomic Unit, National Hospital for Neurology and Neurosurgery, UCL NHS Trust, London, WC1N 3BG, UK
| | - Valeria Iodice
- Autonomic Unit, National Hospital for Neurology and Neurosurgery, UCL NHS Trust, London, WC1N 3BG, UK
| | - Stanislav Groppa
- Neurobiology and Medical Genetics Laboratory, "Nicolae Testemitanu" State University of Medicine and Pharmacy, 165, Stefan cel Mare si Sfant Boulevard, 2004, Chişinău, Republic of Moldova
- Department of Neurology, Epileptology and Internal Diseases, Institute of Emergency Medicine, 1, Toma Ciorba Street, 2004, Chişinău, Republic of Moldova
- Department of Neurology nr. 2, Nicolae Testemitanu" State University of Medicine and Pharmacy, 165, Stefan cel Mare si Sfant Boulevard, 2004, Chişinău, Republic of Moldova
| | - Henry Houlden
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- Neurogenetics Laboratory, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| |
Collapse
|
37
|
Althammer F, Ferreira-Neto HC, Rubaharan M, Roy RK, Patel AA, Murphy A, Cox DN, Stern JE. Three-dimensional morphometric analysis reveals time-dependent structural changes in microglia and astrocytes in the central amygdala and hypothalamic paraventricular nucleus of heart failure rats. J Neuroinflammation 2020; 17:221. [PMID: 32703230 PMCID: PMC7379770 DOI: 10.1186/s12974-020-01892-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cardiovascular diseases, including heart failure, are the most common cause of death globally. Recent studies support a high degree of comorbidity between heart failure and cognitive and mood disorders resulting in memory loss, depression, and anxiety. While neuroinflammation in the hypothalamic paraventricular nucleus contributes to autonomic and cardiovascular dysregulation in heart failure, mechanisms underlying cognitive and mood disorders in this disease remain elusive. The goal of this study was to quantitatively assess markers of neuroinflammation (glial morphology, cytokines, and A1 astrocyte markers) in the central amygdala, a critical forebrain region involved in emotion and cognition, and to determine its time course and correlation to disease severity during the progression of heart failure. METHODS We developed and implemented a comprehensive microglial/astrocyte profiler for precise three-dimensional morphometric analysis of individual microglia and astrocytes in specific brain nuclei at different time points during the progression of heart failure. To this end, we used a well-established ischemic heart failure rat model. Morphometric studies were complemented with quantification of various pro-inflammatory cytokines and A1/A2 astrocyte markers via qPCR. RESULTS We report structural remodeling of central amygdala microglia and astrocytes during heart failure that affected cell volume, surface area, filament length, and glial branches, resulting overall in somatic swelling and deramification, indicative of a change in glial state. These changes occurred in a time-dependent manner, correlated with the severity of heart failure, and were delayed compared to changes in the hypothalamic paraventricular nucleus. Morphometric changes correlated with elevated mRNA levels of pro-inflammatory cytokines and markers of reactive A1-type astrocytes in the paraventricular nucleus and central amygdala during heart failure. CONCLUSION We provide evidence that in addition to the previously described hypothalamic neuroinflammation implicated in sympathohumoral activation during heart failure, microglia, and astrocytes within the central amygdala also undergo structural remodeling indicative of glial shifts towards pro-inflammatory phenotypes. Thus, our studies suggest that neuroinflammation in the amygdala stands as a novel pathophysiological mechanism and potential therapeutic target that could be associated with emotional and cognitive deficits commonly observed at later stages during the course of heart failure.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, USA
| | | | | | - Ranjan K Roy
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, USA
| | - Atit A Patel
- Neuroscience Institute, Georgia State University, Atlanta, USA
| | - Anne Murphy
- Neuroscience Institute, Georgia State University, Atlanta, USA
| | - Daniel N Cox
- Neuroscience Institute, Georgia State University, Atlanta, USA
| | - Javier E Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, USA.
| |
Collapse
|
38
|
Ally A, Powell I, Ally MM, Chaitoff K, Nauli SM. Role of neuronal nitric oxide synthase on cardiovascular functions in physiological and pathophysiological states. Nitric Oxide 2020; 102:52-73. [PMID: 32590118 DOI: 10.1016/j.niox.2020.06.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/15/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022]
Abstract
This review describes and summarizes the role of neuronal nitric oxide synthase (nNOS) on the central nervous system, particularly on brain regions such as the ventrolateral medulla (VLM) and the periaqueductal gray matter (PAG), and on blood vessels and the heart that are involved in the regulation and control of the cardiovascular system (CVS). Furthermore, we shall also review the functional aspects of nNOS during several physiological, pathophysiological, and clinical conditions such as exercise, pain, cerebral vascular accidents or stroke and hypertension. For example, during stroke, a cascade of molecular, neurochemical, and cellular changes occur that affect the nervous system as elicited by generation of free radicals and nitric oxide (NO) from vulnerable neurons, peroxide formation, superoxides, apoptosis, and the differential activation of three isoforms of nitric oxide synthases (NOSs), and can exert profound effects on the CVS. Neuronal NOS is one of the three isoforms of NOSs, the others being endothelial (eNOS) and inducible (iNOS) enzymes. Neuronal NOS is a critical homeostatic component of the CVS and plays an important role in regulation of different systems and disease process including nociception. The functional and physiological roles of NO and nNOS are described at the beginning of this review. We also elaborate the structure, gene, domain, and regulation of the nNOS protein. Both inhibitory and excitatory role of nNOS on the sympathetic autonomic nervous system (SANS) and parasympathetic autonomic nervous system (PANS) as mediated via different neurotransmitters/signal transduction processes will be explored, particularly its effects on the CVS. Because the VLM plays a crucial function in cardiovascular homeostatic mechanisms, the neuroanatomy and cardiovascular regulation of the VLM will be discussed in conjunction with the actions of nNOS. Thereafter, we shall discuss the up-to-date developments that are related to the interaction between nNOS and cardiovascular diseases such as hypertension and stroke. Finally, we shall focus on the role of nNOS, particularly within the PAG in cardiovascular regulation and neurotransmission during different types of pain stimulus. Overall, this review focuses on our current understanding of the nNOS protein, and provides further insights on how nNOS modulates, regulates, and controls cardiovascular function during both physiological activity such as exercise, and pathophysiological conditions such as stroke and hypertension.
Collapse
Affiliation(s)
- Ahmmed Ally
- Arkansas College of Osteopathic Medicine, Fort Smith, AR, USA.
| | - Isabella Powell
- All American Institute of Medical Sciences, Black River, Jamaica
| | | | - Kevin Chaitoff
- Interventional Rehabilitation of South Florida, West Palm Beach, FL, USA
| | - Surya M Nauli
- Chapman University and University of California, Irvine, CA, USA.
| |
Collapse
|
39
|
Chen X, Xin N, Pan Y, Zhu L, Yin P, Liu Q, Yang W, Xu X, Li S, Li XJ. Huntingtin-Associated Protein 1 in Mouse Hypothalamus Stabilizes Glucocorticoid Receptor in Stress Response. Front Cell Neurosci 2020; 14:125. [PMID: 32581713 PMCID: PMC7289054 DOI: 10.3389/fncel.2020.00125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
Huntingtin-associated protein 1 (Hap1) was initially identified as a brain-enriched protein that binds to the Huntington’s disease protein, huntingtin. Unlike huntingtin that is ubiquitously expressed in the brain, Hap1 is enriched in the brain with the highest expression level in the hypothalamus. The selective enrichment of Hap1 in the hypothalamus suggests that Hap1 may play a specific role in hypothalamic function that can regulate metabolism and stress response. Here we report that Hap1 is colocalized and interacts with the glucocorticoid receptor (GR) in mouse hypothalamic neurons. Genetic depletion of Hap1 reduced the expression level of GR in the hypothalamus. Dexamethasone, a GR agonist, treatment or fasting of mice induced stress, resulting in increased expression of Hap1 in the hypothalamus. However, when Hap1 was absent, these treatments promoted GR reduction in the hypothalamus. In cultured cells, loss of Hap1 shortened the half-life of GR. These findings suggest that Hap1 stabilizes GR in the cytoplasm and that Hap1 dysfunction or deficiency may alter animal’s stress response.
Collapse
Affiliation(s)
- Xingxing Chen
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, China.,Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China.,Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| | - Ning Xin
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yongcheng Pan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States.,Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Louyin Zhu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| | - Peng Yin
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Qiong Liu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States.,Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Weili Yang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Xingshun Xu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shihua Li
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Xiao-Jiang Li
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| |
Collapse
|
40
|
Japundžić-Žigon N, Lozić M, Šarenac O, Murphy D. Vasopressin & Oxytocin in Control of the Cardiovascular System: An Updated Review. Curr Neuropharmacol 2020; 18:14-33. [PMID: 31544693 PMCID: PMC7327933 DOI: 10.2174/1570159x17666190717150501] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/03/2019] [Accepted: 07/06/2019] [Indexed: 01/19/2023] Open
Abstract
Since the discovery of vasopressin (VP) and oxytocin (OT) in 1953, considerable knowledge has been gathered about their roles in cardiovascular homeostasis. Unraveling VP vasoconstrictor properties and V1a receptors in blood vessels generated powerful hemostatic drugs and drugs effective in the treatment of certain forms of circulatory collapse (shock). Recognition of the key role of VP in water balance via renal V2 receptors gave birth to aquaretic drugs found to be useful in advanced stages of congestive heart failure. There are still unexplored actions of VP and OT on the cardiovascular system, both at the periphery and in the brain that may open new venues in treatment of cardiovascular diseases. After a brief overview on VP, OT and their peripheral action on the cardiovascular system, this review focuses on newly discovered hypothalamic mechanisms involved in neurogenic control of the circulation in stress and disease.
Collapse
Affiliation(s)
| | - Maja Lozić
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Olivera Šarenac
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - David Murphy
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
41
|
Akbari A, Jelodar G. Cardiovascular responses produced by resistin injected into paraventricular nucleus mediated by the glutamatergic and CRFergic transmissions within rostral ventrolateral medulla. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:344-353. [PMID: 32440321 PMCID: PMC7229507 DOI: 10.22038/ijbms.2019.40316.9547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 09/23/2019] [Indexed: 11/24/2022]
Abstract
OBJECTIVES Resistin, as a 12.5 kDa cysteine-rich polypeptide, is expressed in hypothalamus and regulates sympathetic nerve activity. It is associated with obesity, metabolic syndrome and cardiovascular diseases. In this study, we investigated the neural pathway of cardiovascular responses induced by injection of resistin into paraventricular nucleus (PVN) with rostral ventrolateral medulla (RVLM). MATERIALS AND METHODS Adult male rats were anesthetized with urethane (1.4 g/kg intraperitoneally). Resistin (3 µg/1 µl/rat) was first injected into PVN, and the glutamatergic, corticotrophin-releasing factor (CRF)-ergic and angiotensinogenic transmission was inhibited by injecting of their antagonist in RVLM. Arterial pressure (AP) and heart rate (HR) were monitored before and after the injection. RESULTS The results showed that resistin injection into PVN significantly increased AP and HR compared to control group and prior to its injection (P<0.05). Injection of AP5 ((2R)-amino-5-phosphonovaleric acid; (2R)-amino-5-phosphonopentanoate) (50 nM/rat), losartan (10 nM/rat) and astressin (50 nM/rat) into RVLM reduced cardiovascular responses produced by injected resistin into PVN. Injection of AP5+losartan or astressin+losartan or astressin+AP5 into RVLM could significantly reduce cardiovascular responses produced by resistin compared to before injection (P<0.05). Furthermore, the depressor responses generated by AP5+losartan injected into RVLM were significantly stronger than the depressor responses generated by AP5+astressin and/or astressin+losartan injected into RVLM (P<0.05). CONCLUSION It can be concluded that glutamatergic and CRFergic transmissions have crucial contribution to cardiovascular responses produced by resistin. The results provided new and potentially important insight regarding neural transmission when the plasma level of resistin increases; this reveals the role of resistin in cardiovascular responses such as metabolic syndrome and hypertension.
Collapse
Affiliation(s)
- Abolfazl Akbari
- Department of Physiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Gholamali Jelodar
- Department of Physiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
42
|
Ovalles AC, Contoreggi NH, Marques-Lopes J, Van Kempen TA, Iadecola C, Waters EM, Glass MJ, Milner TA. Plasma Membrane Affiliated AMPA GluA1 in Estrogen Receptor β-containing Paraventricular Hypothalamic Neurons Increases Following Hypertension in a Mouse Model of Post-menopause. Neuroscience 2019; 423:192-205. [PMID: 31682817 DOI: 10.1016/j.neuroscience.2019.09.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/11/2022]
Abstract
Sex and ovarian function contribute to hypertension susceptibility, however, the mechanisms are not well understood. Prior studies show that estrogens and neurogenic factors, including hypothalamic glutamatergic NMDA receptor plasticity, play significant roles in rodent hypertension. Here, we investigated the role of sex and ovarian failure on AMPA receptor plasticity in estrogen-sensitive paraventricular nucleus (PVN) neurons in naïve and angiotensin II (AngII) infused male and female mice and female mice at early and late stages of accelerated ovarian failure (AOF). High-resolution electron microscopy was used to assess the subcellular distribution of AMPA GluA1 in age-matched male and female estrogen receptor beta (ERβ) enhanced green fluorescent protein (EGFP) reporter mice as well as female ERβ-EGFP mice treated with 4-vinylcyclohexene diepoxide. In the absence of AngII, female mice at a late stage of AOF displayed higher levels of GluA1 on the plasma membrane, indicative of functional protein, in ERβ-expressing PVN dendrites when compared to male, naïve female and early stage AOF mice. Following slow-pressor AngII infusion, males, as well as early and late stage AOF females had elevated blood pressure. Significantly, only late stage-AOF female mice infused with AngII had an increase in GluA1 near the plasma membrane in dendrites of ERβ-expressing PVN neurons. In contrast, prior studies reported that plasmalemmal NMDA GluN1 increased in ERβ-expressing PVN dendrites in males and early, but not late stage AOF females. Together, these findings reveal that early and late stage AOF female mice display unique molecular signatures of long-lasting synaptic strength prior to, and following hypertension.
Collapse
Affiliation(s)
- Astrid C Ovalles
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Jose Marques-Lopes
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Tracey A Van Kempen
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Elizabeth M Waters
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Michael J Glass
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA.
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
43
|
Effects of needling acupoints at different nerve segments on oxytocin neurons in rat’s hypothalamic paraventricular nucleus and intragastric pressure. JOURNAL OF ACUPUNCTURE AND TUINA SCIENCE 2019. [DOI: 10.1007/s11726-019-1128-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
44
|
Aggressive behavior and brain neuronal activation in sexually naïve male Mongolian gerbils. Behav Brain Res 2019; 378:112276. [PMID: 31589893 DOI: 10.1016/j.bbr.2019.112276] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/25/2019] [Accepted: 10/01/2019] [Indexed: 12/18/2022]
Abstract
Aggressive behavior plays an important role in animal's survival and reproductive success. Although there has been growing interests in studying neural mechanisms underlying aggressive behavior using traditional laboratory animal models, little is known about mechanisms controlling naturally occurring aggression in sexually naïve animals. In the present study, we characterized aggressive behavior displayed by sexually naïve male Mongolian gerbils (Meriones unguiculatus) and examined the subsequent neuronal activation in the brain measured by Fos-immunoreactive (Fos-ir) staining. We found that resident males initiated attacks and showed intense levels of aggression (including chase, bite, offensive sideway, lunge and on-top) towards a conspecific male intruder. Furthermore, attacks from the resident males towards the intruder produced a nonrandom distribution of bites, with the most on the rump, flank, back and tail and few on the limbs, ventrum and head. In contrast, control males that were exposed to a woodblock (control for novelty) never attacked the woodblock and showed higher levels of object/environmental investigation. Male gerbils exposed to an intruder had significantly higher levels of Fos-ir density in the medial (MeA) and anterior cortical (ACo) subnuclei of the amygdala, principal nucleus (BSTpr) and interfascicular nucleus (BSTif) of the bed nucleus of the stria terminalis, ventrolateral subdivision of the ventromedial hypothalamus (VMHvl), and paraventricular nucleus of the hypothalamus (PVN), compared to control males. Together, our results indicate that sexually naïve, group housed male gerbils naturally display aggression towards conspecific strangers, and such aggressive behavior is associated with special patterns of neuronal activation in the brain.
Collapse
|
45
|
Stofkova A, Murakami M. Neural activity regulates autoimmune diseases through the gateway reflex. Bioelectron Med 2019; 5:14. [PMID: 32232103 PMCID: PMC7098223 DOI: 10.1186/s42234-019-0030-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022] Open
Abstract
The brain, spinal cord and retina are protected from blood-borne compounds by the blood-brain barrier (BBB), blood-spinal cord barrier (BSCB) and blood-retina barrier (BRB) respectively, which create a physical interface that tightly controls molecular and cellular transport. The mechanical and functional integrity of these unique structures between blood vessels and nervous tissues is critical for maintaining organ homeostasis. To preserve the stability of these barriers, interplay between constituent barrier cells, such as vascular endothelial cells, pericytes, glial cells and neurons, is required. When any of these cells are defective, the barrier can fail, allowing blood-borne compounds to encroach neural tissues and cause neuropathologies. Autoimmune diseases of the central nervous system (CNS) and retina are characterized by barrier disruption and the infiltration of activated immune cells. Here we review our recent findings on the role of neural activity in the regulation of these barriers at the vascular endothelial cell level in the promotion of or protection against the development of autoimmune diseases. We suggest nervous system reflexes, which we named gateway reflexes, are fundamentally involved in these diseases. Although their reflex arcs are not completely understood, we identified the activation of specific sensory neurons or receptor cells to which barrier endothelial cells respond as effectors that regulate gateways for immune cells to enter the nervous tissue. We explain this novel mechanism and describe its role in neuroinflammatory conditions, including models of multiple sclerosis and posterior autoimmune uveitis.
Collapse
Affiliation(s)
- Andrea Stofkova
- 1Department of Physiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
- 2Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-0815 Japan
| | - Masaaki Murakami
- 2Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-0815 Japan
| |
Collapse
|
46
|
Baschieri F, Cortelli P. Circadian rhythms of cardiovascular autonomic function: Physiology and clinical implications in neurodegenerative diseases. Auton Neurosci 2019; 217:91-101. [DOI: 10.1016/j.autneu.2019.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/31/2019] [Accepted: 01/31/2019] [Indexed: 12/11/2022]
|
47
|
Dampney RA, Michelini LC, Li DP, Pan HL. Regulation of sympathetic vasomotor activity by the hypothalamic paraventricular nucleus in normotensive and hypertensive states. Am J Physiol Heart Circ Physiol 2018; 315:H1200-H1214. [PMID: 30095973 PMCID: PMC6297824 DOI: 10.1152/ajpheart.00216.2018] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/13/2018] [Accepted: 07/25/2018] [Indexed: 12/22/2022]
Abstract
The hypothalamic paraventricular nucleus (PVN) is a unique and important brain region involved in the control of cardiovascular, neuroendocrine, and other physiological functions pertinent to homeostasis. The PVN is a major source of excitatory drive to the spinal sympathetic outflow via both direct and indirect projections. In this review, we discuss the role of the PVN in the regulation of sympathetic output in normal physiological conditions and in hypertension. In normal healthy animals, the PVN presympathetic neurons do not appear to have a major role in sustaining resting sympathetic vasomotor activity or in regulating sympathetic responses to short-term homeostatic challenges such as acute hypotension or hypoxia. Their role is, however, much more significant during longer-term challenges, such as sustained water deprivation, chronic intermittent hypoxia, and pregnancy. The PVN also appears to have a major role in generating the increased sympathetic vasomotor activity that is characteristic of multiple forms of hypertension. Recent studies in the spontaneously hypertensive rat model have shown that impaired inhibitory and enhanced excitatory synaptic inputs to PVN presympathetic neurons are the basis for the heightened sympathetic outflow in hypertension. We discuss the molecular mechanisms underlying the presynaptic and postsynaptic alterations in GABAergic and glutamatergic inputs to PVN presympathetic neurons in hypertension. In addition, we discuss the ability of exercise training to correct sympathetic hyperactivity by restoring blood-brain barrier integrity, reducing angiotensin II availability, and decreasing oxidative stress and inflammation in the PVN.
Collapse
Affiliation(s)
- Roger A Dampney
- Department of Physiology, University of Sydney , Sydney, New South Wales , Australia
| | - Lisete C Michelini
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo , São Paulo , Brazil
| | - De-Pei Li
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center , Houston, Texas
| | - Hui-Lin Pan
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center , Houston, Texas
| |
Collapse
|
48
|
Ruyle BC, Klutho PJ, Baines CP, Heesch CM, Hasser EM. Hypoxia activates a neuropeptidergic pathway from the paraventricular nucleus of the hypothalamus to the nucleus tractus solitarii. Am J Physiol Regul Integr Comp Physiol 2018; 315:R1167-R1182. [PMID: 30230933 DOI: 10.1152/ajpregu.00244.2018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The paraventricular nucleus of the hypothalamus (PVN) contributes to both autonomic and neuroendocrine function. PVN lesion or inhibition blunts cardiorespiratory responses to peripheral chemoreflex activation, suggesting that the PVN is required for full expression of these effects. However, the role of efferent projections to cardiorespiratory nuclei and the neurotransmitters/neuromodulators that are involved is unclear. The PVN sends dense projections to the nucleus tractus solitarii (nTS), a region that displays neuronal activation following hypoxia. We hypothesized that acute hypoxia activates nTS-projecting PVN neurons. Using a combination of retrograde tracing and immunohistochemistry, we determined whether hypoxia activates PVN neurons that project to the nTS and examined the phenotype of these neurons. Conscious rats underwent 2 h normoxia (21% O2, n = 5) or hypoxia (10% O2, n = 6). Hypoxia significantly increased Fos immunoreactivity in nTS-projecting neurons, primarily in the caudal PVN. The majority of activated nTS-projecting neurons contained corticotropin-releasing hormone (CRH). In the nTS, fibers expressing the CRH receptor corticotropin-releasing factor receptor 2 (CRFR2) were colocalized with oxytocin (OT) fibers and were closely associated with hypoxia-activated nTS neurons. A separate group of animals that received a microinjection of adeno-associated virus type 2-hSyn-green fluorescent protein (GFP) into the PVN exhibited GFP-expressing fibers in the nTS; a proportion of these fibers displayed OT immunoreactivity. Thus, nTS CRFR2s appear to be located on the fibers of PVN OT neurons that project to the nTS. Taken together, our findings suggest that PVN CRH projections to the nTS may modulate nTS neuronal activation, possibly via OTergic mechanisms, and thus contribute to chemoreflex cardiorespiratory responses.
Collapse
Affiliation(s)
- Brian C Ruyle
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Paula J Klutho
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri , Columbia, Missouri
| | - Christopher P Baines
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri , Columbia, Missouri.,Department of Medical Pharmacology and Physiology, University of Missouri , Columbia, Missouri
| | - Cheryl M Heesch
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri , Columbia, Missouri
| | - Eileen M Hasser
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri , Columbia, Missouri.,Department of Medical Pharmacology and Physiology, University of Missouri , Columbia, Missouri
| |
Collapse
|
49
|
Role of Trpv1 and Trpv4 in surgical incision-induced tissue swelling and Fos-like immunoreactivity in the central nervous system of mice. Neurosci Lett 2018; 678:76-82. [PMID: 29733975 DOI: 10.1016/j.neulet.2018.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/05/2018] [Accepted: 05/01/2018] [Indexed: 11/24/2022]
Abstract
Pain management remains a major concern regarding the treatment of postoperative patients. Transient receptor potential (TRP) channels are considered to be new therapeutic targets for pain control. We investigated whether the genes Trpv1 and Trpv4 are involved in hind paw swelling caused after surgical incision in mice or in incision-induced Fos-like immunoreactivity (Fos-LI) levels in the central nervous system. Mice were divided into four groups: wild-type (WT) control, WT incision, Trpv1 knockout (Trpv1-/-) incision, and Trpv4 knockout (Trpv4-/-) incision. Mice were anesthetized, and only those in the incision, and not control, groups received a surgical incision to their right plantar hind paw. Changes in paw diameter and in Fos-LI levels in the dorsal horn of the spinal cord, paraventricular nucleus of the hypothalamus (PVN), paraventricular nucleus of the thalamus, and central amygdala were evaluated 2 h after the incision. There was no significant difference in the paw diameter among groups. In contrast, in laminae I-II of the dorsal horn of the spinal cord and PVN, Fos-LI was significantly higher in all incision groups than in the WT control group. A significant increase in Fos-positive cells was also observed in the dorsal horn laminae III-IV in Trpv1-/- and Trpv4-/- incision groups compared with the WT incision group. Our results indicate that surgical incision activates the PVN and that Trpv1 and Trpv4 might be involved in neuronal activity in the dorsal horn laminae III-IV after surgical incision.
Collapse
|
50
|
Coon EA, Cutsforth-Gregory JK, Benarroch EE. Neuropathology of autonomic dysfunction in synucleinopathies. Mov Disord 2018; 33:349-358. [DOI: 10.1002/mds.27186] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/30/2017] [Accepted: 09/10/2017] [Indexed: 12/16/2022] Open
|