1
|
Tsai J, Malik S, Tjen-A-Looi SC. Pulmonary Hypertension: Pharmacological and Non-Pharmacological Therapies. Life (Basel) 2024; 14:1265. [PMID: 39459565 PMCID: PMC11509317 DOI: 10.3390/life14101265] [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: 08/31/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Pulmonary hypertension (PH) is a severe and chronic disease characterized by increased pulmonary vascular resistance and remodeling, often precipitating right-sided heart dysfunction and death. Although the condition is progressive and incurable, current therapies for the disease focus on multiple different drugs and general supportive therapies to manage symptoms and prolong survival, ranging from medications more specific to pulmonary arterial hypertension (PAH) to exercise training. Moreover, there are multiple studies exploring novel experimental drugs and therapies including unique neurostimulation, to help better manage the disease. Here, we provide a narrative review focusing on current PH treatments that target multiple underlying biochemical mechanisms, including imbalances in vasoconstrictor-vasodilator and autonomic nervous system function, inflammation, and bone morphogenic protein (BMP) signaling. We also focus on the potential of novel therapies for managing PH, focusing on multiple types of neurostimulation including acupuncture. Lastly, we also touch upon the disease's different subgroups, clinical presentations and prognosis, diagnostics, demographics, and cost.
Collapse
Affiliation(s)
- Jason Tsai
- Susan Samueli Integrative Health Institute, College of Health Sciences, University of California-Irvine, Irvine, CA 92617, USA;
| | | | - Stephanie C. Tjen-A-Looi
- Susan Samueli Integrative Health Institute, College of Health Sciences, University of California-Irvine, Irvine, CA 92617, USA;
| |
Collapse
|
2
|
de Barcellos Filho PG, Dantzler HA, Hasser EM, Kline DD. Oxytocin and corticotropin-releasing hormone exaggerate nucleus tractus solitarii neuronal and synaptic activity following chronic intermittent hypoxia. J Physiol 2024; 602:3375-3400. [PMID: 38698722 PMCID: PMC11251298 DOI: 10.1113/jp286069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/28/2024] [Indexed: 05/05/2024] Open
Abstract
Chronic intermittent hypoxia (CIH) in rodents mimics the hypoxia-induced elevation of blood pressure seen in individuals experiencing episodic breathing. The brainstem nucleus tractus solitarii (nTS) is the first site of visceral sensory afferent integration, and thus is critical for cardiorespiratory homeostasis and its adaptation during a variety of stressors. In addition, the paraventricular nucleus of the hypothalamus (PVN), in part through its nTS projections that contain oxytocin (OT) and/or corticotropin-releasing hormone (CRH), contributes to cardiorespiratory regulation. Within the nTS, these PVN-derived neuropeptides alter nTS activity and the cardiorespiratory response to hypoxia. Nevertheless, their contribution to nTS activity after CIH is not fully understood. We hypothesized that OT and CRH would increase nTS activity to a greater extent following CIH, and co-activation of OT+CRH receptors would further magnify nTS activity. Our data show that compared to their normoxic controls, 10 days' CIH exaggerated nTS discharge, excitatory synaptic currents and Ca2+ influx in response to CRH, which were further enhanced by the addition of OT. CIH increased the tonic functional contribution of CRH receptors, which occurred with elevation of mRNA and protein. Together, our data demonstrate that intermittent hypoxia exaggerates the expression and function of neuropeptides on nTS activity. KEY POINTS: Episodic breathing and chronic intermittent hypoxia (CIH) are associated with autonomic dysregulation, including elevated sympathetic nervous system activity. Altered nucleus tractus solitarii (nTS) activity contributes to this response. Neurons originating in the paraventricular nucleus (PVN), including those containing oxytocin (OT) and corticotropin-releasing hormone (CRH), project to the nTS, and modulate the cardiorespiratory system. Their role in CIH is unknown. In this study, we focused on OT and CRH individually and together on nTS activity from rats exposed to either CIH or normoxia control. We show that after CIH, CRH alone and with OT increased to a greater extent overall nTS discharge, neuronal calcium influx, synaptic transmission to second-order nTS neurons, and OT and CRH receptor expression. These results provide insights into the underlying circuits and mechanisms contributing to autonomic dysfunction during periods of episodic breathing.
Collapse
Affiliation(s)
- Procopio Gama de Barcellos Filho
- Department of Biomedical Sciences, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Dalton Cardiovascular Research Center, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
| | - Heather A. Dantzler
- Department of Biomedical Sciences, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Dalton Cardiovascular Research Center, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
| | - Eileen M. Hasser
- Department of Biomedical Sciences, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Dalton Cardiovascular Research Center, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
| | - David D. Kline
- Department of Biomedical Sciences, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Dalton Cardiovascular Research Center, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
| |
Collapse
|
3
|
Zhou J, Zhang B, Zhou X, Zhang F, Shu Q, Wu Y, Chang HM, Hu L, Cai RL, Yu Q. Electroacupuncture pretreatment mediates sympathetic nerves to alleviate myocardial ischemia-reperfusion injury via CRH neurons in the paraventricular nucleus of the hypothalamus. Chin Med 2024; 19:43. [PMID: 38448912 PMCID: PMC10916233 DOI: 10.1186/s13020-024-00916-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/26/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Myocardial ischemia-reperfusion can further exacerbate myocardial injury and increase the risk of death. Our previous research found that the paraventricular nucleus (PVN) of the hypothalamus plays a crucial role in the improvement of myocardial ischemia-reperfusion injury (MIRI) by electroacupuncture (EA) pretreatment, but its mechanism of action is still unclear. CRH neurons exhibit periodic concentrated expression in PVN, but further research is needed to determine whether they are involved in the improvement of MIRI by EA pretreatment. Meanwhile, numerous studies have shown that changes in sympathetic nervous system innervation and activity are associated with many heart diseases. This study aims to investigate whether EA pretreatment improves MIRI through sympathetic nervous system mediated by PVNCRH neurons. METHODS Integrated use of fiber-optic recording, chemical genetics and other methods to detect relevant indicators: ECG signals were acquired through Powerlab standard II leads, and LabChart 8 calculated heart rate, ST-segment offset, and heart rate variability (HRV); Left ventricular ejection fraction (LVEF), left ventricular short-axis shortening (LVFS), left ventricular end-systolic internal diameter (LVIDs) and interventricular septal thickness (IVSs) were measured by echocardiography; Myocardial infarct area (IA) and area at risk (AAR) were calculated by Evans-TTC staining. Pathological changes in cardiomyocytes were observed by HE staining; Changes in PVNCRH neuronal activity were recorded by fiber-optic photometry; Sympathetic nerve discharges were recorded for in vivo electrophysiology; NE and TH protein expression was assayed by Western blot. RESULTS Our data indicated that EA pretreatment can effectively alleviate MIRI. Meanwhile, we found that in the MIRI model, the number and activity of CRH neurons co labeled with c-Fos in the PVN area of the rat brain increased, and the frequency of sympathetic nerve discharge increased. EA pretreatment could reverse this change. In addition, the results of chemical genetics indicated that inhibiting PVNCRH neurons has a similar protective effect on MIRI as EA pretreatment, and the activation of PVNCRH neurons can counteract this protective effect. CONCLUSION EA pretreatment can inhibit PVNCRH neurons and improve MIRI by inhibiting sympathetic nerve, which offers fresh perspectives on the application of acupuncture in the management of cardiovascular disease.
Collapse
Affiliation(s)
- Jie Zhou
- College of Acupuncture and Moxibustion, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Bin Zhang
- College of Acupuncture and Moxibustion, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Xiang Zhou
- College of Acupuncture and Moxibustion, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Fan Zhang
- College of Acupuncture and Moxibustion, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Qi Shu
- College of Acupuncture and Moxibustion, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Yan Wu
- College of Acupuncture and Moxibustion, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Hui-Min Chang
- College of Acupuncture and Moxibustion, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Ling Hu
- Institute of Acupuncture and Meridian Research, Anhui Academy of Chinese Medicine, Hefei, 230038, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230038, China
| | - Rong-Lin Cai
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Insitute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, China.
- Institute of Acupuncture and Meridian Research, Anhui Academy of Chinese Medicine, Hefei, 230038, China.
- Anhui Province Key Laboratory of Meridian Viscera Correlationship, Hefei, 230038, China.
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230038, China.
| | - Qing Yu
- Institute of Acupuncture and Meridian Research, Anhui Academy of Chinese Medicine, Hefei, 230038, China.
- Anhui Province Key Laboratory of Meridian Viscera Correlationship, Hefei, 230038, China.
| |
Collapse
|
4
|
Pace SA, Myers B. Hindbrain Adrenergic/Noradrenergic Control of Integrated Endocrine and Autonomic Stress Responses. Endocrinology 2023; 165:bqad178. [PMID: 38015813 DOI: 10.1210/endocr/bqad178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/07/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Hindbrain adrenergic/noradrenergic nuclei facilitate endocrine and autonomic responses to physical and psychological challenges. Neurons that synthesize adrenaline and noradrenaline target hypothalamic structures to modulate endocrine responses while descending spinal projections regulate sympathetic function. Furthermore, these neurons respond to diverse stress-related metabolic, autonomic, and psychosocial challenges. Accordingly, adrenergic and noradrenergic nuclei are integrative hubs that promote physiological adaptation to maintain homeostasis. However, the precise mechanisms through which adrenaline- and noradrenaline-synthesizing neurons sense interoceptive and exteroceptive cues to coordinate physiological responses have yet to be fully elucidated. Additionally, the regulatory role of these cells in the context of chronic stress has received limited attention. This mini-review consolidates reports from preclinical rodent studies on the organization and function of brainstem adrenaline and noradrenaline cells to provide a framework for how these nuclei coordinate endocrine and autonomic physiology. This includes identification of hindbrain adrenaline- and noradrenaline-producing cell groups and their role in stress responding through neurosecretory and autonomic engagement. Although temporally and mechanistically distinct, the endocrine and autonomic stress axes are complementary and interconnected. Therefore, the interplay between brainstem adrenergic/noradrenergic nuclei and peripheral physiological systems is necessary for integrated stress responses and organismal survival.
Collapse
Affiliation(s)
- Sebastian A Pace
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Brent Myers
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
5
|
Ruyle BC, Lima-Silveira L, Martinez D, Cummings KJ, Heesch CM, Kline DD, Hasser EM. Paraventricular nucleus projections to the nucleus tractus solitarii are essential for full expression of hypoxia-induced peripheral chemoreflex responses. J Physiol 2023; 601:4309-4336. [PMID: 37632733 DOI: 10.1113/jp284907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/13/2023] [Indexed: 08/28/2023] Open
Abstract
The hypothalamic paraventricular nucleus (PVN) is essential to peripheral chemoreflex neurocircuitry, but the specific efferent pathways utilized are not well defined. The PVN sends dense projections to the nucleus tractus solitarii (nTS), which exhibits neuronal activation following a hypoxic challenge. We hypothesized that nTS-projecting PVN (PVN-nTS) neurons contribute to hypoxia-induced nTS neuronal activation and cardiorespiratory responses. To selectively target PVN-nTS neurons, rats underwent bilateral nTS nanoinjection of retrogradely transported adeno-associated virus (AAV) driving Cre recombinase expression. We then nanoinjected into PVN AAVs driving Cre-dependent expression of Gq or Gi designer receptors exclusively activated by designer drugs (DREADDs) to test the degree that selective activation or inhibition, respectively, of the PVN-nTS pathway affects the hypoxic ventilatory response (HVR) of conscious rats. We used immunohistochemistry for Fos and extracellular recordings to examine how DREADD activation influences PVN-nTS neuronal activation by hypoxia. Pathway activation enhanced the HVR at moderate hypoxic intensities and increased PVN and nTS Fos immunoreactivity in normoxia and hypoxia. In contrast, PVN-nTS inhibition reduced both the HVR and PVN and nTS neuronal activation following hypoxia. To further confirm selective pathway effects on central cardiorespiratory output, rats underwent hypoxia before and after bilateral nTS nanoinjections of C21 to activate or inhibit PVN-nTS terminals. PVN terminal activation within the nTS enhanced tachycardic, sympathetic and phrenic (PhrNA) nerve activity responses to hypoxia whereas inhibition attenuated hypoxia-induced increases in nTS neuronal action potential discharge and PhrNA. The results demonstrate the PVN-nTS pathway enhances nTS neuronal activation and is necessary for full cardiorespiratory responses to hypoxia. KEY POINTS: The hypothalamic paraventricular nucleus (PVN) contributes to peripheral chemoreflex cardiorespiratory responses, but specific PVN efferent pathways are not known. The nucleus tractus solitarii (nTS) is the first integration site of the peripheral chemoreflex, and the nTS receives dense projections from the PVN. Selective GqDREADD activation of the PVN-nTS pathway was shown to enhance ventilatory responses to hypoxia and activation (Fos immunoreactivity (IR)) of nTS neurons in conscious rats, augmenting the sympathetic and phrenic nerve activity (SSNA and PhrNA) responses to hypoxia in anaesthetized rats. Selective GiDREADD inhibition of PVN-nTS neurons attenuates ventilatory responses, nTS neuronal Fos-IR, action potential discharge and PhrNA responses to hypoxia. These results demonstrate that a projection from the PVN to the nTS is critical for full chemoreflex responses to hypoxia.
Collapse
Affiliation(s)
- Brian C Ruyle
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
| | - Ludmila Lima-Silveira
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Diana Martinez
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Kevin J Cummings
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Cheryl M Heesch
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - David D Kline
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Eileen M Hasser
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
6
|
Jun S, Ou X, Shi L, Yu H, Deng T, Chen J, Nie X, Hao Y, Shi Y, Liu W, Tian Y, Wang S, Yuan F. Circuit-Specific Control of Blood Pressure by PNMT-Expressing Nucleus Tractus Solitarii Neurons. Neurosci Bull 2023; 39:1193-1209. [PMID: 36588135 PMCID: PMC10387028 DOI: 10.1007/s12264-022-01008-3] [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: 08/21/2022] [Accepted: 10/18/2022] [Indexed: 01/03/2023] Open
Abstract
The nucleus tractus solitarii (NTS) is one of the morphologically and functionally defined centers that engage in the autonomic regulation of cardiovascular activity. Phenotypically-characterized NTS neurons have been implicated in the differential regulation of blood pressure (BP). Here, we investigated whether phenylethanolamine N-methyltransferase (PNMT)-expressing NTS (NTSPNMT) neurons contribute to the control of BP. We demonstrate that photostimulation of NTSPNMT neurons has variable effects on BP. A depressor response was produced during optogenetic stimulation of NTSPNMT neurons projecting to the paraventricular nucleus of the hypothalamus, lateral parabrachial nucleus, and caudal ventrolateral medulla. Conversely, photostimulation of NTSPNMT neurons projecting to the rostral ventrolateral medulla produced a robust pressor response and bradycardia. In addition, genetic ablation of both NTSPNMT neurons and those projecting to the rostral ventrolateral medulla impaired the arterial baroreflex. Overall, we revealed the neuronal phenotype- and circuit-specific mechanisms underlying the contribution of NTSPNMT neurons to the regulation of BP.
Collapse
Affiliation(s)
- Shirui Jun
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xianhong Ou
- Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Luo Shi
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Hongxiao Yu
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Tianjiao Deng
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jinting Chen
- Core Facilities and Centers, Institute of Medicine and Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xiaojun Nie
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yinchao Hao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yishuo Shi
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Wei Liu
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yanming Tian
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Sheng Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China.
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, 050017, China.
| | - Fang Yuan
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China.
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, 050017, China.
| |
Collapse
|
7
|
Carson KE, Alvarez J, Mackley J, Travagli RA, Browning KN. Perinatal high-fat diet exposure alters oxytocin and corticotropin releasing factor inputs onto vagal neurocircuits controlling gastric motility. J Physiol 2023; 601:2853-2875. [PMID: 37154244 PMCID: PMC10524104 DOI: 10.1113/jp284726] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/03/2023] [Indexed: 05/10/2023] Open
Abstract
Perinatal high-fat diet (pHFD) exposure alters the development of vagal neurocircuits that control gastrointestinal (GI) motility and reduce stress resiliency in offspring. Descending oxytocin (OXT; prototypical anti-stress peptide) and corticotropin releasing factor (CRF; prototypical stress peptide) inputs from the paraventricular nucleus (PVN) of the hypothalamus to the dorsal motor nucleus of the vagus (DMV) modulate the GI stress response. How these descending inputs, and their associated changes to GI motility and stress responses, are altered following pHFD exposure are, however, unknown. The present study used retrograde neuronal tracing experiments, cerebrospinal fluid extraction, in vivo recordings of gastric tone, motility and gastric emptying rates, and in vitro electrophysiological recordings from brainstem slice preparations to investigate the hypothesis that pHFD alters descending PVN-DMV inputs and dysregulates vagal brain-gut responses to stress. Compared to controls, rats exposed to pHFD had slower gastric emptying rates and did not respond to acute stress with the expected delay in gastric emptying. Neuronal tracing experiments demonstrated that pHFD reduced the number of PVNOXT neurons that project to the DMV, but increased PVNCRF neurons. Both in vitro electrophysiology recordings of DMV neurons and in vivo recordings of gastric motility and tone demonstrated that, following pHFD, PVNCRF -DMV projections were tonically active, and that pharmacological antagonism of brainstem CRF1 receptors restored the appropriate gastric response to brainstem OXT application. These results suggest that pHFD exposure disrupts descending PVN-DMV inputs, leading to a dysregulated vagal brain-gut response to stress. KEY POINTS: Maternal high-fat diet exposure is associated with gastric dysregulation and stress sensitivity in offspring. The present study demonstrates that perinatal high-fat diet exposure downregulates hypothalamic-vagal oxytocin (OXT) inputs but upregulates hypothalamic-vagal corticotropin releasing factor (CRF) inputs. Both in vitro and in vivo studies demonstrated that, following perinatal high-fat diet, CRF receptors were tonically active at NTS-DMV synapses, and that pharmacological antagonism of these receptors restored the appropriate gastric response to OXT. The current study suggests that perinatal high-fat diet exposure disrupts descending PVN-DMV inputs, leading to a dysregulated vagal brain-gut response to stress.
Collapse
Affiliation(s)
- Kaitlin E. Carson
- Department of Neural and Behavioral Sciences, Pennsylvania State College of Medicine, Hershey, PA
| | - Jared Alvarez
- Barrett Honors College, Arizona State University, Tempe, AZ
| | - Jasmine Mackley
- Schreyer Honors College, Pennsylvania State University, State College, PA
| | | | - Kirsteen N. Browning
- Address for correspondence: Kirsteen N. Browning, PhD, Penn State College of Medicine, 500 University Drive, MC H109, Hershey, PA, 17033;
| |
Collapse
|
8
|
Zhu L, Ye Z, Zhang M, Xu W, Wang R, Wu S, Gao H. Electroacupuncture intervention on stress-induced cardiac autonomic imbalance in rats involves corticotropin-releasing hormone system activity. Neuroreport 2023; 34:401-410. [PMID: 37096785 DOI: 10.1097/wnr.0000000000001905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Autonomic imbalance is a core aspect of stress response that strongly correlates to cardiovascular diseases. Enhanced activity of the central corticotropin-releasing hormone (CRH) system may result in autonomic imbalance to cause cardiovascular responses in a stress state. Electroacupuncture at PC6 acupoints has been demonstrated to prevent and treat cardiovascular diseases. In this study, we aim to demonstrate the protective role of electroacupuncture at PC6 in ameliorating cardiac autonomic imbalance and investigate the underlying mechanisms in immobilization stress rats. Four groups were subjected. Immobilization stress was applied to three groups. And the rats in two electroacupuncture-intervened groups exerted electroacupuncture at PC6 or tail respectively. Then, we performed ECG recording for heart rate variability (HRV) analysis, and rats were sacrificed after experiments for biological analysis. HRV analysis indicated that electroacupuncture at PC6 improved the enhanced low-frequency band of the power spectrum (LF), the reduced high-frequency band of the power spectrum (HF), and the enhanced LF/HF ratio caused by immobilization stress. Besides, electroacupuncture at PC6 significantly decreased phosphorylated tyrosine hydroxylase expression and increased acetylcholine esterase expression in heart of immobilization stress rats. Furthermore, electroacupuncture at PC6 significantly decreased CRH level and CRH 1 type receptor and CRH 2 type receptor (CRHR2) expressions in the rostral ventrolateral medulla (RVLM), and CRH level and CRHR2 expression in the nucleus of the solitary tract (NTS) of immobilization stress rats. Our findings suggest that electroacupuncture at PC6 can ameliorate stress-induced cardiac autonomic imbalance by modulating the CRHergic input in the RVLM and NTS.
Collapse
Affiliation(s)
- Li Zhu
- School of Acupuncture and Tuina, Anhui University of Chinese Medicine
| | - Zhen Ye
- School of Acupuncture and Tuina, Anhui University of Chinese Medicine
| | - Mengting Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine
- CAS Key Laboratory of Brain Function and Disease, and School of Life Sciences, University of Science Technology of China
| | - Weichen Xu
- School of Acupuncture and Tuina, Anhui University of Chinese Medicine
| | - Ruwen Wang
- School of Acupuncture and Tuina, Anhui University of Chinese Medicine
| | - Shengbing Wu
- School of Acupuncture and Tuina, Anhui University of Chinese Medicine
- Research Institute of Acupuncture and Meridian, Anhui Academy of Chinese Medicine
- Key Laboratory of Acupuncture-moxibustion Basis and Technology of Anhui Higher Education Institutes, Anhui University of Chinese Medicine (KLABT)
- Key Laboratory of Xin'an Medicine (Anhui University of Chinese Medicine), the Ministry of Education, Hefei, Anhui, China
| | - Heren Gao
- School of Acupuncture and Tuina, Anhui University of Chinese Medicine
- CAS Key Laboratory of Brain Function and Disease, and School of Life Sciences, University of Science Technology of China
- Research Institute of Acupuncture and Meridian, Anhui Academy of Chinese Medicine
| |
Collapse
|
9
|
Xu Z, Hu SW, Zhou Y, Guo Q, Wang D, Gao YH, Zhao WN, Tang HM, Yang JX, Yu X, Ding HL, Cao JL. Corticotropin-releasing factor neurones in the paraventricular nucleus of the hypothalamus modulate isoflurane anaesthesia and its responses to acute stress in mice. Br J Anaesth 2023; 130:446-458. [PMID: 36737387 DOI: 10.1016/j.bja.2022.12.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 11/23/2022] [Accepted: 12/23/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Corticotropin-releasing factor (CRF) neurones in the paraventricular nucleus (PVN) of the hypothalamus (PVNCRF neurones) can promote wakefulness and are activated under anaesthesia. However, whether these neurones contribute to anaesthetic effects is unknown. METHODS With a combination of chemogenetic and molecular approaches, we examined the roles of PVNCRF neurones in isoflurane anaesthesia in mice and further explored the underlying cellular and molecular mechanisms. RESULTS PVN neurones exhibited increased Fos expression during isoflurane anaesthesia (mean [standard deviation], 218 [69.3] vs 21.3 [7.3]; P<0.001), and ∼75% were PVNCRF neurones. Chemogenetic inhibition of PVNCRF neurones facilitated emergence from isoflurane anaesthesia (11.7 [1.1] vs 13.9 [1.2] min; P=0.001), whereas chemogenetic activation of these neurones delayed emergence from isoflurane anaesthesia (16.9 [1.2] vs 13.9 [1.3] min; P=0.002). Isoflurane exposure increased CRF protein expression in PVN (4.0 [0.1] vs 2.2 [0.3], respectively; P<0.001). Knockdown of CRF in PVNCRF neurones mimicked the effects of chemogenetic inhibition of PVNCRF neurones in facilitating emergence (9.6 [1.1] vs 13.0 [1.4] min; P=0.003) and also abolished the effects of chemogenetic activation of PVNCRF neurones on delaying emergence from isoflurane anaesthesia (10.3 [1.3] vs 16.0 [2.6] min; P<0.001). Acute, but not chronic, stress delayed emergence from isoflurane anaesthesia (15.5 [1.5] vs 13.0 [1.4] min; P=0.004). This effect was reversed by chemogenetic inhibition of PVNCRF neurones (11.7 [1.6] vs 14.7 [1.4] min; P=0.001) or knockdown of CRF in PVNCRF neurones (12.3 [1.5] vs 15.3 [1.6] min; P=0.002). CONCLUSIONS CRF neurones in the PVN of the hypothalamus neurones modulate isoflurane anaesthesia and acute stress effects on anaesthesia through CRF signalling.
Collapse
Affiliation(s)
- Zheng Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Su-Wan Hu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Yu Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Qingchen Guo
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Di Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Yi-Hong Gao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Wei-Nan Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Hui-Mei Tang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Jun-Xia Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Xiaolu Yu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Hai-Lei Ding
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China; Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
10
|
Rasiah NP, Loewen SP, Bains JS. Windows into stress: a glimpse at emerging roles for CRH PVN neurons. Physiol Rev 2023; 103:1667-1691. [PMID: 36395349 DOI: 10.1152/physrev.00056.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The corticotropin-releasing hormone cells in the paraventricular nucleus of the hypothalamus (CRHPVN) control the slow endocrine response to stress. The synapses on these cells are exquisitely sensitive to acute stress, leveraging local signals to leave a lasting imprint on this system. Additionally, recent work indicates that these cells also play key roles in the control of distinct stress and survival behaviors. Here we review these observations and provide a perspective on the role of CRHPVN neurons as integrative and malleable hubs for behavioral, physiological, and endocrine responses to stress.
Collapse
Affiliation(s)
- Neilen P Rasiah
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Spencer P Loewen
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jaideep S Bains
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
11
|
Jiang S, Chen L, Huang ZL, Chen CR. Role of the paraventricular nucleus of the hypothalamus in sleep–wake regulation. BRAIN SCIENCE ADVANCES 2022. [DOI: 10.26599/bsa.2022.9050017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The paraventricular nucleus of the hypothalamus (PVH) acts as a cohesive functional unit that regulates neuroendocrine and autonomic function, complex behavior, and negative emotions after stress. However, how the PVH integrates arousal with these biological functions has only recently been explored. Clinical reports, combined with neurotoxic lesioning, immunochemistry, neuronal activity recordings, and the polysomnographic analyses of genetically modified animals, have revealed that the PVH is important for the control of wakefulness. Here, we review emerging anatomical and neural mechanisms for sleep–wake regulation in the PVH to support its essential role in the promotion and maintenance of wakefulness.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College Fudan University, Shanghai 200030, China
| | - Lu Chen
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College Fudan University, Shanghai 200030, China
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College Fudan University, Shanghai 200030, China
| | - Chang-Rui Chen
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College Fudan University, Shanghai 200030, China
| |
Collapse
|
12
|
Holt MK. The ins and outs of the caudal nucleus of the solitary tract: An overview of cellular populations and anatomical connections. J Neuroendocrinol 2022; 34:e13132. [PMID: 35509189 PMCID: PMC9286632 DOI: 10.1111/jne.13132] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/25/2022] [Accepted: 03/22/2022] [Indexed: 11/29/2022]
Abstract
The body and brain are in constant two-way communication. Driving this communication is a region in the lower brainstem: the dorsal vagal complex. Within the dorsal vagal complex, the caudal nucleus of the solitary tract (cNTS) is a major first stop for incoming information from the body to the brain carried by the vagus nerve. The anatomy of this region makes it ideally positioned to respond to signals of change in both emotional and bodily states. In turn, the cNTS controls the activity of regions throughout the brain that are involved in the control of both behaviour and physiology. This review is intended to help anyone with an interest in the cNTS. First, I provide an overview of the architecture of the cNTS and outline the wide range of neurotransmitters expressed in subsets of neurons in the cNTS. Next, in detail, I discuss the known inputs and outputs of the cNTS and briefly highlight what is known regarding the neurochemical makeup and function of those connections. Then, I discuss one group of cNTS neurons: glucagon-like peptide-1 (GLP-1)-expressing neurons. GLP-1 neurons serve as a good example of a group of cNTS neurons, which receive input from varied sources and have the ability to modulate both behaviour and physiology. Finally, I consider what we might learn about other cNTS neurons from our study of GLP-1 neurons and why it is important to remember that the manipulation of molecularly defined subsets of cNTS neurons is likely to affect physiology and behaviours beyond those monitored in individual experiments.
Collapse
Affiliation(s)
- Marie K. Holt
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| |
Collapse
|
13
|
Copper exposure for 30 days at a daily dose twice the recommended increases blood pressure and cardiac contractility. Life Sci 2022; 300:120579. [PMID: 35489564 DOI: 10.1016/j.lfs.2022.120579] [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: 03/02/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 11/21/2022]
Abstract
Copper is an essential factor for the body's homeostasis. However, excess copper compromises organic functions. AIMS We investigated the effects of copper exposure for 30 days on blood pressure (BP) and cardiac contractility and the putative involvement of nitric oxide (NO) and reactive oxygen species. MAIN METHODS Wistar rats (12 weeks old, 280 g) were randomized to the treated group that was exposed for 30 days to copper (2000 μg/kg/day CuCl2) and the control (Ct) group that received intraperitoneal saline (0.9%). KEY FINDINGS The blood concentration of copper was ~1.26 μg/mL in the copper-exposed group and ~0.024 μg/mL in the Ct group. The main metal accumulations occurred in the liver and kidneys. Copper exposure increased systolic BP (Cu: 141 ± 3 mmHg; Ct: 133 ± 3 mmHg) (tail cuff method), left ventricular systolic pressure and papillary muscle force. Calcium release from the sarcoplasmic reticulum was reduced. The contractile response to Ca2+ was increased by copper, and the effect was not altered by L-NAME. Copper increased contractions dependent on sarcolemmal Ca2+ influx, and this effect was not altered by L-NAME. The percentage response to isoproterenol decreased in the copper-exposed group, but L-NAME did not alter this reduction. Papillary force development at the peak and plateau of tetanic contractions also increased after copper exposure, but this effect was not altered by L-NAME. In situ detection of OH local production increased. SIGNIFICANCE Copper increased BP and cardiac force, increased Ca2+ inflow, reduced Ca2+ reuptake by the sarcoplasmic reticulum, and increased OH local production. Copper exposure at doses considered tolerable affects cardiac contractility.
Collapse
|
14
|
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: 21] [Impact Index Per Article: 10.5] [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
|
15
|
Forstenpointner J, Elman I, Freeman R, Borsook D. The Omnipresence of Autonomic Modulation in Health and Disease. Prog Neurobiol 2022; 210:102218. [PMID: 35033599 DOI: 10.1016/j.pneurobio.2022.102218] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/13/2021] [Accepted: 01/10/2022] [Indexed: 10/19/2022]
Abstract
The Autonomic Nervous System (ANS) is a critical part of the homeostatic machinery with both central and peripheral components. However, little is known about the integration of these components and their joint role in the maintenance of health and in allostatic derailments leading to somatic and/or neuropsychiatric (co)morbidity. Based on a comprehensive literature search on the ANS neuroanatomy we dissect the complex integration of the ANS: (1) First we summarize Stress and Homeostatic Equilibrium - elucidating the responsivity of the ANS to stressors; (2) Second we describe the overall process of how the ANS is involved in Adaptation and Maladaptation to Stress; (3) In the third section the ANS is hierarchically partitioned into the peripheral/spinal, brainstem, subcortical and cortical components of the nervous system. We utilize this anatomical basis to define a model of autonomic integration. (4) Finally, we deploy the model to describe human ANS involvement in (a) Hypofunctional and (b) Hyperfunctional states providing examples in the healthy state and in clinical conditions.
Collapse
Affiliation(s)
- Julia Forstenpointner
- Center for Pain and the Brain, Boston Children's Hospital, Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, USA; Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, SH, Germany.
| | - Igor Elman
- Center for Pain and the Brain, Boston Children's Hospital, Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, USA; Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA
| | - Roy Freeman
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - David Borsook
- Center for Pain and the Brain, Boston Children's Hospital, Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, USA; Departments of Psychiatry and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Holt MK. Mind affects matter: Hindbrain GLP1 neurons link stress, physiology and behaviour. Exp Physiol 2021; 106:1853-1862. [PMID: 34302307 DOI: 10.1113/ep089445] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022]
Abstract
NEW FINDINGS What is the topic of this review? This Lecture covers the role of caudal brainstem GLP1 neurons in acute and chronic stress responses. What advances does it highlight? This Lecture focuses on the recent advances in our understanding of GLP1 neurons and their physiological role in many aspects of stress. Particular focus is given to the recent elucidation, in part, of the anatomical basis for recruitment of GLP1 neurons in response to acute stress. Finally, the potential, but at this time somewhat speculative, role of GLP1 neurons in chronic stress is discussed. ABSTRACT The brain responds rapidly to stressful stimuli by increasing sympathetic outflow, activating the hypothalamic-pituitary-adrenal axis and eliciting avoidance behaviours to limit risks to safety. Stress responses are adaptive and essential but can become maladaptive when the stress is chronic, causing autonomic imbalance, hypothalamic-pituitary-adrenal axis hyper-reactivity and a state of hypervigilance. Ultimately, this contributes to the development of cardiovascular disease and affective disorders, including major depression and anxiety. Stress responses are often thought to be driven mainly by forebrain areas; however, the brainstem nucleus of the solitary tract (NTS) is ideally located to control both autonomic outflow and behaviour in response to stress. Here, I review the preclinical evidence that the NTS and its resident glucagon-like peptide-1 (GLP1)-expressing neurons are prominent mediators of stress responses. This Lecture introduces the reader to the idea of good and bad stress and outlines the types of stress that engage the NTS and GLP1 neurons. I describe in particular detail the recent studies by myself and others aimed at mapping sources of synaptic inputs to GLP1 neurons and consider the implications for our understanding of the role of GLP1 neurons in stress. This is followed by a discussion of the contribution of brain GLP1 and GLP1 neurons to behavioural and physiological stress responses. The evidence reviewed highlights a potentially prominent role for GLP1 neurons in the response of the brain to acute stress and reveals important unanswered questions regarding their role in chronic stress.
Collapse
Affiliation(s)
- Marie K Holt
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| |
Collapse
|
17
|
Elsaafien K, Kirchner MK, Mohammed M, Eikenberry SA, West C, Scott KA, de Kloet AD, Stern JE, Krause EG. Identification of Novel Cross-Talk between the Neuroendocrine and Autonomic Stress Axes Controlling Blood Pressure. J Neurosci 2021; 41:4641-4657. [PMID: 33858944 PMCID: PMC8260250 DOI: 10.1523/jneurosci.0251-21.2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 11/21/2022] Open
Abstract
The hypothalamic paraventricular nucleus (PVN) controls neuroendocrine axes and the autonomic nervous system to mount responses that cope with the energetic burdens of psychological or physiological stress. Neurons in the PVN that express the angiotensin Type 1a receptor (PVNAgtr1a) are implicated in neuroendocrine and autonomic stress responses; however, the mechanism by which these neurons coordinate activation of neuroendocrine axes with sympathetic outflow remains unknown. Here, we use a multidisciplinary approach to investigate intra-PVN signaling mechanisms that couple the activity of neurons synthesizing corticotropin-releasing-hormone (CRH) to blood pressure. We used the Cre-Lox system in male mice with in vivo optogenetics and cardiovascular recordings to demonstrate that excitation of PVNAgtr1a promotes elevated blood pressure that is dependent on the sympathetic nervous system. Next, neuroanatomical experiments found that PVNAgtr1a synthesize CRH, and intriguingly, fibers originating from PVNAgtr1a make appositions onto neighboring neurons that send projections to the rostral ventrolateral medulla and express CRH type 1 receptor (CRHR1) mRNA. We then used an ex vivo preparation that combined optogenetics, patch-clamp electrophysiology, and Ca2+ imaging to discover that excitation of PVNAgtr1a drives the local, intra-PVN release of CRH, which activates rostral ventrolateral medulla-projecting neurons via stimulation of CRHR1(s). Finally, we returned to our in vivo preparation and found that CRH receptor antagonism specifically within the PVN lowered blood pressure basally and during optogenetic activation of PVNAgtr1a Collectively, these results demonstrate that angiotensin II acts on PVNAgtr1a to conjoin hypothalamic-pituitary-adrenal axis activity with sympathetically mediated vasoconstriction in male mice.SIGNIFICANCE STATEMENT The survival of an organism is dependent on meeting the energetic demands imposed by stressors. This critical function is accomplished by the CNS's ability to orchestrate simultaneous activities of neurosecretory and autonomic axes. Here, we unveil a novel signaling mechanism within the paraventricular nucleus of the hypothalamus that links excitation of neurons producing corticotropin-releasing-hormone with excitation of neurons controlling sympathetic nervous system activity and blood pressure. The implication is that chronic stress exposure may promote cardiometabolic disease by dysregulating the interneuronal cross-talk revealed by our experiments.
Collapse
Affiliation(s)
- Khalid Elsaafien
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32611
- Center of Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida 32611
| | - Matthew K Kirchner
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, Georgia 30302
| | - Mazher Mohammed
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32611
| | - Sophia A Eikenberry
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida 32611
| | - Chloe West
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, Georgia 30302
| | - Karen A Scott
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32611
- Center of Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida 32611
| | - Annette D de Kloet
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida 32611
- Center of Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida 32611
- McKnight Brain Institute, University of Florida, Gainesville, Florida 32611
| | - Javier E Stern
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, Georgia 30302
| | - Eric G Krause
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32611
- Center of Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida 32611
- McKnight Brain Institute, University of Florida, Gainesville, Florida 32611
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Rapidly emerging evidence implicates an important role of gut-brain-bone marrow (BM) axis involving gut microbiota (GM), gut epithelial wall permeability, increased production of pro-inflammatory BM cells and neuroinflammation in hypertension (HTN). However, the precise sequence of events involving these organs remains to be established. Furthermore, whether an impaired gut-brain-BM axis is a cause or consequence of HTN is actively under investigation. This will be extremely important for translation of this fundamental knowledge to novel, innovative approaches for the control and management of HTN. Therefore, our objectives are to summarize the latest hypothesis, provide evidence for and against the impaired gut, BM and brain interactions in HTN and discuss perspectives and future directions. RECENT FINDINGS Hypertensive stimuli activate autonomic neural pathways resulting in increased sympathetic and decreased parasympathetic cardiovascular modulation. This directly affects the functions of cardiovascular-relevant organs to increase blood pressure. Increases in sympathetic drive to the gut and BM also trigger sequences of signaling events that ultimately contribute to altered GM, increased gut permeability, enhanced gut- and brain-targeted pro-inflammatory cells from the BM in perpetuation and establishment of HTN. SUMMARY In this review, we present the mechanisms involving the brain, gut, and BM, whose dysfunctional interactions may be critical in persistent neuroinflammation and key in the development and establishment of HTN.
Collapse
Affiliation(s)
- Jing Li
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, USA
| | | | | |
Collapse
|
19
|
Kalsbeek A, Buijs RM. Organization of the neuroendocrine and autonomic hypothalamic paraventricular nucleus. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:45-63. [PMID: 34225948 DOI: 10.1016/b978-0-12-820107-7.00004-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A major function of the nervous system is to maintain a relatively constant internal environment. The distinction between our external environment (i.e., the environment that we live in and that is subject to major changes, such as temperature, humidity, and food availability) and our internal environment (i.e., the environment formed by the fluids surrounding our bodily tissues and that has a very stable composition) was pointed out in 1878 by Claude Bernard (1814-1878). Later on, it was indicated by Walter Cannon (1871-1945) that the internal environment is not really constant, but rather shows limited variability. Cannon named the mechanism maintaining this limited variability homeostasis. Claude Bernard envisioned that, for optimal health, all physiologic processes in the body needed to maintain homeostasis and should be in perfect harmony with each other. This is illustrated by the fact that, for instance, during the sleep-wake cycle important elements of our physiology such as body temperature, circulating glucose, and cortisol levels show important variations but are in perfect synchrony with each other. These variations are driven by the biologic clock in interaction with hypothalamic target areas, among which is the paraventricular nucleus of the hypothalamus (PVN), a core brain structure that controls the neuroendocrine and autonomic nervous systems and thus is key for integrating central and peripheral information and implementing homeostasis. This chapter focuses on the anatomic connections between the biologic clock and the PVN to modulate homeostasis according to the daily sleep-wake rhythm. Experimental studies have revealed a highly specialized organization of the connections between the clock neurons and neuroendocrine system as well as preautonomic neurons in the PVN. These complex connections ensure a logical coordination between behavioral, endocrine, and metabolic functions that helps the organism maintain homeostasis throughout the day.
Collapse
Affiliation(s)
- Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (Amsterdam UMC), University of Amsterdam, Amsterdam, The Netherlands; Department of Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands.
| | - Ruud M Buijs
- Hypothalamic Integration Mechanisms Laboratory, Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| |
Collapse
|
20
|
Li SB, Borniger JC, Yamaguchi H, Hédou J, Gaudilliere B, de Lecea L. Hypothalamic circuitry underlying stress-induced insomnia and peripheral immunosuppression. SCIENCE ADVANCES 2020; 6:eabc2590. [PMID: 32917689 PMCID: PMC11206529 DOI: 10.1126/sciadv.abc2590] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/28/2020] [Indexed: 06/11/2023]
Abstract
The neural substrates of insomnia/hyperarousal induced by stress remain unknown. Here, we show that restraint stress leads to hyperarousal associated with strong activation of corticotropin-releasing hormone neurons in the paraventricular nucleus of hypothalamus (CRHPVN) and hypocretin neurons in the lateral hypothalamus (HcrtLH). CRHPVN neurons directly innervate HcrtLH neurons, and optogenetic stimulation of LH-projecting CRHPVN neurons elicits hyperarousal. CRISPR-Cas9-mediated knockdown of the crh gene in CRHPVN neurons abolishes hyperarousal induced by stimulating LH-projecting CRHPVN neurons. Genetic ablation of Hcrt neurons or crh gene knockdown significantly counteracts restraint stress-induced hyperarousal. Single-cell mass cytometry by time of flight (CyTOF) revealed extensive changes to immune cell distribution and functional responses in peripheral blood during hyperarousal upon optogenetic stimulation of CRHPVN neurons simulating stress-induced insomnia. Our findings suggest both central and peripheral systems are synergistically engaged in the response to stress via CRHPVN circuitry.
Collapse
Affiliation(s)
- Shi-Bin Li
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA 94305, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Jeremy C Borniger
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA 94305, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Hiroshi Yamaguchi
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA 94305, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Julien Hédou
- Department of Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brice Gaudilliere
- Department of Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA 94305, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
21
|
Gut microbiota modulates stress-induced hypertension through the HPA axis. Brain Res Bull 2020; 162:49-58. [PMID: 32535221 DOI: 10.1016/j.brainresbull.2020.05.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/07/2020] [Accepted: 05/29/2020] [Indexed: 12/20/2022]
Abstract
Stress is associated with an increased risk of hypertension, and the incidence of stress-related hypertension has risen rapidly in recent years; however, the underlying mechanisms remain elusive. Gut dysbiosis has been demonstrated to contribute to hypertension and hyperactivation of the hypothalamus-pituitary-adrenal (HPA) axis. Based on our previous findings showing the altered gut microbiota in the rats of stress-induced hypertension (SIH), the present study aims to investigate whether the stress-induced alteration in gut microbiota can lead to the dysfunction of the HPA axis which contributes to the development of SIH. SIH was developed in rats subjected to electric foot-shock combined with buzzer noise stressors. The gut microbiota of rats were deleted by administering an antibiotic cocktail containing ampicillin (1 g/L), vancomycin (500 mg/L), neomycin (1 g/L), and metronidazole (1 g/L) in drinking water. The serum levels of adrenocorticotropic hormone (ACTH) and corticosterone (CORT) were tested using enzyme-linked immunosorbent assay (ELISA). The mRNA expression of glucocorticoid receptor (GR) and corticotropin-releasing factor (CRF), CRFR1 and CRFR2 was detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR). The cellular protein expressions of corticotropin-releasing hormone (CRH), c-fos, and GR were examined by immunohistochemical staining. In the present study, SIH rats showed a hyperactive HPA axis as indicated by the increased CRH expression in the paraventricular nucleus (PVN) of the hypothalamus, the elevated serum ACTH or CORT concentrations, and increased adrenal gland index. The decreased GR expression and increased CRFR1 in the hypothalamus might underlie the hyperactivation of the HPA axis. The microbial deletion by antibiotics mitigated the hyperactivation of the HPA axis and attenuated the stress-induced elevation of blood pressure, indicating that the causal link of gut microbiota to SIH is mediated, at least in part, by the HPA axis activity. Our findings shed new light on the mechanisms underlying SIH.
Collapse
|
22
|
Matovic S, Ichiyama A, Igarashi H, Salter EW, Sunstrum JK, Wang XF, Henry M, Kuebler ES, Vernoux N, Martinez-Trujillo J, Tremblay ME, Inoue W. Neuronal hypertrophy dampens neuronal intrinsic excitability and stress responsiveness during chronic stress. J Physiol 2020; 598:2757-2773. [PMID: 32347541 DOI: 10.1113/jp279666] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/17/2020] [Indexed: 01/16/2023] Open
Abstract
KEY POINTS The hypothalamic-pituitary-adrenal (HPA) axis habituates to repeated stress exposure. We studied hypothalamic corticotropin-releasing hormone (CRH) neurons that form the apex of the HPA axis in a mouse model of stress habituation using repeated restraint. The intrinsic excitability of CRH neurons decreased after repeated stress in a time course that coincided with the development of HPA axis habituation. This intrinsic excitability plasticity co-developed with an expansion of surface membrane area, which increased a passive electric load and dampened membrane depolarization in response to the influx of positive charge. We report a novel structure-function relationship for intrinsic excitability plasticity as a neural correlate for HPA axis habituation. ABSTRACT Encountering a stressor immediately activates the hypothalamic-pituitary-adrenal (HPA) axis, but this stereotypic stress response also undergoes experience-dependent adaptation. Despite the biological and clinical importance, how the brain adjusts stress responsiveness in the long term remains poorly understood. We studied hypothalamic corticotropin-releasing hormone neurons that form the apex of the HPA axis in a mouse model of stress habituation using repeated restraint. Using patch-clamp electrophysiology in acute slices, we found that the intrinsic excitability of these neurons substantially decreased after daily repeated stress in a time course that coincided with their loss of stress responsiveness in vivo. This intrinsic excitability plasticity co-developed with an expansion of surface membrane area, which increased a passive electric load, and dampened membrane depolarization in response to the influx of positive charge. Multiphoton imaging and electron microscopy revealed that repeated stress augmented ruffling of the plasma membrane, suggesting an ultrastructural plasticity that may efficiently accommodate the membrane area expansion. Overall, we report a novel structure-function relationship for intrinsic excitability plasticity as a neural correlate for adaptation of the neuroendocrine stress response.
Collapse
Affiliation(s)
- Sara Matovic
- Robarts Research Institute, University of Western Ontario.,Neuroscience Program, University of Western Ontario
| | - Aoi Ichiyama
- Neuroscience Program, University of Western Ontario
| | | | - Eric W Salter
- Robarts Research Institute, University of Western Ontario.,Current address: University of Toronto
| | | | - Xue Fan Wang
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario
| | - Mathilde Henry
- Axe Neurosciences, CRCHU de Quebec-Université Laval.,Current address: INRAE, Univ. Bordeaux, Bordeaux INP, Nutrineuro, UMR 1286, Bordeaux, F-33000, France
| | - Eric S Kuebler
- Robarts Research Institute, University of Western Ontario
| | | | - Julio Martinez-Trujillo
- Robarts Research Institute, University of Western Ontario.,Neuroscience Program, University of Western Ontario.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario
| | - Marie-Eve Tremblay
- Axe Neurosciences, CRCHU de Quebec-Université Laval.,Département de médecine moléculaire, Université Laval.,Division of Medical Sciences, University of Victoria
| | - Wataru Inoue
- Robarts Research Institute, University of Western Ontario.,Neuroscience Program, University of Western Ontario.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario
| |
Collapse
|
23
|
Ruyle BC, Martinez D, Heesch CM, Kline DD, Hasser EM. The PVN enhances cardiorespiratory responses to acute hypoxia via input to the nTS. Am J Physiol Regul Integr Comp Physiol 2019; 317:R818-R833. [PMID: 31509428 PMCID: PMC6962628 DOI: 10.1152/ajpregu.00135.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/26/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
Abstract
Chemoreflex neurocircuitry includes the paraventricular nucleus (PVN), but the role of PVN efferent projections to specific cardiorespiratory nuclei is unclear. We hypothesized that the PVN contributes to cardiorespiratory responses to hypoxia via projections to the nucleus tractus solitarii (nTS). Rats received bilateral PVN microinjections of adeno-associated virus expressing inhibitory designer receptor exclusively activated by designer drug (GiDREADD) or green fluorescent protein (GFP) control. Efficacy of GiDREADD inhibition by the designer receptor exclusively activated by designer drug (DREADD) agonist Compound 21 (C21) was verified in PVN slices; C21 reduced evoked action potential discharge by reducing excitability to injected current in GiDREADD-expressing PVN neurons. We evaluated hypoxic ventilatory responses (plethysmography) and PVN and nTS neuronal activation (cFos immunoreactivity) to 2 h hypoxia (10% O2) in conscious GFP and GiDREADD rats after intraperitoneal C21 injection. Generalized PVN inhibition via systemic C21 blunted hypoxic ventilatory responses and reduced PVN and also nTS neuronal activation during hypoxia. To determine if the PVN-nTS pathway contributes to these effects, we evaluated cardiorespiratory responses to hypoxia during selective PVN terminal inhibition in the nTS. Anesthetized GFP and GiDREADD rats exposed to brief hypoxia (10% O2, 45 s) exhibited depressor and tachycardic responses and increased sympathetic and phrenic nerve activity. C21 was then microinjected into the nTS, followed after 60 min by another hypoxic episode. In GiDREADD but not GFP rats, PVN terminal inhibition by nTS C21 strongly attenuated the phrenic amplitude response to hypoxia. Interestingly, C21 augmented tachycardic and sympathetic responses without altering the coupling of splanchnic sympathetic nerve activity to phrenic nerve activity during hypoxia. Data demonstrate that the PVN, including projections to the nTS, is critical in shaping sympathetic and respiratory responses to hypoxia.
Collapse
Affiliation(s)
- Brian C Ruyle
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Diana Martinez
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, 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
| | - David D Kline
- 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
|
24
|
Synaptic Inputs to the Mouse Dorsal Vagal Complex and Its Resident Preproglucagon Neurons. J Neurosci 2019; 39:9767-9781. [PMID: 31666353 PMCID: PMC6891065 DOI: 10.1523/jneurosci.2145-19.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/07/2019] [Accepted: 10/13/2019] [Indexed: 12/17/2022] Open
Abstract
Stress responses are coordinated by widespread neural circuits. Homeostatic and psychogenic stressors activate preproglucagon (PPG) neurons in the caudal nucleus of the solitary tract (cNTS) that produce glucagon-like peptide-1; published work in rodents indicates that these neurons play a crucial role in stress responses. While the axonal targets of PPG neurons are well established, their afferent inputs are unknown. Stress responses are coordinated by widespread neural circuits. Homeostatic and psychogenic stressors activate preproglucagon (PPG) neurons in the caudal nucleus of the solitary tract (cNTS) that produce glucagon-like peptide-1; published work in rodents indicates that these neurons play a crucial role in stress responses. While the axonal targets of PPG neurons are well established, their afferent inputs are unknown. Here we use retrograde tracing with cholera toxin subunit b to show that the cNTS in male and female mice receives axonal inputs similar to those reported in rats. Monosynaptic and polysynaptic inputs specific to cNTS PPG neurons were revealed using Cre-conditional pseudorabies and rabies viruses. The most prominent sources of PPG monosynaptic input include the lateral (LH) and paraventricular (PVN) nuclei of the hypothalamus, parasubthalamic nucleus, lateral division of the central amygdala, and Barrington's nucleus (Bar). Additionally, PPG neurons receive monosynaptic vagal sensory input from the nodose ganglia and spinal sensory input from the dorsal horn. Sources of polysynaptic input to cNTS PPG neurons include the hippocampal formation, paraventricular thalamus, and prefrontal cortex. Finally, cNTS-projecting neurons within PVN, LH, and Bar express the activation marker cFOS in mice after restraint stress, identifying them as potential sources of neurogenic stress-induced recruitment of PPG neurons. In summary, cNTS PPG neurons in mice receive widespread monosynaptic and polysynaptic input from brain regions implicated in coordinating behavioral and physiological stress responses, as well as from vagal and spinal sensory neurons. Thus, PPG neurons are optimally positioned to integrate signals of homeostatic and psychogenic stress. SIGNIFICANCE STATEMENT Recent research has indicated a crucial role for glucagon-like peptide-1-producing preproglucagon (PPG) neurons in regulating both appetite and behavioral and autonomic responses to acute stress. Intriguingly, the central glucagon-like peptide-1 system defined in rodents is conserved in humans, highlighting the translational importance of understanding its anatomical organization. Findings reported here indicate that PPG neurons receive significant monosynaptic and polysynaptic input from brain regions implicated in autonomic and behavioral responses to stress, as well as direct input from vagal and spinal sensory neurons. Improved understanding of the neural pathways underlying the recruitment of PPG neurons may facilitate the development of novel therapies for the treatment of stress-related disorders.
Collapse
|
25
|
Jiang Z, Rajamanickam S, Justice NJ. CRF signaling between neurons in the paraventricular nucleus of the hypothalamus (PVN) coordinates stress responses. Neurobiol Stress 2019; 11:100192. [PMID: 31516918 PMCID: PMC6732729 DOI: 10.1016/j.ynstr.2019.100192] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 12/22/2022] Open
Abstract
The importance of a precisely coordinated neuroendocrine, autonomic, and behavioral stress response was a primary theme at the Stress Neurobiology Workshop 2018, held in the beautiful setting of Banff Provincial Park in Alberta, Canada. Much of the research featured at this meeting reinforced the importance of appropriately responding to stress in order to avoid various neuropsychiatric pathologies, including Post-Traumatic Stress Disorder (PTSD), depression, and addiction. Corticotropin-Releasing Factor (CRF) neurons in the paraventricular nucleus of the hypothalamus (PVN) are central players in the stress response, integrating both external and visceral stress-relevant information, then directing neuroendocrine, autonomic and behavioral adaptations via endocrine and neural outputs of the PVN. The PVN contains a densely packed array of neuron types that respond to stress, including CRF neurons that activate the Hypothalamic-Pituitary-Adrenal (HPA) axis. Recently, identification of a new population of neurons in the PVN that express CRF Receptor 1 (CRFR1) has suggested that CRF release in the PVN signals to neighboring CRF responsive neurons, potentially functioning in HPA axis feedback, neuroendocrine coordination, and autonomic signaling. Here, we review our recent work characterizing an intra-PVN microcircuit in which locally released CRF release activates CRFR1+ neurons that make recurrent inhibitory GABAergic synapses onto CRF neurons to dampen excitability , therebylimiting HPA axis hyperactivity in response to stress and promoting stress recovery, which we presented in a poster session at the conference. We then discuss questions that have arisen following publication of our initial characterization of the microcircuit, regarding specific features of intra-PVN CRF signaling and its potential role in coordinating neuroendocrine, autonomic, and behavioral outputs of the PVN. Our presented work, as well as many of the presentations at the Stress Neurobiology Workshop 2018 together establish intra-PVN signaling as an important regulatory node in stress response pathways, which are central to the pathogenesis of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Zhiying Jiang
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Texas, 77030, USA
| | - Shivakumar Rajamanickam
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Texas, 77030, USA
| | - Nicholas J Justice
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Texas, 77030, USA
| |
Collapse
|
26
|
Gao HR, Gao HY. Cardiovascular functions of central corticotropin-releasing factor related peptides system. Neuropeptides 2019; 75:18-24. [PMID: 30922523 DOI: 10.1016/j.npep.2019.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 02/26/2019] [Accepted: 03/19/2019] [Indexed: 10/27/2022]
Abstract
The corticotropin-releasing factor (CRF) related peptides system has widespread distributions in central nervous system, to perform many physiological and pathophysiological functions, including cardiovascular functions. A complex connection exists between the central CRF related peptides system and cardiovascular system. There are multiple pathways and mechanisms through which the central CRF related peptides system influences cardiovascular functions. A dysfunction in the central CRF related peptides system may lead to a wide range of alterations in cardiovascular functions. Though there are difficulties or limitations in establishing exact modulatory roles of the central CRF related peptides system in cardiovascular functions. The central CRF related peptides system as target to prevent cardiovascular diseases is being pursued with increasing interest. In this review, we summarize recent understanding on cardiovascular functions of the CRF related peptides system in limbic forebrain, hypothalamus and brain stem structures, discuss mechanisms of the central CRF related peptides system in control of cardiovascular functions, and suggest that the central CRF related peptides system may be a potent candidate for prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- He-Ren Gao
- Research Institute of Acupuncture and Meridian, Anhui University of Chinese Medicine, Hefei 230038, China.
| | - He-Yuan Gao
- Department of Pediatrics, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei 066000, China
| |
Collapse
|