1
|
Martinez P, Sabatier JM. Malignant tumors in vagal-innervated organs: Exploring its homeostatic role. Cancer Lett 2025; 617:217539. [PMID: 39954934 DOI: 10.1016/j.canlet.2025.217539] [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: 12/01/2024] [Revised: 01/28/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
Cancer remains a significant global health challenge, with its progression shaped by complex and multifactorial mechanisms. Recent research suggests that the vagus nerve could play a critical role in mediating communication between the tumor microenvironment and the central nervous system (CNS). This review highlights the diversity of vagal afferent receptors, which could position the vagus nerve as a unique pathway for transmitting immune, metabolic, mechanical, and chemical signals from tumors to the CNS. Such signaling could influence systemic disease progression and tumor-related responses. Additionally, the vagus nerve's interactions with the microbiome and the renin-angiotensin system (RAS)-both implicated in cancer biology-further underscore its potential central role in modulating tumor-related processes. Contradictions in the literature, particularly concerning vagal fibers, illustrate the complexity of its involvement in tumor progression, with both tumor-promoting and tumor-suppressive effects reported depending on cancer type and context. These contradictions often overlook certain experimental biases, such as the failure to distinguish between vagal afferent and efferent fibers during vagotomies or the localized parasympathetic effects that cannot always be extrapolated to the systemic level. By focusing on the homeostatic role of the vagus nerve, understanding these mechanisms could open the door to new perspectives in cancer research related to the vagus nerve and lead to potential therapeutic innovations.
Collapse
Affiliation(s)
| | - Jean-Marc Sabatier
- Institut de NeuroPhysiopathologie (INP), CNRS UMR 7051, 27 Bd Jean Moulin, 13005, Marseille, France
| |
Collapse
|
2
|
Ruyle BC, Masud S, Kesaraju R, Tahirkheli M, Modh J, Roth CG, Angulo-Lopera S, Lintz T, Higginbotham JA, Massaly N, Morón JA. Peripheral opioid receptor antagonism alleviates fentanyl-induced cardiorespiratory depression and is devoid of aversive behavior. eLife 2025; 13:RP104469. [PMID: 40167443 PMCID: PMC11961120 DOI: 10.7554/elife.104469] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Millions of Americans suffering from Opioid Use Disorders face a high risk of fatal overdose due to opioid-induced respiratory depression (OIRD). Fentanyl, a powerful synthetic opioid, is a major contributor to the rising rates of overdose deaths. Reversing fentanyl overdoses has proved challenging due to its high potency and the rapid onset of OIRD. We assessed the contributions of central and peripheral mu opioid receptors (MORs) in mediating fentanyl-induced physiological responses. The peripherally restricted MOR antagonist naloxone methiodide (NLXM) both prevented and reversed OIRD to a degree comparable to that of naloxone (NLX), indicating substantial involvement of peripheral MORs to OIRD. Interestingly, NLXM-mediated OIRD reversal did not produce aversive behaviors observed after NLX. We show that neurons in the nucleus of the solitary tract (nTS), the first central synapse of peripheral afferents, exhibit a biphasic activity profile following fentanyl exposure. NLXM pretreatment attenuates this activity, suggesting that these responses are mediated by peripheral MORs. Together, these findings establish a critical role for peripheral MORs, including ascending inputs to the nTS, as sites of dysfunction during OIRD. Furthermore, selective peripheral MOR antagonism could be a promising therapeutic strategy for managing OIRD by sparing CNS-driven acute opioid-associated withdrawal and aversion observed after NLX.
Collapse
Affiliation(s)
- Brian C Ruyle
- Department of Anesthesiology, Washington University in St. LouisSt. LouisUnited States
- Pain Center, Washington University in St. LouisSt. LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Sarah Masud
- Department of Anesthesiology, Washington University in St. LouisSt. LouisUnited States
- Pain Center, Washington University in St. LouisSt. LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Rohith Kesaraju
- Department of Anesthesiology, Washington University in St. LouisSt. LouisUnited States
- Pain Center, Washington University in St. LouisSt. LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Mubariz Tahirkheli
- Department of Anesthesiology, Washington University in St. LouisSt. LouisUnited States
- Pain Center, Washington University in St. LouisSt. LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Juhi Modh
- Department of Anesthesiology, Washington University in St. LouisSt. LouisUnited States
- Pain Center, Washington University in St. LouisSt. LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Caroline G Roth
- Department of Anesthesiology, Washington University in St. LouisSt. LouisUnited States
- Pain Center, Washington University in St. LouisSt. LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Sofia Angulo-Lopera
- Department of Anesthesiology, Washington University in St. LouisSt. LouisUnited States
- Pain Center, Washington University in St. LouisSt. LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Tania Lintz
- Department of Anesthesiology, Washington University in St. LouisSt. LouisUnited States
- Pain Center, Washington University in St. LouisSt. LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Jessica A Higginbotham
- Department of Anesthesiology, Washington University in St. LouisSt. LouisUnited States
- Pain Center, Washington University in St. LouisSt. LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Nicolas Massaly
- Department of Anesthesiology, Washington University in St. LouisSt. LouisUnited States
- Pain Center, Washington University in St. LouisSt. LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Jose A Morón
- Department of Anesthesiology, Washington University in St. LouisSt. LouisUnited States
- Pain Center, Washington University in St. LouisSt. LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
- Department of Neuroscience, Washington University in St. LouisSt. LouisUnited States
- Department of Psychiatry, Washington University in St. LouisSt. LouisUnited States
| |
Collapse
|
3
|
Ruyle BC, Masud S, Kesaraju R, Tahirkheli M, Modh J, Roth CG, Angulo-Lopera S, Lintz T, Higginbotham JA, Massaly N, Moron JA. Peripheral opioid receptor antagonism alleviates fentanyl-induced cardiorespiratory depression and is devoid of aversive behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.16.613257. [PMID: 39345613 PMCID: PMC11429738 DOI: 10.1101/2024.09.16.613257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Millions of Americans suffering from Opioid Use Disorders (OUD) face a high risk of fatal overdose due to opioid-induced respiratory depression (OIRD). Fentanyl, a powerful synthetic opioid, is a major contributor to the rising rates of overdose deaths. Reversing fentanyl overdoses has proved challenging due to its high potency and the rapid onset of OIRD. We assessed the contributions of central and peripheral mu opioid receptors (MORs) in mediating fentanyl-induced physiological responses. The peripherally restricted MOR antagonist naloxone methiodide (NLXM) both prevented and reversed OIRD to a degree comparable to that of naloxone (NLX), indicating substantial involvement of peripheral MORs to OIRD. Interestingly, NLXM-mediated OIRD reversal did not produce aversive behaviors observed after NLX. We show that neurons in the nucleus of the solitary tract (nTS), the first central synapse of peripheral afferents, exhibit a biphasic activity profile following fentanyl exposure. NLXM pretreatment attenuates this activity, suggesting that these responses are mediated by peripheral MORs. Together, these findings establish a critical role for peripheral MORs, including ascending inputs to the nTS, as sites of dysfunction during OIRD. Furthermore, selective peripheral MOR antagonism could be a promising therapeutic strategy for managing OIRD by sparing CNS-driven acute opioid-associated withdrawal and aversion observed after NLX. Significance Statement In this study, we compare the central versus peripheral components underlying fentanyl-induced cardiorespiratory depression to prevent overdose deaths. Our data indicate that these effects are, at least partially, due to the activation of mu opioid receptors present in peripheral sites. These findings provide insight into peripheral contributions to fentanyl-induced overdoses and could potentially lead to the development of treatments selectively targeting the peripheral system, sparing individuals from the CNS-driven acute opioid withdrawal generally observed with the use of naloxone.
Collapse
|
4
|
Dempsey JA, Gibbons TD. Rethinking O 2, CO 2 and breathing during wakefulness and sleep. J Physiol 2024; 602:5571-5585. [PMID: 37750243 DOI: 10.1113/jp284551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/11/2023] [Indexed: 09/27/2023] Open
Abstract
We have examined the importance of three long-standing questions concerning chemoreceptor influences on cardiorespiratory function which are currently experiencing a resurgence of study among physiologists and clinical investigators. Firstly, while carotid chemoreceptors (CB) are required for hypoxic stimulation of breathing, use of an isolated, extracorporeally perfused CB preparation in unanaesthetized animals with maintained tonic input from the CB, reveals that extra-CB hypoxaemia also provides dose-dependent ventilatory stimulation sufficient to account for 40-50% of the total ventilatory response to steady-state hypoxaemia. Extra-CB hyperoxia also provides a dose- and time-dependent hyperventilation. Extra-CB sites of O2-driven ventilatory stimulation identified to date include the medulla, kidney and spinal cord. Secondly, using the isolated or denervated CB preparation in awake animals and humans has demonstrated a hyperadditive effect of CB sensory input on central CO2 sensitivity, so that tonic CB activity accounts for as much as 35-40% of the normal, air-breathing eupnoeic drive to breathe. Thirdly, we argue for a key role for CO2 chemoreception and the neural drive to breathe in the pathogenesis of upper airway obstruction during sleep (OSA), based on the following evidence: (1) removal of the wakefulness drive to breathe enhances the effects of transient CO2 changes on breathing instability; (2) oscillations in respiratory motor output precipitate pharyngeal obstruction in sleeping subjects with compliant, collapsible airways; and (3) in the majority of patients in a large OSA cohort, a reduced neural drive to breathe accompanied reductions in both airflow and pharyngeal airway muscle dilator activity, precipitating airway obstruction.
Collapse
Affiliation(s)
| | - Travis D Gibbons
- University of British Columbia-Okanagan, Kelowna, British Columbia, Canada
| |
Collapse
|
5
|
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
|
6
|
Mohammadi M, Oghabian MA, Ghaderi S, Jalali M, Samadi S. Volumetric analysis of the hypothalamic subunits in obstructive sleep apnea. Brain Behav 2024; 14:e70026. [PMID: 39236146 PMCID: PMC11376441 DOI: 10.1002/brb3.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/22/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is a prevalent sleep disorder that is associated with structural brain damage and cognitive impairment. The hypothalamus plays a crucial role in regulating sleep and wakefulness. We aimed to evaluate hypothalamic subunit volumes in patients with OSA. METHODS We enrolled 30 participants (15 patients with OSA and 15 healthy controls (HC)). Patients with OSA underwent complete overnight polysomnography (PSG) examination. All the participants underwent MRI. The hypothalamic subunit volumes were calculated using a segmentation technique that trained a 3D convolutional neural network. RESULTS Although hypothalamus subunit volumes were comparable between the HC and OSA groups (lowest p = .395), significant negative correlations were found in OSA patients between BMI and whole left hypothalamus volume (R = -0.654, p = .008), as well as between BMI and left posterior volume (R = -0.556, p = .032). Furthermore, significant positive correlations were found between ESS and right anterior inferior volume (R = 0.548, p = .042), minimum SpO2 and the whole left hypothalamus (R = 0.551, p = .033), left tubular inferior volumes (R = 0.596, p = .019), and between the percentage of REM stage and left anterior inferior volume (R = 0.584, p = .022). CONCLUSIONS While there were no notable differences in the hypothalamic subunit volumes between the OSA and HC groups, several important correlations were identified in the OSA group. These relationships suggest that factors related to sleep apnea severity could affect hypothalamic structure in patients.
Collapse
Affiliation(s)
- Mahdi Mohammadi
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Neuroimaging and Analysis Group, Research Center for Molecular and Cellular Imaging, Advanced Medical Technologies and Equipment Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Oghabian
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Neuroimaging and Analysis Group, Research Center for Molecular and Cellular Imaging, Advanced Medical Technologies and Equipment Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadegh Ghaderi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Jalali
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Neuroimaging and Analysis Group, Research Center for Molecular and Cellular Imaging, Advanced Medical Technologies and Equipment Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahram Samadi
- Sleep Breathing Disorders Research Center, Imam Khomeini Hospital Complex, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Anesthesia, Critical Care and Pain Management Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Hao WY, Wang JX, Xu XY, Chen JL, Chen Q, Li YH, Zhu GQ, Chen AD. Chemerin in caudal division of nucleus tractus solitarius increases sympathetic activity and blood pressure. Eur J Neurosci 2024; 60:4830-4842. [PMID: 39044301 DOI: 10.1111/ejn.16475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/25/2024]
Abstract
Chemerin is an adipokine that contributes to metabolism regulation. Nucleus tractus solitarius (NTS) is the first relay station in the brain for accepting various visceral afferent activities for regulating cardiovascular activity. However, the roles of chemerin in the NTS in regulating sympathetic activity and blood pressure are almost unknown. This study aimed to determine the role and potential mechanism of chemerin in the NTS in modulating sympathetic outflow and blood pressure. Bilateral NTS microinjections were performed in anaesthetized adult male Sprague-Dawley rats. Renal sympathetic nerve activity (RSNA), mean arterial pressure (MAP) and heart rate (HR) were continuously recorded. Chemerin and its receptor chemokine-like receptor 1 (CMKLR1) were highly expressed in caudal NTS (cNTS). Microinjection of chemerin-9 to the cNTS increased RSNA, MAP and HR, which were prevented by CMKLR1 antagonist α-NETA, superoxide scavenger tempol or N-acetyl cysteine, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors diphenyleneiodonium or apocynin. Chemerin-9 increased superoxide production and NADPH oxidase activity in the cNTS. The increased superoxide production induced by chemerin-9 was inhibited by α-NETA. The effects of cNTS microinjection of chemerin-9 on the RSNA, MAP and HR were attenuated by the pretreatment with paraventricular nucleus (PVN) microinjection of NMDA receptor antagonist MK-801 rather than AMPA/kainate receptor antagonist CNQX. These results indicate that chemerin-9 in the NTS increases sympathetic outflow, blood pressure and HR via CMKLR1-mediated NADPH oxidase activation and subsequent superoxide production in anaesthetized normotensive rats. Glutamatergic inputs in the PVN are needed for the chemerin-9-induced responses.
Collapse
Affiliation(s)
- Wen-Yuan Hao
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing-Xiao Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao-Yu Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun-Liu Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qi Chen
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yue-Hua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guo-Qing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ai-Dong Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
8
|
Ben Musa R, Cornelius-Green J, Zhang H, Li DP, Kline DD, Hasser EM, Cummings KJ. Orexin Facilitates the Peripheral Chemoreflex via Corticotropin-Releasing Hormone Neurons Projecting to the Nucleus of the Solitary Tract. J Neurosci 2024; 44:e2383232024. [PMID: 38789262 PMCID: PMC11223477 DOI: 10.1523/jneurosci.2383-23.2024] [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: 01/02/2024] [Revised: 04/14/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
We previously showed that orexin neurons are activated by hypoxia and facilitate the peripheral chemoreflex (PCR)-mediated hypoxic ventilatory response (HVR), mostly by promoting the respiratory frequency response. Orexin neurons project to the nucleus of the solitary tract (nTS) and the paraventricular nucleus of the hypothalamus (PVN). The PVN contributes significantly to the PCR and contains nTS-projecting corticotropin-releasing hormone (CRH) neurons. We hypothesized that in male rats, orexin neurons contribute to the PCR by activating nTS-projecting CRH neurons. We used neuronal tract tracing and immunohistochemistry (IHC) to quantify the degree that hypoxia activates PVN-projecting orexin neurons. We coupled this with orexin receptor (OxR) blockade with suvorexant (Suvo, 20 mg/kg, i.p.) to assess the degree that orexin facilitates the hypoxia-induced activation of CRH neurons in the PVN, including those projecting to the nTS. In separate groups of rats, we measured the PCR following systemic orexin 1 receptor (Ox1R) blockade (SB-334867; 1 mg/kg) and specific Ox1R knockdown in PVN. OxR blockade with Suvo reduced the number of nTS and PVN neurons activated by hypoxia, including those CRH neurons projecting to nTS. Hypoxia increased the number of activated PVN-projecting orexin neurons but had no effect on the number of activated nTS-projecting orexin neurons. Global Ox1R blockade and partial Ox1R knockdown in the PVN significantly reduced the PCR. Ox1R knockdown also reduced the number of activated PVN neurons and the number of activated tyrosine hydroxylase neurons in the nTS. Our findings suggest orexin facilitates the PCR via nTS-projecting CRH neurons expressing Ox1R.
Collapse
Affiliation(s)
- Ruwaida Ben Musa
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| | - Jennifer Cornelius-Green
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| | - Hua Zhang
- Department of Medicine, Center for Precision Medicine, School of Medicine, University of Missouri, Columbia, Missouri 65212
| | - De-Pei Li
- Department of Medicine, Center for Precision Medicine, School of Medicine, University of Missouri, Columbia, Missouri 65212
| | - David D Kline
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| | - Eileen M Hasser
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| | - Kevin J Cummings
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| |
Collapse
|
9
|
Shafer BM, West CR, Foster GE. Advancements in the neurocirculatory reflex response to hypoxia. Am J Physiol Regul Integr Comp Physiol 2024; 327:R1-R13. [PMID: 38738293 PMCID: PMC11380992 DOI: 10.1152/ajpregu.00237.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 04/16/2024] [Accepted: 04/29/2024] [Indexed: 05/14/2024]
Abstract
Hypoxia is a pivotal factor in the pathophysiology of various clinical conditions, including obstructive sleep apnea, which has a strong association with cardiovascular diseases like hypertension, posing significant health risks. Although the precise mechanisms linking hypoxemia-associated clinical conditions with hypertension remains incompletely understood, compelling evidence suggests that hypoxia induces plasticity of the neurocirculatory control system. Despite variations in experimental designs and the severity, frequency, and duration of hypoxia exposure, evidence from animal and human models consistently demonstrates the robust effects of hypoxemia in triggering reflex-mediated sympathetic activation. Both acute and chronic hypoxia alters neurocirculatory regulation and, in some circumstances, leads to sympathetic outflow and elevated blood pressures that persist beyond the hypoxic stimulus. Dysregulation of autonomic control could lead to adverse cardiovascular outcomes and increase the risk of developing hypertension.
Collapse
Affiliation(s)
- Brooke M Shafer
- Centre for Heart, Lung, and Vascular Health, School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia, Canada
| | - Christopher R West
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Chronic Disease Prevention and Management, University of British Columbia, Kelowna, British Columbia, Canada
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Glen E Foster
- Centre for Heart, Lung, and Vascular Health, School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia, Canada
| |
Collapse
|
10
|
Fukushi I, Yokota S, Hasebe Y, Pokorski M, Okada Y. Modulation of respiration and hypothalamus. VITAMINS AND HORMONES 2024; 127:125-152. [PMID: 39864940 DOI: 10.1016/bs.vh.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The hypothalamus is the gray matter of the ventral portion of the diencephalon. The hypothalamus is the higher center of the autonomic nervous system and is involved in the regulation of various homeostatic mechanisms. It also modulates respiration by facilitating the respiratory network. Among subregions of the hypothalamus, the paraventricular nucleus, lateral hypothalamic area, perifornical area, dorsomedial and posterior hypothalamus play particularly important roles in respiratory control. Neurons in these regions have extensive and complex interconnectivity with the cerebral cortex, pons, medulla, spinal cord, and other brain areas. These hypothalamic regions are involved in the maintenance of basal ventilation, respiratory responses to hypoxic and hypercapnic conditions, respiratory augmentation during dynamic exercise, and respiratory modulation in awake and sleep states. Disorders affecting the hypothalamus such as narcolepsy, ROHHAD syndrome, and Prader-Willi syndrome could lead to respiratory abnormalities. However, the role of the hypothalamus in respiratory control, especially its interplay with other local respiratory networks has not yet been fully elucidated. Further clarification of these issues would contribute to a better understanding of the hypothalamus-mediated respiratory control and the pathophysiology of respiratory disorders underlain by hypothalamic dysfunction, as well as to the development of new targeted therapies.
Collapse
Affiliation(s)
- Isato Fukushi
- Graduate School of Health Sciences, Aomori University of Health and Welfare, Aomori, Japan; Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan.
| | - Shigefumi Yokota
- Department of Anatomy and Neuroscience, Shimane University School of Medicine, Izumo, Japan
| | - Yohei Hasebe
- Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan; Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | | | - Yasumasa Okada
- Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan
| |
Collapse
|
11
|
Alvarez-Araos P, Jiménez S, Salazar-Ardiles C, Núñez-Espinosa C, Paez V, Rodriguez-Fernandez M, Raberin A, Millet GP, Iturriaga R, Andrade DC. Baroreflex and chemoreflex interaction in high-altitude exposure: possible role on exercise performance. Front Physiol 2024; 15:1422927. [PMID: 38895516 PMCID: PMC11184637 DOI: 10.3389/fphys.2024.1422927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
The hypoxic chemoreflex and the arterial baroreflex are implicated in the ventilatory response to exercise. It is well known that long-term exercise training increases parasympathetic and decreases sympathetic tone, both processes influenced by the arterial baroreflex and hypoxic chemoreflex function. Hypobaric hypoxia (i.e., high altitude [HA]) markedly reduces exercise capacity associated with autonomic reflexes. Indeed, a reduced exercise capacity has been found, paralleled by a baroreflex-related parasympathetic withdrawal and a pronounced chemoreflex potentiation. Additionally, it is well known that the baroreflex and chemoreflex interact, and during activation by hypoxia, the chemoreflex is predominant over the baroreflex. Thus, the baroreflex function impairment may likely facilitate the exercise deterioration through the reduction of parasympathetic tone following acute HA exposure, secondary to the chemoreflex activation. Therefore, the main goal of this review is to describe the main physiological mechanisms controlling baro- and chemoreflex function and their role in exercise capacity during HA exposure.
Collapse
Affiliation(s)
- Pablo Alvarez-Araos
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura, Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
- Departamento de Kinesiología, Facultad de Ciencias de la Salud, Universidad de Atacama, Copiapó, Chile
| | - Sergio Jiménez
- Departamento de Kinesiología, Facultad de Ciencias de la Salud, Universidad de Atacama, Copiapó, Chile
| | - Camila Salazar-Ardiles
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura, Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Cristian Núñez-Espinosa
- Escuela de Medicina de la Universidad de Magallanes, Punta Arenas, Chile
- Centro Asistencial de Docencia e Investigación (CADI-UMAG), Santiago, Chile
| | - Valeria Paez
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura, Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Maria Rodriguez-Fernandez
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Antoine Raberin
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Gregoire P. Millet
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Rodrigo Iturriaga
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura, Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - David C. Andrade
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura, Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| |
Collapse
|
12
|
Zhu M, Jun S, Nie X, Chen J, Hao Y, Yu H, Zhang X, Sun L, Liu Y, Yuan X, Yuan F, Wang S. Mapping of afferent and efferent connections of phenylethanolamine N-methyltransferase-expressing neurons in the nucleus tractus solitarii. CNS Neurosci Ther 2024; 30:e14808. [PMID: 38887205 PMCID: PMC11183208 DOI: 10.1111/cns.14808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/18/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE Phenylethanolamine N-methyltransferase (PNMT)-expressing neurons in the nucleus tractus solitarii (NTS) contribute to the regulation of autonomic functions. However, the neural circuits linking these neurons to other brain regions remain unclear. This study aims to investigate the connectivity mechanisms of the PNMT-expressing neurons in the NTS (NTSPNMT neurons). METHODS The methodologies employed in this study included a modified rabies virus-based retrograde neural tracing technique, conventional viral anterograde tracing, and immunohistochemical staining procedures. RESULTS A total of 43 upstream nuclei projecting to NTSPNMT neurons were identified, spanning several key brain regions including the medulla oblongata, pons, midbrain, cerebellum, diencephalon, and telencephalon. Notably, dense projections to the NTSPNMT neurons were observed from the central amygdaloid nucleus, paraventricular nucleus of the hypothalamus, area postrema, and the gigantocellular reticular nucleus. In contrast, the ventrolateral medulla, lateral parabrachial nucleus, and lateral hypothalamic area were identified as the primary destinations for axon terminals originating from NTSPNMT neurons. Additionally, reciprocal projections were evident among 21 nuclei, primarily situated within the medulla oblongata. CONCLUSION Our research findings demonstrate that NTSPNMT neurons form extensive connections with numerous nuclei, emphasizing their essential role in the homeostatic regulation of vital autonomic functions.
Collapse
Affiliation(s)
- Mengchu Zhu
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
- Department of Laboratory DiagnosticsHebei Medical UniversityShijiazhuangHebeiChina
| | - Shirui Jun
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Xiaojun Nie
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Jinting Chen
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Yinchao Hao
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Hongxiao Yu
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Xiang Zhang
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Lu Sun
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Yuelin Liu
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Xiangshan Yuan
- Department of Anatomy and Histoembryology, School of Basic Medical SciencesFudan UniversityShanghaiChina
- Department of NeurologyJinshan Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Fang Yuan
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
- Hebei Key Laboratory of NeurophysiologyShijiazhuangHebei ProvinceChina
| | - Sheng Wang
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
- Hebei Key Laboratory of NeurophysiologyShijiazhuangHebei ProvinceChina
| |
Collapse
|
13
|
Kawamura M, Yoshimoto A, Ikegaya Y, Matsumoto N. Low Atmospheric Oxygen Attenuates Alpha Oscillations in the Primary Motor Cortex of Awake Rats. Biol Pharm Bull 2024; 47:462-468. [PMID: 38382999 DOI: 10.1248/bpb.b23-00863] [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] [Indexed: 02/23/2024]
Abstract
Oxygen is pivotal for survival of animals. Their cellular activity and cognitive behavior are impaired when atmospheric oxygen is insufficient, called hypoxia. However, concurrent effects of hypoxia on physiological signals are poorly understood. To address this question, we simultaneously recorded local field potentials in the primary motor cortex, primary somatosensory, and anterior cingulate cortex, electrocardiograms, electroolfactograms, and electromyograms of rats under acute hypoxic conditions (i.e., 5.0% O2). Exposure to acute hypoxia significantly attenuated alpha oscillations alone in the primary motor cortex, while we failed to find any effects of acute hypoxia on the oscillatory power in the somatosensory cortex or anterior cingulate cortex. These area- and frequency-specific effects by hypoxia may be accounted for by neural innervation from the brainstem to each cortical area via thalamic relay nuclei. Moreover, we found that heart rate and respiratory rate were increased during acute hypoxia and high heart rate was maintained even after the oxygen level returned to the baseline. Altogether, our study characterizes a systemic effect of atmospheric hypoxia on neural and peripheral signals from physiological viewpoints, leading to bridging a gap between cellular and behavioral levels.
Collapse
Affiliation(s)
- Masashi Kawamura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Airi Yoshimoto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
- Institute for AI and Beyond, The University of Tokyo
- Center for Information and Neural Networks, National Institute of Information and Communications Technology
| | - Nobuyoshi Matsumoto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
- Institute for AI and Beyond, The University of Tokyo
| |
Collapse
|
14
|
Ahmadzadeh E, Polglase GR, Stojanovska V, Herlenius E, Walker DW, Miller SL, Allison BJ. Does fetal growth restriction induce neuropathology within the developing brainstem? J Physiol 2023; 601:4667-4689. [PMID: 37589339 PMCID: PMC10953350 DOI: 10.1113/jp284191] [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: 01/29/2023] [Accepted: 08/04/2023] [Indexed: 08/18/2023] Open
Abstract
Fetal growth restriction (FGR) is a complex obstetric issue describing a fetus that does not reach its genetic growth potential. The primary cause of FGR is placental dysfunction resulting in chronic fetal hypoxaemia, which in turn causes altered neurological, cardiovascular and respiratory development, some of which may be pathophysiological, particularly for neonatal life. The brainstem is the critical site of cardiovascular, respiratory and autonomic control, but there is little information describing how chronic hypoxaemia and the resulting FGR may affect brainstem neurodevelopment. This review provides an overview of the brainstem-specific consequences of acute and chronic hypoxia, and what is known in FGR. In addition, we discuss how brainstem structural alterations may impair functional control of the cardiovascular and respiratory systems. Finally, we highlight the clinical and translational findings of the potential roles of the brainstem in maintaining cardiorespiratory adaptation in the transition from fetal to neonatal life under normal conditions and in response to the pathological environment that arises during development in growth-restricted infants. This review emphasises the crucial role that the brainstem plays in mediating cardiovascular and respiratory responses during fetal and neonatal life. We assess whether chronic fetal hypoxaemia might alter structure and function of the brainstem, but this also serves to highlight knowledge gaps regarding FGR and brainstem development.
Collapse
Affiliation(s)
- Elham Ahmadzadeh
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVictoriaAustralia
| | - Graeme R. Polglase
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVictoriaAustralia
| | - Vanesa Stojanovska
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVictoriaAustralia
| | - Eric Herlenius
- Department of Women's and Children's HealthKarolinska InstitutetSolnaSweden
- Astrid Lindgren Children´s HospitalKarolinska University Hospital StockholmSolnaSweden
| | - David W. Walker
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical SciencesRoyal Melbourne Institute of Technology (RMIT)MelbourneVictoriaAustralia
| | - Suzanne L. Miller
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVictoriaAustralia
| | - Beth J. Allison
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
15
|
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: 9] [Impact Index Per Article: 4.5] [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
|
16
|
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: 9] [Impact Index Per Article: 4.5] [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
|
17
|
Rasiah NP, Loewen SP, Bains JS. Windows into stress: a glimpse at emerging roles for CRH PVN neurons. Physiol Rev 2023; 103:1667-1691. [PMID: 36395349 DOI: 10.1152/physrev.00056.2021] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The corticotropin-releasing hormone cells in the paraventricular nucleus of the hypothalamus (CRHPVN) control the slow endocrine response to stress. The synapses on these cells are exquisitely sensitive to acute stress, leveraging local signals to leave a lasting imprint on this system. Additionally, recent work indicates that these cells also play key roles in the control of distinct stress and survival behaviors. Here we review these observations and provide a perspective on the role of CRHPVN neurons as integrative and malleable hubs for behavioral, physiological, and endocrine responses to stress.
Collapse
Affiliation(s)
- Neilen P Rasiah
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Spencer P Loewen
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jaideep S Bains
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
18
|
Krohn F, Novello M, van der Giessen RS, De Zeeuw CI, Pel JJM, Bosman LWJ. The integrated brain network that controls respiration. eLife 2023; 12:83654. [PMID: 36884287 PMCID: PMC9995121 DOI: 10.7554/elife.83654] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/29/2023] [Indexed: 03/09/2023] Open
Abstract
Respiration is a brain function on which our lives essentially depend. Control of respiration ensures that the frequency and depth of breathing adapt continuously to metabolic needs. In addition, the respiratory control network of the brain has to organize muscular synergies that integrate ventilation with posture and body movement. Finally, respiration is coupled to cardiovascular function and emotion. Here, we argue that the brain can handle this all by integrating a brainstem central pattern generator circuit in a larger network that also comprises the cerebellum. Although currently not generally recognized as a respiratory control center, the cerebellum is well known for its coordinating and modulating role in motor behavior, as well as for its role in the autonomic nervous system. In this review, we discuss the role of brain regions involved in the control of respiration, and their anatomical and functional interactions. We discuss how sensory feedback can result in adaptation of respiration, and how these mechanisms can be compromised by various neurological and psychological disorders. Finally, we demonstrate how the respiratory pattern generators are part of a larger and integrated network of respiratory brain regions.
Collapse
Affiliation(s)
- Friedrich Krohn
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Manuele Novello
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands.,Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Johan J M Pel
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | |
Collapse
|
19
|
Ostrowski D, Heesch CM, Kline DD, Hasser EM. Nucleus tractus solitarii is required for the development and maintenance of phrenic and sympathetic long-term facilitation after acute intermittent hypoxia. Front Physiol 2023; 14:1120341. [PMID: 36846346 PMCID: PMC9949380 DOI: 10.3389/fphys.2023.1120341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
Exposure to acute intermittent hypoxia (AIH) induces prolonged increases (long term facilitation, LTF) in phrenic and sympathetic nerve activity (PhrNA, SNA) under basal conditions, and enhanced respiratory and sympathetic responses to hypoxia. The mechanisms and neurocircuitry involved are not fully defined. We tested the hypothesis that the nucleus tractus solitarii (nTS) is vital to augmentation of hypoxic responses and the initiation and maintenance of elevated phrenic (p) and splanchnic sympathetic (s) LTF following AIH. nTS neuronal activity was inhibited by nanoinjection of the GABAA receptor agonist muscimol before AIH exposure or after development of AIH-induced LTF. AIH but not sustained hypoxia induced pLTF and sLTF with maintained respiratory modulation of SSNA. nTS muscimol before AIH increased baseline SSNA with minor effects on PhrNA. nTS inhibition also markedly blunted hypoxic PhrNA and SSNA responses, and prevented altered sympathorespiratory coupling during hypoxia. Inhibiting nTS neuronal activity before AIH exposure also prevented the development of pLTF during AIH and the elevated SSNA after muscimol did not increase further during or following AIH exposure. Furthermore, nTS neuronal inhibition after the development of AIH-induced LTF substantially reversed but did not eliminate the facilitation of PhrNA. Together these findings demonstrate that mechanisms within the nTS are critical for initiation of pLTF during AIH. Moreover, ongoing nTS neuronal activity is required for full expression of sustained elevations in PhrNA following exposure to AIH although other regions likely also are important. Together, the data indicate that AIH-induced alterations within the nTS contribute to both the development and maintenance of pLTF.
Collapse
Affiliation(s)
- Daniela Ostrowski
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States,Department of Biology, Truman State University, Kirksville, MO, United States
| | - Cheryl M. Heesch
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - David D. Kline
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | - Eileen M. Hasser
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States,*Correspondence: Eileen M. Hasser,
| |
Collapse
|
20
|
Maletz SN, Reid BT, Varga AG, Levitt ES. Nucleus Tractus Solitarius Neurons Activated by Hypercapnia and Hypoxia Lack Mu Opioid Receptor Expression. Front Mol Neurosci 2022; 15:932189. [PMID: 35898697 PMCID: PMC9309891 DOI: 10.3389/fnmol.2022.932189] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022] Open
Abstract
Impaired chemoreflex responses are a central feature of opioid-induced respiratory depression, however, the mechanism through which mu opioid receptor agonists lead to diminished chemoreflexes is not fully understood. One brainstem structure involved in opioid-induced impairment of chemoreflexes is the nucleus of the solitary tract (NTS), which contains a population of neurons that express mu opioid receptors. Here, we tested whether caudal NTS neurons activated during the chemoreflex challenge express mu opioid receptors and overlap with neurons activated by opioids. Using genetic labeling of mu opioid receptor-expressing neurons and cFos immunohistochemistry as a proxy for neuronal activation, we examined the distribution of activated NTS neurons following hypercapnia, hypoxia, and morphine administration. The main finding was that hypoxia and hypercapnia primarily activated NTS neurons that did not express mu opioid receptors. Furthermore, concurrent administration of morphine with hypercapnia induced cFos expression in non-overlapping populations of neurons. Together these results suggest an indirect effect of opioids within the NTS, which could be mediated through mu opioid receptors on afferents and/or inhibitory interneurons.
Collapse
Affiliation(s)
- Sebastian N. Maletz
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
| | - Brandon T. Reid
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
| | - Adrienn G. Varga
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, United States
| | - Erica S. Levitt
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, United States
- *Correspondence: Erica S. Levitt ; orcid.org/0000-0002-3634-6594
| |
Collapse
|
21
|
Schäfer E, Scheer C, Saljé K, Fritz A, Kohlmann T, Hübner NO, Napp M, Fiedler-Lacombe L, Stahl D, Rauch B, Nauck M, Völker U, Felix S, Lucchese G, Flöel A, Engeli S, Hoffmann W, Hahnenkamp K, Tzvetkov MV. Course of disease and risk factors for hospitalization in outpatients with a SARS-CoV-2 infection. Sci Rep 2022; 12:7249. [PMID: 35508524 PMCID: PMC9065670 DOI: 10.1038/s41598-022-11103-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 04/11/2022] [Indexed: 12/02/2022] Open
Abstract
We analyzed symptoms and comorbidities as predictors of hospitalization in 710 outpatients in North-East Germany with PCR-confirmed SARS-CoV-2 infection. During the first 3 days of infection, commonly reported symptoms were fatigue (71.8%), arthralgia/myalgia (56.8%), headache (55.1%), and dry cough (51.8%). Loss of smell (anosmia), loss of taste (ageusia), dyspnea, and productive cough were reported with an onset of 4 days. Anosmia or ageusia were reported by only 18% of the participants at day one, but up to 49% between days 7 and 9. Not all participants who reported ageusia also reported anosmia. Individuals suffering from ageusia without anosmia were at highest risk of hospitalization (OR 6.8, 95% CI 2.5-18.1). They also experienced more commonly dyspnea and nausea (OR of 3.0, 2.9, respectively) suggesting pathophysiological connections between these symptoms. Other symptoms significantly associated with increased risk of hospitalization were dyspnea, vomiting, and fever. Among basic parameters and comorbidities, age > 60 years, COPD, prior stroke, diabetes, kidney and cardiac diseases were also associated with increased risk of hospitalization. In conclusion, due to the delayed onset, ageusia and anosmia may be of limited use in differential diagnosis of SARS-CoV-2. However, differentiation between ageusia and anosmia may be useful for evaluating risk for hospitalization.
Collapse
Affiliation(s)
- Eik Schäfer
- Department of Clinical Pharmacology, Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany
- Department of Anesthesiology, University Medicine Greifswald, Greifswald, Germany
| | - Christian Scheer
- Department of Anesthesiology, University Medicine Greifswald, Greifswald, Germany
| | - Karen Saljé
- Department of Clinical Pharmacology, Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany
| | - Anja Fritz
- Department of General Pharmacology, Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, 17489, Greifswald, Germany
| | - Thomas Kohlmann
- Institute for Community Medicine, Section Epidemiology of Health Care and Community Health, University Medicine Greifswald, Greifswald, Germany
| | - Nils-Olaf Hübner
- Central Unit for Infection Prevention and Control, University Medicine Greifswald, Greifswald, Germany
- Institute of Hygiene and Environmental Medicine, University of Greifswald, Greifswald, Germany
| | - Matthias Napp
- Departments of Emergency and Acute Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Lizon Fiedler-Lacombe
- Independent Trusted Third Party, University Medicine Greifswald, Greifswald, Germany
| | - Dana Stahl
- Independent Trusted Third Party, University Medicine Greifswald, Greifswald, Germany
| | - Bernhard Rauch
- Department of General Pharmacology, Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, 17489, Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Uwe Völker
- Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Stephan Felix
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, Cardiology, Pneumology, Infectious Diseases, Intensive Care Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Guglielmo Lucchese
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Agnes Flöel
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Engeli
- Department of Clinical Pharmacology, Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany
| | - Wolfgang Hoffmann
- Institute for Community Medicine, Section Epidemiology of Health Care and Community Health, University Medicine Greifswald, Greifswald, Germany
| | - Klaus Hahnenkamp
- Department of Anesthesiology, University Medicine Greifswald, Greifswald, Germany
| | - Mladen V Tzvetkov
- Department of General Pharmacology, Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, 17489, Greifswald, Germany.
| |
Collapse
|
22
|
Katayama PL, Leirão IP, Kanashiro A, Luiz JPM, Cunha FQ, Navegantes LCC, Menani JV, Zoccal DB, Colombari DSA, Colombari E. The carotid body detects circulating tumor necrosis factor-alpha to activate a sympathetic anti-inflammatory reflex. Brain Behav Immun 2022; 102:370-386. [PMID: 35339628 DOI: 10.1016/j.bbi.2022.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/10/2022] [Accepted: 03/21/2022] [Indexed: 12/28/2022] Open
Abstract
Recent evidence has suggested that the carotid bodies might act as immunological sensors, detecting pro-inflammatory mediators and signalling to the central nervous system, which, in turn, orchestrates autonomic responses. Here, we confirmed that the TNF-α receptor type I is expressed in the carotid bodies of rats. The systemic administration of TNF-α increased carotid body afferent discharge and activated glutamatergic neurons in the nucleus tractus solitarius (NTS) that project to the rostral ventrolateral medulla (RVLM), where many pre-sympathetic neurons reside. The activation of these neurons was accompanied by an increase in splanchnic sympathetic nerve activity. Carotid body ablation blunted the TNF-α-induced activation of RVLM-projecting NTS neurons and the increase in splanchnic sympathetic nerve activity. Finally, plasma and spleen levels of cytokines after TNF-α administration were higher in rats subjected to either carotid body ablation or splanchnic sympathetic denervation. Collectively, our findings indicate that the carotid body detects circulating TNF-α to activate a counteracting sympathetic anti-inflammatory mechanism.
Collapse
Affiliation(s)
- Pedro L Katayama
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University, Araraquara, São Paulo, Brazil.
| | - Isabela P Leirão
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Alexandre Kanashiro
- Department of Neurosciences and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - João P M Luiz
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Luiz C C Navegantes
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Jose V Menani
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Daniel B Zoccal
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Débora S A Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Eduardo Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University, Araraquara, São Paulo, Brazil.
| |
Collapse
|
23
|
Joyce W, Wang T. Regulation of heart rate in vertebrates during hypoxia: A comparative overview. Acta Physiol (Oxf) 2022; 234:e13779. [PMID: 34995393 DOI: 10.1111/apha.13779] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/12/2021] [Accepted: 01/01/2022] [Indexed: 12/18/2022]
Abstract
Acute exposure to low oxygen (hypoxia) places conflicting demands on the heart. Whilst an increase in heart rate (tachycardia) may compensate systemic oxygen delivery as arterial oxygenation falls, the heart itself is an energetically expensive organ that may benefit from slowing (bradycardia) to reduce work when oxygen is limited. Both strategies are apparent in vertebrates, with tetrapods (mammals, birds, reptiles, and amphibians) classically exhibiting hypoxic tachycardia and fishes displaying characteristic hypoxic bradycardia. With a richer understanding of the ontogeny and evolution of the responses, however, we see similarities in the underlying mechanisms between vertebrate groups. For example, in adult mammals, primary bradycardia results from the hypoxic stimulation of carotid body chemoreceptors that are overwhelmed by mechano-sensory feedback from the lung associated with hyperpnoea. Fish-like bradycardia prevails in the mammalian foetus (which, at this stage, is incapable of pulmonary ventilation), and in fish and foetus alike, the bradycardia ensues despite an elevation of circulating catecholamines. In both cases, the reduced heart rate may primarily serve to protect the heart. Thus, the comparative perspective offers fundamental insight into how and why different vertebrates regulate heart rate in different ways during periods of hypoxia.
Collapse
Affiliation(s)
- William Joyce
- Department of Biology—Zoophysiology Aarhus University Aarhus C Denmark
| | - Tobias Wang
- Department of Biology—Zoophysiology Aarhus University Aarhus C Denmark
| |
Collapse
|
24
|
Kisielinski K, Giboni P, Prescher A, Klosterhalfen B, Graessel D, Funken S, Kempski O, Hirsch O. Is a Mask That Covers the Mouth and Nose Free from Undesirable Side Effects in Everyday Use and Free of Potential Hazards? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:4344. [PMID: 33923935 PMCID: PMC8072811 DOI: 10.3390/ijerph18084344] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022]
Abstract
Many countries introduced the requirement to wear masks in public spaces for containing SARS-CoV-2 making it commonplace in 2020. Up until now, there has been no comprehensive investigation as to the adverse health effects masks can cause. The aim was to find, test, evaluate and compile scientifically proven related side effects of wearing masks. For a quantitative evaluation, 44 mostly experimental studies were referenced, and for a substantive evaluation, 65 publications were found. The literature revealed relevant adverse effects of masks in numerous disciplines. In this paper, we refer to the psychological and physical deterioration as well as multiple symptoms described because of their consistent, recurrent and uniform presentation from different disciplines as a Mask-Induced Exhaustion Syndrome (MIES). We objectified evaluation evidenced changes in respiratory physiology of mask wearers with significant correlation of O2 drop and fatigue (p < 0.05), a clustered co-occurrence of respiratory impairment and O2 drop (67%), N95 mask and CO2 rise (82%), N95 mask and O2 drop (72%), N95 mask and headache (60%), respiratory impairment and temperature rise (88%), but also temperature rise and moisture (100%) under the masks. Extended mask-wearing by the general population could lead to relevant effects and consequences in many medical fields.
Collapse
Affiliation(s)
| | | | - Andreas Prescher
- Institute of Molecular and Cellular Anatomy (MOCA), Wendlingweg 2, 52074 Aachen, Germany;
| | - Bernd Klosterhalfen
- Institute of Pathology, Dueren Hospital, Roonstrasse 30, 52351 Dueren, Germany;
| | - David Graessel
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, 52425 Jülich, Germany;
| | | | - Oliver Kempski
- Institute of Neurosurgical Pathophysiology, University Medical Centre of the Johannes Gutenberg University of Mainz Langenbeckstr. 1, 55131 Mainz, Germany;
| | - Oliver Hirsch
- Department of Psychology, FOM University of Applied Sciences, 57078 Siegen, Germany
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Pascoaloti-Lima JC, Machado BH, Accorsi-Mendonça D. Sustained Hypoxia Reduces GABAergic Modulation on NTS Neurons Sending Projections to Ventral Medulla of Rats. Neuroscience 2021; 457:1-11. [PMID: 33421568 DOI: 10.1016/j.neuroscience.2020.12.028] [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: 07/10/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 11/26/2022]
Abstract
Peripheral chemoreflex is activated during short-term sustained hypoxia (SH), and the first synapse of these afferents is located in Nucleus Tractus Solitarius(NTS). NTS neurons projecting to the ventral lateral medulla (NTS-VLM) are part of the respiratory pathways of the chemoreflex. SH increases the magnitude of basal respiratory parameters in rats from Wistar-Hannover strain. In this study, we hypothesized that the observed changes in the respiratory pattern in response to SH were due to changes in the GABAergic modulation of the synaptic transmission of NTS-VLM neurons. We used an electrophysiological approach to record the synaptic activity of NTS neurons labeled with a retrograde tracer previously microinjected into VLM of Wistar-Hannover rats submitted to 24 h SH. The data are showing that: (a) the amplitude of evoked inhibitory currents in NTS-VLM neurons of SH rats was reduced and not accompanied by any change in rise-time and decay-time; (b) the 1/CV2 and the number of failures in response to evoked currents were also affected by SH; (c) the frequency of spontaneous inhibitory currents was reduced by SH without changes in amplitude and half-width. These effects of SH were observed in NTS-VLM neurons located in caudal and intermediate NTS, but not in NTS-VLM neurons located in the rostral NTS. We conclude that SH causes a reduction in inhibitory modulation onto NTS-VLM neurons by pre-synaptic mechanisms, which may contribute to the observed changes in the respiratory pattern of Wistar-Hannover rats submitted to SH.
Collapse
Affiliation(s)
- Júlio C Pascoaloti-Lima
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Benedito H Machado
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Daniela Accorsi-Mendonça
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
28
|
Wang W, Zheng Y, Li M, Lin S, Lin H. Recent Advances in Studies on the Role of Neuroendocrine Disorders in Obstructive Sleep Apnea-Hypopnea Syndrome-Related Atherosclerosis. Nat Sci Sleep 2021; 13:1331-1345. [PMID: 34349578 PMCID: PMC8326525 DOI: 10.2147/nss.s315375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is a common cause of death worldwide, and atherosclerosis (AS) and obstructive sleep apnea-hypopnea syndrome (OSAHS) critically contribute to the initiation and progression of cardiovascular diseases. OSAHS promotes endothelial injury, vascular smooth muscle cell (VSMC) proliferation, abnormal lipid metabolism, and elevated arterial blood pressure. However, the exact OSAHS mechanism that causes AS remains unclear. The nervous system is widely distributed in the central and peripheral regions. It regulates appetite, energy metabolism, inflammation, oxidative stress, insulin resistance, and vasoconstriction by releasing regulatory factors and participates in the occurrence and development of AS. Studies showed that OSAHS can cause changes in neurophysiological plasticity and affect modulator release, suggesting that neuroendocrine dysfunction may be related to the OSAHS mechanism causing AS. In this article, we review the possible mechanisms of neuroendocrine disorders in the pathogenesis of OSAHS-induced AS and provide a new basis for further research on the development of corresponding effective intervention strategies.
Collapse
Affiliation(s)
- Wanda Wang
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| | - Yanli Zheng
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| | - Meimei Li
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| | - Shu Lin
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China.,Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China.,Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | - Huili Lin
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| |
Collapse
|
29
|
Martinez D, Rogers RC, Hasser EM, Hermann GE, Kline DD. Loss of excitatory amino acid transporter restraint following chronic intermittent hypoxia contributes to synaptic alterations in nucleus tractus solitarii. J Neurophysiol 2020; 123:2122-2135. [PMID: 32347148 PMCID: PMC7311725 DOI: 10.1152/jn.00766.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Peripheral viscerosensory afferent signals are transmitted to the nucleus tractus solitarii (nTS) via release of glutamate. Following release, glutamate is removed from the extrasynaptic and synaptic cleft via excitatory amino acid transporters (EAATs), thus limiting glutamate receptor activation or over activation, and maintaining its working range. We have shown that EAAT block with the antagonist threo-β-benzyloxyaspartic acid (TBOA) depolarized nTS neurons and increased spontaneous excitatory postsynaptic current (sEPSC) frequency yet reduced the amplitude of afferent (TS)-evoked EPSCs (TS-EPSCs). Interestingly, chronic intermittent hypoxia (CIH), a model of obstructive sleep apnea (OSA), produces similar synaptic responses as EAAT block. We hypothesized EAAT expression or function are downregulated after CIH, and this reduction in glutamate removal contributes to the observed neurophysiological responses. To test this hypothesis, we used brain slice electrophysiology and imaging of glutamate release and TS-afferent Ca2+ to compare nTS properties of rats exposed to 10 days of normoxia (Norm; 21%O2) or CIH. Results show that EAAT blockade with (3S)-3-[[3-[[4-(trifluoromethyl)benzoyl]-amino]phenyl]methoxy]-l-aspartic acid (TFB-TBOA) in Norm caused neuronal depolarization, generation of an inward current, and increased spontaneous synaptic activity. The latter augmentation was eliminated by inclusion of tetrodotoxin in the perfusate. TS stimulation during TFB-TBOA also elevated extracellular glutamate and decreased presynaptic Ca2+ and TS-EPSC amplitude. In CIH, the effects of EAAT block are eliminated or attenuated. CIH reduced EAAT expression in nTS, which may contribute to the attenuated function seen in this condition. Therefore, CIH reduces EAAT influence on synaptic and neuronal activity, which may lead to the physiological consequences seen in OSA and CIH.NEW & NOTEWORTHY Removal of excitatory amino acid transporter (EAAT) restraint increases spontaneous synaptic activity yet decreases afferent [tractus solitarius (TS)]-driven excitatory postsynaptic current (EPSC) amplitude. In the chronic intermittent hypoxia model of obstructive sleep apnea, this restraint is lost due to reduction in EAAT expression and function. Thus EAATs are important in controlling elevated glutamatergic signaling, and loss of such control results in maladaptive synaptic signaling.
Collapse
Affiliation(s)
- Diana Martinez
- 1Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | | | - Eileen M. Hasser
- 1Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,2Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | | | - David D. Kline
- 1Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
30
|
Evans AM, Hardie DG. AMPK and the Need to Breathe and Feed: What's the Matter with Oxygen? Int J Mol Sci 2020; 21:ijms21103518. [PMID: 32429235 PMCID: PMC7279029 DOI: 10.3390/ijms21103518] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
We live and to do so we must breathe and eat, so are we a combination of what we eat and breathe? Here, we will consider this question, and the role in this respect of the AMP-activated protein kinase (AMPK). Emerging evidence suggests that AMPK facilitates central and peripheral reflexes that coordinate breathing and oxygen supply, and contributes to the central regulation of feeding and food choice. We propose, therefore, that oxygen supply to the body is aligned with not only the quantity we eat, but also nutrient-based diet selection, and that the cell-specific expression pattern of AMPK subunit isoforms is critical to appropriate system alignment in this respect. Currently available information on how oxygen supply may be aligned with feeding and food choice, or vice versa, through our motivation to breathe and select particular nutrients is sparse, fragmented and lacks any integrated understanding. By addressing this, we aim to provide the foundations for a clinical perspective that reveals untapped potential, by highlighting how aberrant cell-specific changes in the expression of AMPK subunit isoforms could give rise, in part, to known associations between metabolic disease, such as obesity and type 2 diabetes, sleep-disordered breathing, pulmonary hypertension and acute respiratory distress syndrome.
Collapse
Affiliation(s)
- A. Mark Evans
- Centre for Discovery Brain Sciences and Cardiovascular Science, Edinburgh Medical School, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK
- Correspondence:
| | - D. Grahame Hardie
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK;
| |
Collapse
|
31
|
Shimoura CG, Andrade MA, Toney GM. Central AT1 receptor signaling by circulating angiotensin II is permissive to acute intermittent hypoxia-induced sympathetic neuroplasticity. J Appl Physiol (1985) 2020; 128:1329-1337. [PMID: 32240022 DOI: 10.1152/japplphysiol.00094.2020] [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: 01/16/2023] Open
Abstract
Acute intermittent hypoxia (AIH) triggers sympathetic long-term facilitation (sLTF), a progressive increase in sympathetic nerve activity (SNA) linked to central AT1 receptor (AT1R) activation by circulating angiotensin II (ANG II). Here, we investigated AIH activation of the peripheral renin-angiotensin system (RAS) and the extent to which the magnitude of RAS activation predicts the magnitude of AIH-induced sLTF. In anesthetized male Sprague-Dawley rats, plasma renin activity (PRA) increased in a linear fashion in response to 5 (P = 0.0342) and 10 (P < 0.0001) cycles of AIH, with PRA remaining at the 10th cycle level 1 h later, a period over which SNA progressively increased. On average, SNA ramping began at the AIH cycle 4.6 ± 0.9 (n = 12) and was similar in magnitude 1 h later whether AIH consisted of 5 or 10 cycles (n = 6/group). Necessity of central AT1R in post-AIH sLTF was affirmed by intracerebroventricular (icv) losartan (40 nmol, 2 µL; n = 5), which strongly attenuated both splanchnic (P = 0.0469) and renal (P = 0.0018) sLTF compared with vehicle [artificial cerebrospinal fluid (aCSF), 2 µL; n = 5]. Bilateral nephrectomy largely prevented sLTF, affirming the necessity of peripheral RAS activation. Sufficiency of central ANG II signaling was assessed in nephrectomized rats. Whereas ICV ANG II (0.5 ng/0.5 µL, 30 min) in nephrectomized rats exposed to sham AIH (n = 4) failed to cause SNA ramping, it rescued sLTF in nephrectomized rats exposed to five cycles of AIH [splanchnic SNA (SSNA), P = 0.0227; renal SNA (RSNA), P = 0.0390; n = 5]. Findings indicate that AIH causes progressive peripheral RAS activation, which stimulates an apparent threshold level of central AT1R signaling that plays a permissive role in triggering sLTF.NEW & NOTEWORTHY Acute intermittent hypoxia (AIH) triggers sympathetic long-term facilitation (sLTF) that relies on peripheral renin-angiotensin system (RAS) activation. Here, increasing AIH cycles from 5 to 10 proportionally increased RAS activity, but not the magnitude of post-AIH sLTF. Brain angiotensin II (ANG II) receptor blockade and nephrectomy each largely prevented sLTF, whereas central ANG II rescued it following nephrectomy. Peripheral RAS activation by AIH induces time-dependent neuroplasticity at an apparent central ANG II signaling threshold, triggering a stereotyped sLTF response.
Collapse
Affiliation(s)
- Caroline G Shimoura
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Mary Ann Andrade
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Glenn M Toney
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas.,Center for Biomedical Neuroscience, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
32
|
Martinez D, Rogers RC, Hermann GE, Hasser EM, Kline DD. Astrocytic glutamate transporters reduce the neuronal and physiological influence of metabotropic glutamate receptors in nucleus tractus solitarii. Am J Physiol Regul Integr Comp Physiol 2020; 318:R545-R564. [PMID: 31967862 PMCID: PMC7099463 DOI: 10.1152/ajpregu.00319.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Astrocytic excitatory amino acid transporters (EAATs) are critical to restraining synaptic and neuronal activity in the nucleus tractus solitarii (nTS). Relief of nTS EAAT restraint generates two opposing effects, an increase in neuronal excitability that reduces blood pressure and breathing and an attenuation in afferent [tractus solitarius (TS)]-driven excitatory postsynaptic current (EPSC) amplitude. Although the former is due, in part, to activation of ionotropic glutamate receptors, there remains a substantial contribution from another unidentified glutamate receptor. In addition, the mechanism(s) by which EAAT inhibition reduced TS-EPSC amplitude is unknown. Metabotropic glutamate receptors (mGluRs) differentially modulate nTS excitability. Activation of group I mGluRs on nTS neuron somas leads to depolarization, whereas group II/III mGluRs on sensory afferents decrease TS-EPSC amplitude. Thus we hypothesize that EAATs control postsynaptic excitability and TS-EPSC amplitude via restraint of mGluR activation. To test this hypothesis, we used in vivo recording, brain slice electrophysiology, and imaging of glutamate release and TS-afferent Ca2+. Results show that EAAT blockade in the nTS with (3S)-3-[[3-[[4-(trifluoromethyl)benzoyl]amino]phenyl]methoxy]-l-aspartic acid (TFB-TBOA) induced group I mGluR-mediated depressor, bradycardic, and apneic responses that were accompanied by neuronal depolarization, elevated discharge, and increased spontaneous synaptic activity. Conversely, upon TS stimulation TFB-TBOA elevated extracellular glutamate to decrease presynaptic Ca2+ and TS-EPSC amplitude via activation of group II/III mGluRs. Together, these data suggest an important role of EAATs in restraining mGluR activation and overall cardiorespiratory function.
Collapse
Affiliation(s)
- Diana Martinez
- 1Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | | | | | - Eileen M. Hasser
- 1Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,2Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - David D. Kline
- 1Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
33
|
Venkataraman S, Vungarala S, Covassin N, Somers VK. Sleep Apnea, Hypertension and the Sympathetic Nervous System in the Adult Population. J Clin Med 2020; 9:jcm9020591. [PMID: 32098169 PMCID: PMC7073618 DOI: 10.3390/jcm9020591] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/16/2020] [Accepted: 02/19/2020] [Indexed: 02/06/2023] Open
Abstract
Sleep apnea is very common in patients with cardiovascular disease, especially in patients with hypertension. Over the last few decades a number of discoveries have helped support a causal relationship between the two and even resistant hypertension. The role neurogenic mechanisms play has gathered more attention in the recent past due to their immediate bedside utility. Several innovative discoveries in pathogenesis including those exploring the role of baroreflex gain, cardiovascular variability, chemoreceptor reflex activation and the sympathetic nervous system have emerged. In this review, we discuss the epidemiology of sleep apnea and hypertension and the pathogenic mechanisms contributing to neurogenic hypertension. Furthermore, recent management strategies in addition to continuous positive airway pressure (CPAP), such as upper airway stimulation and renal denervation that target these pathogenic mechanisms, are also discussed.
Collapse
|
34
|
Zera T, Moraes DJA, da Silva MP, Fisher JP, Paton JFR. The Logic of Carotid Body Connectivity to the Brain. Physiology (Bethesda) 2020; 34:264-282. [PMID: 31165684 DOI: 10.1152/physiol.00057.2018] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The carotid body has emerged as a therapeutic target for cardio-respiratory-metabolic diseases. With the expansive functions of the chemoreflex, we sought mechanisms to explain differential control of individual responses. We purport a remarkable correlation between phenotype of a chemosensory unit (glomus cell-sensory afferent) with a distinct component of the reflex response. This logic could permit differential modulation of distinct chemoreflex responses, a strategy ideal for therapeutic exploitation.
Collapse
Affiliation(s)
- Tymoteusz Zera
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw , Warsaw , Poland
| | - Davi J A Moraes
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo , São Paulo , Brazil
| | - Melina P da Silva
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo , São Paulo , Brazil
| | - James P Fisher
- Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland , Auckland , New Zealand
| | - Julian F R Paton
- Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland , Auckland , New Zealand
| |
Collapse
|
35
|
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: 3.3] [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
|
36
|
Milanick WJ, Polo-Parada L, Dantzler HA, Kline DD. Activation of alpha-1 adrenergic receptors increases cytosolic calcium in neurones of the paraventricular nucleus of the hypothalamus. J Neuroendocrinol 2019; 31:e12791. [PMID: 31494990 PMCID: PMC7003713 DOI: 10.1111/jne.12791] [Citation(s) in RCA: 5] [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: 03/15/2019] [Revised: 08/22/2019] [Accepted: 09/04/2019] [Indexed: 12/15/2022]
Abstract
Norepinephrine (NE) activates adrenergic receptors (ARs) in the hypothalamic paraventricular nucleus (PVN) to increase excitatory currents, depolarise neurones and, ultimately, augment neuro-sympathetic and endocrine output. Such cellular events are known to potentiate intracellular calcium ([Ca2+ ]i ); however, the role of NE with respect to modulating [Ca2+ ]i in PVN neurones and the mechanisms by which this may occur remain unclear. We evaluated the effects of NE on [Ca2+ ]i of acutely isolated PVN neurones using Fura-2 imaging. NE induced a slow increase in [Ca2+ ]i compared to artificial cerebrospinal fluid vehicle. NE-induced Ca2+ elevations were mimicked by the α1 -AR agonist phenylephrine (PE) but not by α2 -AR agonist clonidine (CLON). NE and PE but not CLON also increased the overall number of neurones that increase [Ca2+ ]i (ie, responders). Elimination of extracellular Ca2+ or intracellular endoplasmic reticulum Ca2+ stores abolished the increase in [Ca2+ ]i and reduced responders. Blockade of voltage-dependent Ca2+ channels abolished the α1 -AR induced increase in [Ca2+ ]i and number of responders, as did inhibition of phospholipase C inhibitor, protein kinase C and inositol triphosphate receptors. Spontaneous phasic Ca2+ events, however, were not altered by NE, PE or CLON. Repeated K+ -induced membrane depolarisation produced repetitive [Ca2+ ]i elevations. NE and PE increased baseline Ca2+ , whereas NE decreased the peak amplitude. CLON also decreased peak amplitude but did not affect baseline [Ca2+ ]i . Taken together, these data suggest receptor-specific influence of α1 and α2 receptors on the various modes of calcium entry in PVN neurones. They further suggest Ca2+ increase via α1 -ARs is co-dependent on extracellular Ca2+ influx and intracellular Ca2+ release, possibly via a phospholipase C inhibitor-mediated signalling cascade.
Collapse
Affiliation(s)
- William J. Milanick
- Department of Biomedical Sciences, University of Missouri, Columbia MO 65211
- Dalton Cardiovascular Research Center, University of Missouri, Columbia MO 65211
| | - Luis Polo-Parada
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia MO 65211
- Dalton Cardiovascular Research Center, University of Missouri, Columbia MO 65211
| | - Heather A. Dantzler
- Department of Biomedical Sciences, University of Missouri, Columbia MO 65211
- Dalton Cardiovascular Research Center, University of Missouri, Columbia MO 65211
| | - David D. Kline
- Department of Biomedical Sciences, University of Missouri, Columbia MO 65211
- Dalton Cardiovascular Research Center, University of Missouri, Columbia MO 65211
| |
Collapse
|
37
|
AMPK breathing and oxygen supply. Respir Physiol Neurobiol 2019; 265:112-120. [DOI: 10.1016/j.resp.2018.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/06/2018] [Accepted: 08/31/2018] [Indexed: 01/28/2023]
|
38
|
Lima-Silveira L, Accorsi-Mendonça D, Bonagamba LGH, Almado CEL, da Silva MP, Nedoboy PE, Pilowsky PM, Machado BH. Enhancement of excitatory transmission in NTS neurons projecting to ventral medulla of rats exposed to sustained hypoxia is blunted by minocycline. J Physiol 2019; 597:2903-2923. [PMID: 30993693 DOI: 10.1113/jp277532] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 04/16/2019] [Indexed: 01/13/2023] Open
Abstract
KEY POINTS Rats subjected to sustained hypoxia (SH) present increases in arterial pressure (AP) and in glutamatergic transmission in the nucleus tractus solitarius (NTS) neurons sending projections to ventrolateral medulla (VLM). Treatment with minocycline, a microglial inhibitor, attenuated the increase in AP in response to SH. The increase in the amplitude of glutamatergic postsynaptic currents in the NTS-VLM neurons, induced by postsynaptic mechanisms, was blunted by minocycline treatment. The number of microglial cells was increased in the NTS of vehicle-treated SH rats but not in the NTS of minocycline-treated rats. The data show that microglial recruitment/proliferation induced by SH is associated with the enhancement of excitatory neurotransmission in NTS-VLM neurons, which may contribute to the observed increase in AP. ABSTRACT Short-term sustained hypoxia (SH) produces significant autonomic and respiratory adjustments and triggers activation of microglia, the resident immune cells in the brain. SH also enhances glutamatergic neurotransmission in the NTS. Here we evaluated the role of microglial activation induced by SH on the cardiovascular changes and mainly on glutamatergic neurotransmission in NTS neurons sending projections to the ventrolateral medulla (NTS-VLM), using a microglia inhibitor (minocycline). Direct measurement of arterial pressure (AP) in freely moving rats showed that SH (24 h, fraction of inspired oxygen ( F I , O 2 ) 0.1) in vehicle and minocycline (30 mg/kg i.p. for 3 days)-treated groups produced a significant increase in AP in relation to control groups under normoxic conditions, but this increase was significantly lower in minocycline-treated rats. Whole-cell patch-clamp recordings revealed that the active properties of the membrane were comparable among the groups. Nevertheless, the amplitudes of glutamatergic postsynaptic currents, evoked by tractus solitarius stimulation, were increased in NTS-VLM neurons of SH rats. Changes in asynchronous glutamatergic currents indicated that the observed increase in amplitude was due to postsynaptic mechanisms. These changes were blunted in the SH group previously treated with minocycline. Using immunofluorescence, we found that the number of microglial cells was increased in the NTS of vehicle-treated SH rats but not in the NTS neurons of minocycline-treated rats. Our data support the concept that microglial activation induced by SH is associated with the enhancement of excitatory neurotransmission in NTS-VLM neurons, which may contribute to the increase in AP observed in this experimental model.
Collapse
Affiliation(s)
- Ludmila Lima-Silveira
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, 14049-900, SP, Brazil
| | - Daniela Accorsi-Mendonça
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, 14049-900, SP, Brazil
| | - Leni G H Bonagamba
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, 14049-900, SP, Brazil
| | - Carlos Eduardo L Almado
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, 14049-900, SP, Brazil
| | - Melina P da Silva
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, 14049-900, SP, Brazil
| | - Polina E Nedoboy
- The Heart Research Institute, Sydney, New South Wales, 2042, Australia
| | - Paul M Pilowsky
- The Heart Research Institute, Sydney, New South Wales, 2042, Australia
| | - Benedito H Machado
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, 14049-900, SP, Brazil
| |
Collapse
|
39
|
Yamagishi A, Okada M, Masuda M, Sato N. Oxytocin administration modulates rats' helping behavior depending on social context. Neurosci Res 2019; 153:56-61. [PMID: 30953682 DOI: 10.1016/j.neures.2019.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/19/2019] [Accepted: 04/02/2019] [Indexed: 02/08/2023]
Abstract
The affiliative effect of oxytocin on behavior toward other individuals can be modulated by positive and negative aspects of those individuals. However, the context-dependent effect of oxytocin on helping behavior is still unclear. In this study, we examined the effect of oxytocin administration on helping behavior in rats. The rats learned to open a door to help a cagemate soaked with water. The rats were divided into Pair and Solo groups. The rats in the Pair group were housed with their cagemates and those in the Solo group were housed individually. The rats in both groups received oxytocin (1.0 mg/kg) or saline injections intraperitoneally for 5 consecutive days before starting the experimental sessions. In the rats injected with oxytocin, the Solo group showed helping behavior faster than those in the Pair group. The results suggest that the effects of oxytocin administration on helping behavior are dependent on the social context.
Collapse
Affiliation(s)
- Atsuhito Yamagishi
- Department of Psychological Sciences, Kwansei Gakuin University, 1-1-155, Uegahara, Nishinomiya, Hyogo 662-8501, Japan
| | - Maya Okada
- Department of Psychological Sciences, Kwansei Gakuin University, 1-1-155, Uegahara, Nishinomiya, Hyogo 662-8501, Japan
| | - Masatoshi Masuda
- Department of Psychological Sciences, Kwansei Gakuin University, 1-1-155, Uegahara, Nishinomiya, Hyogo 662-8501, Japan
| | - Nobuya Sato
- Department of Psychological Sciences, Kwansei Gakuin University, 1-1-155, Uegahara, Nishinomiya, Hyogo 662-8501, Japan.
| |
Collapse
|
40
|
Maruyama NO, Mitchell NC, Truong TT, Toney GM. Activation of the hypothalamic paraventricular nucleus by acute intermittent hypoxia: Implications for sympathetic long-term facilitation neuroplasticity. Exp Neurol 2018; 314:1-8. [PMID: 30605624 DOI: 10.1016/j.expneurol.2018.12.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/03/2018] [Accepted: 12/30/2018] [Indexed: 02/07/2023]
Abstract
Exposure to acute intermittent hypoxia (AIH) induces a progressive increase of sympathetic nerve activity (SNA) that reflects a form of neuroplasticity known as sympathetic long-term facilitation (sLTF). Our recent findings indicate that activity of neurons in the hypothalamic paraventricular nucleus (PVN) contributes to AIH-induced sLTF, but neither the intra-PVN distribution nor the neurochemical identity of AIH responsive neurons has been determined. Here, awake rats were exposed to 10 cycles of AIH and c-Fos immunohistochemistry was performed to identify transcriptionally activated neurons in rostral, middle and caudal planes of the PVN. Effects of graded intensities of AIH were investigated in separate groups of rats (n = 6/group) in which inspired oxygen (O2) was reduced every 6 min from 21% to nadirs of 10%, 8% or 6%. All intensities of AIH failed to increase c-Fos counts in the caudally located lateral parvocellular region of the PVN. c-Fos counts increased in the dorsal parvocellular and central magnocellular regions, but significance was achieved only with AIH to 6% O2 (P < 0.002). By contrast, graded intensities of AIH induced graded c-Fos activation in the stress-related medial parvocellular (MP) region. Focusing on AIH exposure to 8% O2, experiments next investigated the stress-regulatory neuropeptide content of AIH-activated MP neurons. Tissue sections immunostained for corticotropin-releasing hormone (CRH) or arginine vasopressin (AVP) revealed a significantly greater number of neurons stained for CRH than AVP (P < 0.0001), though AIH induced expression of c-Fos in a similar fraction (~14%) of each neurochemical class. Amongst AIH-activated MP neurons, ~30% stained for CRH while only ~2% stained for AVP. Most AIH-activated CRH neurons (~82%) were distributed in the rostral one-half of the PVN. Results indicate that AIH recruits CRH, but not AVP, neurons in rostral to middle levels of the MP region of PVN, and raise the possibility that these CRH neurons may be a substrate for AIH-induced sLTF neuroplasticity.
Collapse
Affiliation(s)
- Nadia Oliveira Maruyama
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Nathan C Mitchell
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Tamara T Truong
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Glenn M Toney
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
41
|
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
|
42
|
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
|
43
|
Litvin DG, Dick TE, Smith CB, Jacono FJ. Lung-injury depresses glutamatergic synaptic transmission in the nucleus tractus solitarii via discrete age-dependent mechanisms in neonatal rats. Brain Behav Immun 2018; 70:398-422. [PMID: 29601943 PMCID: PMC6075724 DOI: 10.1016/j.bbi.2018.03.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 03/20/2018] [Accepted: 03/26/2018] [Indexed: 12/26/2022] Open
Abstract
Transition periods (TPs) are brief stages in CNS development where neural circuits can exhibit heightened vulnerability to pathologic conditions such as injury or infection. This susceptibility is due in part to specialized mechanisms of synaptic plasticity, which may become activated by inflammatory mediators released under pathologic conditions. Thus, we hypothesized that the immune response to lung injury (LI) mediated synaptic changes through plasticity-like mechanisms that depended on whether LI occurred just before or after a TP. We studied the impact of LI on brainstem 2nd-order viscerosensory neurons located in the nucleus tractus solitarii (nTS) during a TP for respiratory control spanning (postnatal day (P) 11-15). We injured the lungs of Sprague-Dawley rats by intratracheal instillation of Bleomycin (or saline) just before (P9-11) or after (P17-19) the TP. A week later, we prepared horizontal slices of the medulla and recorded spontaneous and evoked excitatory postsynaptic currents (sEPSCs/eEPSCs) in vitro from neurons in the nTS that received monosynaptic glutamatergic input from the tractus solitarii (TS). In rats injured before the TP (pre-TP), neurons exhibited blunted sEPSCs and TS-eEPSCs compared to controls. The decreased TS-eEPSCs were mediated by differences in postsynaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic-acid receptors (AMPAR). Specifically, compared to controls, LI rats had more Ca2+-impermeable AMPARs (CI-AMPARs) as indicated by: 1) the absence of current-rectification, 2) decreased sensitivity to polyamine, 1-Naphthyl-acetyl-spermine-trihydrochloride (NASPM) and 3) augmented immunoreactive staining for the CI-AMPAR GluA2. Thus, pre-TP-LI acts postsynaptically to blunt glutamatergic transmission. The neuroimmune response to pre-TP-LI included microglia hyper-ramification throughout the nTS. Daily intraperitoneal administration of minocycline, an inhibitor of microglial/macrophage function prevented hyper-ramification and abolished the pre-TP-LI evoked synaptic changes. In contrast, rat-pups injured after the TP (post-TP) exhibited microglia hypo-ramification in the nTS and had increased sEPSC amplitudes/frequencies, and decreased TS-eEPSC amplitudes compared to controls. These synaptic changes were not associated with changes in CI-AMPARs, and instead involved greater TS-evoked use-dependent depression (reduced paired pulse ratio), which is a hallmark of presynaptic plasticity. Thus we conclude that LI regulates the efficacy of TS → nTS synapses through discrete plasticity-like mechanisms that are immune-mediated and depend on whether the injury occurs before or after the TP for respiratory control.
Collapse
Affiliation(s)
- David G Litvin
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Division of Pulmonary, Critical Care and Sleep Medicine, Louis Stokes VA Medical Center, Cleveland, OH 44106, United States
| | - Thomas E Dick
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States
| | - Corey B Smith
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States
| | - Frank J Jacono
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Division of Pulmonary, Critical Care and Sleep Medicine, Louis Stokes VA Medical Center, Cleveland, OH 44106, United States.
| |
Collapse
|
44
|
Tanida M, Zhang T, Sun L, Song J, Yang W, Kuda Y, Kurata Y, Shibamoto T. Anaphylactic hypotension causes renal and adrenal sympathoexcitaion and induces c-fos in the hypothalamus and medulla oblongata. Exp Physiol 2018. [PMID: 29524326 DOI: 10.1113/ep086809] [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] [Indexed: 01/12/2023]
Abstract
NEW FINDINGS What is the central question of this study? Whether anaphylaxis affects sympathetic outflows to the brown adipose tissue (BAT) and adrenal gland and whether anaphylaxis affects some brain areas in association with sympathetic regulation. What is the main finding and its importance? Sympathoexcitatory responses to anaphylaxis occurred regionally in the kidney and adrenal gland, but not in the thermogenesis-related BAT. Further, anaphylactic hypotension also caused increase in c-fos immunoreactivity in the hypothalamic and medullary areas. Moreover, catecholaminergic neurons of the brainstem cause adrenal sympathoexcitation in a baroreceptor-independent manner. ABSTRACT We previously reported that sympathetic nerve activity (SNA) to the kidney and the hindlimb increases during anaphylactic hypotension in anaesthetized rats. Based on this evidence, we examined effects of anaphylactic hypotension on SNA to the brown adipose tissue (BAT), and the adrenal gland and kidney in anaesthetized rats. We demonstrated that adrenal and renal SNA, but not BAT-SNA, were stimulated. In addition, the effects of anaphylaxis on neural activities of the hypothalamic and medullary nuclei, which are candidates for relaying efferent SNA to the peripheral organs, were investigated via immunohistochemical staining of c-fos. Anaphylaxis increased c-fos expression in the neurons of the paraventricular nucleus (PVN) of the hypothalamus and in those of the nucleus tractus solitarii (NTS) and rostral ventrolateral medulla (RVLM) of the medulla oblongata; c-fos was expressed in γ-aminobutyric acid (GABA)-ergic neurons of the NTS and in the catecholaminergic neurons of the RVLM. In addition, c-fos expression in the rostral NTS and mid NTS during anaphylaxis was reduced by sinoaortic baroreceptor denervation; however, increased c-fos expression in the caudal NTS and RVLM or adrenal sympathoexcitation were not affected by sinoaortic baroreceptor denervation. These results indicated that anaphylactic hypotension activates the hypothalamic PVN and the medullary NTS and RVLM independently of the baroreflex pathway. Further, it stimulated efferent SNA to the adrenal gland and kidney to restore blood pressure.
Collapse
Affiliation(s)
- Mamoru Tanida
- Department of Physiology II, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Tao Zhang
- Department of Physiology II, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan.,Department of Colorectal and Hernia Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Lingling Sun
- Department of Physiology II, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan.,Department of Hematology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Jie Song
- Department of Physiology II, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan.,Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Wei Yang
- Department of Physiology II, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan.,Department of Infectious Disease, The Sheng Jing Hospital of China Medical University, Shenyang, 110009, China
| | - Yuichi Kuda
- Department of Physiology II, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Yasutaka Kurata
- Department of Physiology II, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Toshishige Shibamoto
- Department of Physiology II, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| |
Collapse
|
45
|
Faulk KE, Nedungadi TP, Cunningham JT. Angiotensin converting enzyme 1 in the median preoptic nucleus contributes to chronic intermittent hypoxia hypertension. Physiol Rep 2018; 5:e13277. [PMID: 28536140 PMCID: PMC5449561 DOI: 10.14814/phy2.13277] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 04/13/2017] [Accepted: 04/17/2017] [Indexed: 02/07/2023] Open
Abstract
Obstructive sleep apnea is associated with hypertension and cardiovascular disease. Chronic intermittent hypoxia is used to model the arterial hypoxemia seen in sleep apnea patients and is associated with increased sympathetic nerve activity and a sustained diurnal increase in blood pressure. The renin angiotensin system has been associated with hypertension seen in chronic intermittent hypoxia. Angiotensin converting enzyme 1, which cleaves angiotensin I to the active counterpart angiotensin II, is present within the central nervous system and has been shown to be regulated by AP‐1 transcription factors, such as ΔFosB. Our previous study suggested that this transcriptional regulation in the median preoptic nucleus contributes to the sustained blood pressure seen following chronic intermittent hypoxia. Viral mediated delivery of a short hairpin RNA against angiotensin converting enzyme 1 in the median preoptic nucleus was used along with radio‐telemetry measurements of blood pressure to test this hypothesis. FosB immunohistochemistry was utilized in order to assess the effects of angiotensin converting enzyme 1 knockdown on the activity of nuclei downstream from median preoptic nucleus. Angiotensin converting enzyme 1 knockdown within median preoptic nucleus significantly attenuated the sustained hypertension seen in chronic intermittent hypoxia. Angiotensin converting enzyme 1 seems to be partly responsible for regulating downstream regions involved in sympathetic and blood pressure control, such as the paraventricular nucleus and the rostral ventrolateral medulla. The data suggest that angiotensin converting enzyme 1 within median preoptic nucleus plays a critical role in the sustained hypertension seen in chronic intermittent hypoxia.
Collapse
Affiliation(s)
- Katelynn E Faulk
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Centre at Fort Worth, Fort Worth, Texas
| | - T Prashant Nedungadi
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Centre at Fort Worth, Fort Worth, Texas
| | - J Thomas Cunningham
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Centre at Fort Worth, Fort Worth, Texas
| |
Collapse
|
46
|
Onishi M, Yamanaka K, Miyamoto Y, Waki H, Gouraud S. Trpv4 involvement in the sex differences in blood pressure regulation in spontaneously hypertensive rats. Physiol Genomics 2018; 50:272-286. [PMID: 29373075 DOI: 10.1152/physiolgenomics.00096.2017] [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] [Indexed: 01/24/2023] Open
Abstract
Arterial pressure (AP) is lower in premenopausal women than in men of a similar age. Premenopausal women exhibit a lower sympathetic activity and a greater baroreceptor reflex; however, mechanisms controlling sex differences in blood pressure regulation are not well understood. We hypothesized that different neuronal functions in the cardiovascular centers of the brains of men and women may contribute to the sex difference in cardiovascular homeostasis. Our previous studies on male spontaneously hypertensive rats (SHRs) and their normotensive counterparts, Wistar Kyoto (WKY) rats, revealed that the gene-expression profile of the nucleus tractus solitarius (NTS), a region of the medulla oblongata that is pivotal for regulating the set point of AP, is strongly associated with AP. Thus, we hypothesized that gene-expression profiles in the rat NTS are related to sex differences in AP regulation. Because female SHRs clearly exhibit lower AP than their male counterparts of a similar age, we investigated whether SHR NTS exhibits sex differences in gene expression by using microarray and RT-qPCR experiments. The transcript for transient receptor potential cation channel subfamily V member 4 ( Trpv4) was found to be upregulated in SHR NTS in females compared with that in males. The channel was expressed in neurons and glial cells within NTS. The TRPV4 agonist 4-alpha-phorbol-12,13-didecanoate (4α-PDD) decreased blood pressure when injected into NTS of rats. These findings suggest that altered TRPV4 expression might be involved in the sex differences in blood pressure regulation.
Collapse
Affiliation(s)
- Makiko Onishi
- Graduate School of Humanities and Sciences, Ochanomizu University, Otsuka, Bunkyo-ku, Tokyo , Japan.,Institute for Human Life Innovation, Ochanomizu University, Otsuka, Bunkyo-ku, Tokyo , Japan
| | - Ko Yamanaka
- Department of Physiology, Graduate School of Health and Sports Science, Juntendo University, Inzai-city, Chiba , Japan
| | - Yasunori Miyamoto
- Graduate School of Humanities and Sciences, Ochanomizu University, Otsuka, Bunkyo-ku, Tokyo , Japan.,Program for Leading Graduate Schools, Ochanomizu University, Otsuka, Bunkyo-ku, Tokyo , Japan.,Institute for Human Life Innovation, Ochanomizu University, Otsuka, Bunkyo-ku, Tokyo , Japan
| | - Hidefumi Waki
- Department of Physiology, Graduate School of Health and Sports Science, Juntendo University, Inzai-city, Chiba , Japan
| | - Sabine Gouraud
- Program for Leading Graduate Schools, Ochanomizu University, Otsuka, Bunkyo-ku, Tokyo , Japan.,Department of Biology, Ochanomizu University, Otsuka, Bunkyo-ku, Tokyo , Japan
| |
Collapse
|
47
|
Summanen M, Bäck S, Voipio J, Kaila K. Surge of Peripheral Arginine Vasopressin in a Rat Model of Birth Asphyxia. Front Cell Neurosci 2018; 12:2. [PMID: 29403357 PMCID: PMC5780440 DOI: 10.3389/fncel.2018.00002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/03/2018] [Indexed: 12/29/2022] Open
Abstract
Mammalian birth is accompanied by a period of obligatory asphyxia, which consists of hypoxia (drop in blood O2 levels) and hypercapnia (elevation of blood CO2 levels). Prolonged, complicated birth can extend the asphyxic period, leading to a pathophysiological situation, and in humans, to the diagnosis of clinical birth asphyxia, the main cause of hypoxic-ischemic encephalopathy (HIE). The neuroendocrine component of birth asphyxia, in particular the increase in circulating levels of arginine vasopressin (AVP), has been extensively studied in humans. Here we show for the first time that normal rat birth is also accompanied by an AVP surge, and that the fetal AVP surge is further enhanced in a model of birth asphyxia, based on exposing 6-day old rat pups to a gas mixture containing 4% O2 and 20% CO2 for 45 min. Instead of AVP, which is highly unstable with a short plasma half-life, we measured the levels of copeptin, the C-terminal part of prepro-AVP that is biochemically much more stable. In our animal model, the bulk of AVP/copeptin release occurred at the beginning of asphyxia (mean 7.8 nM after 15 min of asphyxia), but some release was still ongoing even 90 min after the end of the 45 min experimental asphyxia (mean 1.2 nM). Notably, the highest copeptin levels were measured after hypoxia alone (mean 14.1 nM at 45 min), whereas copeptin levels were low during hypercapnia alone (mean 2.7 nM at 45 min), indicating that the hypoxia component of asphyxia is responsible for the increase in AVP/copeptin release. Alternating the O2 level between 5 and 9% (CO2 at 20%) with 5 min intervals to mimic intermittent asphyxia during prolonged labor resulted in a slower but quantitatively similar rise in copeptin (peak of 8.3 nM at 30 min). Finally, we demonstrate that our rat model satisfies the standard acid-base criteria for birth asphyxia diagnosis, namely a drop in blood pH below 7.0 and the formation of a negative base excess exceeding -11.2 mmol/l. The mechanistic insights from our work validate the use of the present rodent model in preclinical work on birth asphyxia.
Collapse
Affiliation(s)
- Milla Summanen
- Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Susanne Bäck
- Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Juha Voipio
- Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Kai Kaila
- Department of Biosciences, University of Helsinki, Helsinki, Finland.,Neuroscience Center and HiLife, University of Helsinki, Helsinki, Finland
| |
Collapse
|
48
|
Blackburn MB, Andrade MA, Toney GM. Hypothalamic PVN contributes to acute intermittent hypoxia-induced sympathetic but not phrenic long-term facilitation. J Appl Physiol (1985) 2017; 124:1233-1243. [PMID: 29357503 DOI: 10.1152/japplphysiol.00743.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Blackburn MB, Andrade MA, Toney GM. Hypothalamic PVN contributes to acute intermittent hypoxia-induced sympathetic but not phrenic long-term facilitation. J Appl Physiol 124: 1233-1243, 2018. First published December 19, 2017; doi: 10.1152/japplphysiol.00743.2017 .- Acute intermittent hypoxia (AIH) repetitively activates the arterial chemoreflex and triggers a progressive increase of sympathetic nerve activity (SNA) and phrenic nerve activity (PNA) referred to as sympathetic and phrenic long-term facilitation (S-LTF and P-LTF), respectively. Neurons of the hypothalamic paraventricular nucleus (PVN) participate in the arterial chemoreflex, but their contribution to AIH-induced LTF is unknown. To determine this, anesthetized rats were vagotomized and exposed to 10 cycles of AIH, each consisting of ventilation for 3 min with 100% O2 followed by 3 min with 15% O2. Before AIH, rats received bilateral PVN injections of artificial cerebrospinal fluid (aCSF; vehicle) or the GABA-A receptor agonist muscimol (100 pmol in 50 nl) to inhibit neuronal activity. Thirty minutes after completing the AIH protocol, during which rats were continuously ventilated with 100% O2, S-LTF and P-LTF were quantified from recordings of integrated splanchnic SNA and PNA, respectively. PVN muscimol attenuated increases of SNA during hypoxic episodes occurring in later cycles (6-10) of AIH ( P < 0.03) and attenuated post-AIH S-LTF ( P < 0.001). Muscimol, however, did not consistently affect peak PNA responses during hypoxic episodes and did not alter AIH-induced P-LTF. These findings indicate that PVN neuronal activity contributes to sympathetic responses during AIH and to subsequent generation of S-LTF. NEW & NOTEWORTHY Neural circuits mediating acute intermittent hypoxia (AIH)-induced sympathetic and phrenic long-term facilitation (LTF) have not been fully elucidated. We found that paraventricular nucleus (PVN) inhibition attenuated sympathetic activation during episodes of AIH and reduced post-AIH sympathetic LTF. Neither phrenic burst patterning nor the magnitude of AIH-induced phrenic LTF was affected. Findings indicate that PVN neurons contribute to AIH-induced sympathetic LTF. Defining mechanisms of sympathetic LTF could improve strategies to reduce sympathetic activity in cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Megan B Blackburn
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| | - Mary Ann Andrade
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| | - Glenn M Toney
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio , San Antonio, Texas.,Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| |
Collapse
|
49
|
Carter DA, Choong YT, Connelly AA, Bassi JK, Hunter NO, Thongsepee N, Llewellyn-Smith IJ, Fong AY, McDougall SJ, Allen AM. Functional and neurochemical characterization of angiotensin type 1A receptor-expressing neurons in the nucleus of the solitary tract of the mouse. Am J Physiol Regul Integr Comp Physiol 2017; 313:R438-R449. [PMID: 28701322 DOI: 10.1152/ajpregu.00168.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/10/2017] [Accepted: 07/10/2017] [Indexed: 11/22/2022]
Abstract
Angiotensin II acts via two main receptors within the central nervous system, with the type 1A receptor (AT1AR) most widely expressed in adult neurons. Activation of the AT1R in the nucleus of the solitary tract (NTS), the principal nucleus receiving central synapses of viscerosensory afferents, modulates cardiovascular reflexes. Expression of the AT1R occurs in high density within the NTS of most mammals, including humans, but the fundamental electrophysiological and neurochemical characteristics of the AT1AR-expressing NTS neurons are not known. To address this, we have used a transgenic mouse, in which the AT1AR promoter drives expression of green fluorescent protein (GFP). Approximately one-third of AT1AR-expressing neurons express the catecholamine-synthetic enzyme tyrosine hydroxylase (TH), and a subpopulation of these stained for the transcription factor paired-like homeobox 2b (Phox2b). A third group, comprising approximately two-thirds of the AT1AR-expressing NTS neurons, showed Phox2b immunoreactivity alone. A fourth group in the ventral subnucleus expressed neither TH nor Phox2b. In whole cell recordings from slices in vitro, AT1AR-GFP neurons exhibited voltage-activated potassium currents, including the transient outward current and the M-type potassium current. In two different mouse strains, both AT1AR-GFP neurons and TH-GFP neurons showed similar AT1AR-mediated depolarizing responses to superfusion with angiotensin II. These data provide a comprehensive description of AT1AR-expressing neurons in the NTS and increase our understanding of the complex actions of this neuropeptide in the modulation of viscerosensory processing.
Collapse
Affiliation(s)
- D A Carter
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Y-T Choong
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - A A Connelly
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - J K Bassi
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - N O Hunter
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - N Thongsepee
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - I J Llewellyn-Smith
- Cardiovascular Medicine and Human Physiology, School of Medicine, Flinders University, Bedford Park, South Australia, Australia; and
| | - A Y Fong
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - S J McDougall
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - A M Allen
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia; .,Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
50
|
Matott MP, Kline DD, Hasser EM. Glial EAAT2 regulation of extracellular nTS glutamate critically controls neuronal activity and cardiorespiratory reflexes. J Physiol 2017; 595:6045-6063. [PMID: 28677303 PMCID: PMC5577520 DOI: 10.1113/jp274620] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 06/27/2017] [Indexed: 01/24/2023] Open
Abstract
KEY POINTS Excitatory amino acid transporter 2 (EAAT2) is present on astrocytes in the nucleus tractus solitarii (nTS), an important nucleus in cardiorespiratory control. Its specific role in influencing nTS neuronal activity and thereby basal and reflex cardiorespiratory function is unknown. The specific role of nTS EAAT2 was determined via whole animal and brainstem slice patch clamp experiments. Astrocytic EAAT2 buffers basal glutamate activation of AMPA-type glutamate receptors and therefore decreases baseline excitability of nTS neurons. EAAT2 modulates cardiorespiratory control and tempers excitatory cardiorespiratory responses to activation of the peripheral chemoreflex. This study supports the concept that nTS astrocyte transporters influence sympathetic nervous system activity and cardiorespiratory reflex function in health and disease. ABSTRACT Glutamatergic signalling is critical in the nucleus tractus solitarii (nTS) for cardiorespiratory homeostasis and initiation of sensory reflexes, including the chemoreflex activated during hypoxia. Maintenance of nTS glutamate concentration occurs in part through astrocytic excitatory amino acid transporters (EAATs). We previously established the importance of EAATs in the nTS by demonstrating their inhibition produced neuronal excitation to alter basal cardiorespiratory function. Since EAAT2 is the most expressed EAAT in the nTS, this study specifically determined EAAT2's role in nTS astrocytes, its influence on neuronal and synaptic properties, and ultimately on basal and reflex cardiorespiratory function. The EAAT2-specific antagonist dihydrokainate (DHK) was microinjected into the anaesthetized rat nTS or applied to rat nTS slices. DHK produced depressor, bradycardic and sympathoinhibitory responses and reduced neural respiration in the intact rat, mimicking responses to glutamate excitation. DHK also enhanced responses to glutamate microinjection. DHK elevated extracellular nTS glutamate concentration, depolarized neurons and enhanced spontaneous EPSCs. EAAT2 block also augmented action potential discharge in chemosensitive nTS neurons. Glial recordings confirmed EAAT2 is functional on nTS astrocytes. Neuronal excitation and cardiorespiratory effects following EAAT2 inhibition were due to activation of putative extrasynaptic AMPA receptors as their antagonism blocked DHK responses in the intact rat nTS and the slice. The DHK-induced elevation of extracellular glutamate and neuronal excitation augmented chemoreflex-mediated pressor, sympathoexcitatory and minute neural ventilation responses in the rat. These data shed new light on the important role astrocytic EAAT2 plays on buffering nTS excitation and overall cardiorespiratory function.
Collapse
Affiliation(s)
- Michael P. Matott
- Department of Biomedical Sciences, Dalton Cardiovascular Research CenterUniversity of Missouri134 Research Park DriveColumbiaMO65211USA
| | - David D. Kline
- Department of Biomedical Sciences, Dalton Cardiovascular Research CenterUniversity of Missouri134 Research Park DriveColumbiaMO65211USA
| | - Eileen M. Hasser
- Department of Biomedical SciencesDepartment of Medical Pharmacology and Physiology, Dalton Cardiovascular Research CenterUniversity of Missouri134 Research Park DriveColumbiaMO65211USA
| |
Collapse
|