1
|
Schwarz KG, Pereyra KV, Díaz-Jara E, Vicencio SC, Del Rio R. Brainstem C1 neurons mediate heart failure decompensation and mortality during acute salt loading. Cardiovasc Res 2025; 121:241-253. [PMID: 39775485 PMCID: PMC12012444 DOI: 10.1093/cvr/cvae261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/12/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025] Open
Abstract
AIMS Heart failure (HF) is an emerging epidemic worldwide. Despite advances in treatment, the morbidity and mortality rate of HF remain high, and the global prevalence continues to rise. Common clinical features of HF include cardiac sympathoexcitation, disordered breathing, and kidney dysfunction; kidney dysfunction strongly contributes to sodium retention and fluid overload, leading to poor outcomes of HF patients. We have previously shown that brainstem pre-sympathetic neurons (C1) from the rostral ventrolateral medulla (RVLM) play a key role in sympathetic regulation in experimental models of HF. However, the role of RVLM-C1 neurons during salt-loading in the context of HF is unknown. This study tests whether RVLM C1 neurons drive cardiorespiratory decompensation and ultimately lead to sudden death in HF rats. METHODS AND RESULTS Adult male Sprague-Dawley rats underwent arteriovenous shunt to induce HF with preserved ejection fraction (HFpEF). Two weeks after HFpEF induction, bilateral selective ablation of RVLM C1 neurons was performed using anti-dopamine β-hydroxylase-saporin toxin. Animals were then fed a high Na+ diet (3% Na+ in food and 2% Na+ in water) for 3 weeks to induce compensated-to-decompensated HF state transition. Echocardiography, cardiac autonomic function, breathing function, and survival were assessed during the progression of HF. Salt loading resulted in marked decompensation in HF rats, as evidenced by a significant decrease in survival rates (survival: 10% vs. 100% HFpEF + Na+ vs. HFpEF). Furthermore, HFpEF + Na+ animals showed a further increase in cardiac sympathetic drive and more severe disordered breathing, including higher hypoxia-related epochs (i.e. apnoeas/hypopnoeas), compared with HF. Ablation of RVLM C1 neurons partly reduced the excessive cardiac sympathoexcitation during salt loading in HF, improved the exaggerated disordered breathing in HFpEF+ Na+ rats, and reduced decompensation-linked mortality. We found that hypoxia, but not high sodium, was the major contributor to impaired calcium handling in isolated adult cardiomyocytes. CONCLUSION Our results strongly suggest that RVLM C1 neurons contribute to acute HF decompensation during salt loading by a mechanism encompassing further increases in sympathetic outflow and hypoxia-related breathing disorders. This mechanism may ultimately impact cardiac contractility through cardiomyocyte calcium mishandling, increasing morbidity and mortality.
Collapse
Affiliation(s)
- Karla G Schwarz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
| | - Katherin V Pereyra
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
| | - Esteban Díaz-Jara
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
| | - Sinay C Vicencio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Av. los Flamencos 01364, Punta Arenas 6210005, Chile
- Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd HLSIC-2091, Kansas City, KS 66160, USA
| |
Collapse
|
2
|
Jia X, Sun J, Zhuo Q, Zhao B, Liu Y. Effect of the NLRP3 inflammasome on increased hypoxic ventilation response after CIH exposure in mice. Respir Physiol Neurobiol 2024; 321:104204. [PMID: 38128772 DOI: 10.1016/j.resp.2023.104204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Chronic intermittent hypoxia (CIH) increases the hypoxic ventilation response (HVR). The downstream cytokine IL-1β of the NLRP3 inflammasome regulates respiration by acting on the carotid body (CB) and neurons in the respiratory center, but the effect of the NLRP3 inflammasome on HVR induced by CIH remains unclear. OBJECTIVE To investigate the effect of NLRP3 on the increased HVR and spontaneous apnea events and duration induced by CIH, the expression and localization of NLRP3 in the respiratory regulatory center of the rostral ventrolateral medulla (RVLM), and the effect of CIH on the activation of the NLRP3 inflammasome in the RVLM. METHODS Eighteen male, 7-week-old C57BL/6 N mice and eighteen male, 7-week-old C57BL/6 N NLRP3 knockout mice were randomly divided into CON-WT, CON-NLRP3-/-, CIH-WT and CIH-NLRP3-/- groups. Respiratory changes in mice were continuously detected using whole-body plethysmography. The expression and localization of the NLRP3 protein and the formation of apoptosis-associated speck-like protein containing CARD (ASC) specks were detected using immunofluorescence staining. RESULTS NLRP3 knockout reduced the increased HVR and the incidence and duration of spontaneous apnea events associated with CIH. The increase in HVR caused by CIH partially recovered after reoxygenation. After CIH, NLRP3 inflammasome activation in the RVLM, which is related to respiratory regulation after hypoxia, increased, which was consistent with the trend of the ventilation response. CONCLUSION The NLRP3 inflammasome may be involved in the increase in the HVR and the incidence and duration of spontaneous apnea induced by CIH. NLRP3 inhibitors may help reduce the increase in the HVR after CIH, which is important for ensuring sleep quality at night in patients with obstructive sleep apnea.
Collapse
Affiliation(s)
- Xinyun Jia
- Department of Respiratory, The First Affiliated Hospital of Xinxiang Medical University, 88 Jiankang Road, Weihui 453100, Henan, China; Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, 88 Jiankang Road, Weihui 453100, Henan, China
| | - Jianxia Sun
- Department of Respiratory, The First Affiliated Hospital of Xinxiang Medical University, 88 Jiankang Road, Weihui 453100, Henan, China; Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, 88 Jiankang Road, Weihui 453100, Henan, China
| | - Qingya Zhuo
- Department of Respiratory, The First Affiliated Hospital of Xinxiang Medical University, 88 Jiankang Road, Weihui 453100, Henan, China; Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, 88 Jiankang Road, Weihui 453100, Henan, China
| | - Baosheng Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China
| | - Yuzhen Liu
- Department of Respiratory, The First Affiliated Hospital of Xinxiang Medical University, 88 Jiankang Road, Weihui 453100, Henan, China; Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, 88 Jiankang Road, Weihui 453100, Henan, China; Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China.
| |
Collapse
|
3
|
Schwarz KG, Vicencio SC, Inestrosa NC, Villaseca P, Del Rio R. Autonomic nervous system dysfunction throughout menopausal transition: A potential mechanism underpinning cardiovascular and cognitive alterations during female ageing. J Physiol 2024; 602:263-280. [PMID: 38064358 DOI: 10.1113/jp285126] [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: 08/02/2023] [Accepted: 11/24/2023] [Indexed: 01/16/2024] Open
Abstract
Cardiovascular diseases (CVD) and neurodegenerative disorders, such as Alzheimer's disease (AD), are highly prevalent conditions in middle-aged women that severely impair quality of life. Recent evidence suggests the existence of an intimate cross-talk between the heart and the brain, resulting from a complex network of neurohumoral circuits. From a pathophysiological perspective, the higher prevalence of AD in women may be explained, at least in part, by sex-related differences in the incidence/prevalence of CVD. Notably, the autonomic nervous system, the main heart-brain axis physiological orchestrator, has been suggested to play a role in the incidence of adverse cardiovascular events in middle-aged women because of decreases in oestrogen-related signalling during transition into menopause. Despite its overt relevance for public health, this hypothesis has not been thoroughly tested. Accordingly, in this review, we aim to provide up to date evidence supporting how changes in circulating oestrogen levels during transition to menopause may trigger autonomic dysfunction, thus promoting cardiovascular and cognitive decline in women. A main focus on the effects of oestrogen-mediated signalling at CNS structures related to autonomic regulation is provided, particularly on the role of oestrogens in sympathoexcitation. Improving the understanding of the contribution of the autonomic nervous system on the development, maintenance and/or progression of both cardiovascular and cognitive dysfunction during the transition to menopause should help improve the clinical management of elderly women, with the outcome being an improved life quality during the natural ageing process.
Collapse
Affiliation(s)
- Karla G Schwarz
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sinay C Vicencio
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Paulina Villaseca
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
- Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
4
|
Toledo C, Andrade DC, Diaz-Jara E, Ortolani D, Bernal-Santander I, Schwarz KG, Ortiz FC, Marcus NJ, Oliveira LM, Takakura AC, Moreira TS, Del Rio R. Cardiorespiratory alterations following intermittent photostimulation of RVLM C1 neurons: Implications for long-term blood pressure, breathing and sleep regulation in freely moving rats. Acta Physiol (Oxf) 2022; 236:e13864. [PMID: 35959519 DOI: 10.1111/apha.13864] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 01/29/2023]
Abstract
AIM Sympathoexcitation and sleep-disordered breathing are common contributors for disease progression. Catecholaminergic neurons from the rostral ventrolateral medulla (RVLM-C1) modulate sympathetic outflow and have anatomical projections to respiratory neurons; however, the contribution of highly selective activation of RVLM-C1 neurons on long-term autonomic and breathing (dys)regulation remains to be understood. METHODS To explore this relationship, a lentiviral vector carrying the light-sensitive cation channel channelrhodopsin-2 (LVV-PRSX8-ChR2-YFP) was unilaterally injected into the RVLM of healthy rats. On the contralateral side, LVV-PRSX8-ChR2-YFP was co-injected with a specific immunotoxin (DβH-SAP) targeted to eliminate C1 neurons. RESULTS Intermittent photostimulation of RVLM-C1 in vivo, in unrestrained freely moving rats, elicited long-term facilitation of the sympathetic drive, a rise in blood pressure and sympatho-respiratory coupling. In addition, photoactivation of RVLM-C1 induced long-lasting ventilatory instability, characterized by oscillations in tidal volume and increased breathing variability, but only during non-rapid eye movement sleep. These effects were not observed when photostimulation of the RVLM was performed in the presence of DβH-SAP toxin. CONCLUSIONS The finding that intermittent activation of RVLM-C1 neurons induces autonomic and breathing dysfunction suggest that episodic stimulation of RVLM-C1 may serve as a pathological substrate for the long-term development of cardiorespiratory disorders.
Collapse
Affiliation(s)
- Camilo Toledo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - David C Andrade
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Fisiología y Medicina de Altura, Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Esteban Diaz-Jara
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Domiziana Ortolani
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ignacio Bernal-Santander
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karla G Schwarz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernando C Ortiz
- Mechanisms of Myelin Formation and Repair Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad, Autónoma de Chile, Santiago, Chile
| | - Noah J Marcus
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, Iowa, USA
| | - Luiz M Oliveira
- Department of Pharmacology, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Ana C Takakura
- Department of Pharmacology, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
5
|
Toledo C, Díaz-Jara E, Diaz HS, Schwarz KG, Pereyra KV, Las Heras A, Rios-Gallardo A, Andrade DC, Moreira T, Takakura A, Marcus NJ, Del Rio R. Medullary astrocytes mediate irregular breathing patterns generation in chronic heart failure through purinergic P2X7 receptor signalling. EBioMedicine 2022; 80:104044. [PMID: 35533501 PMCID: PMC9097632 DOI: 10.1016/j.ebiom.2022.104044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 11/05/2022] Open
Abstract
Background Breathing disorders (BD) (apnoeas/hypopneas, periodic breathing) are highly prevalent in chronic heart failure (CHF) and are associated with altered central respiratory control. Ample evidence identifies the retrotrapezoid nucleus (RTN) as an important chemosensitivity region for ventilatory control and generation of BD in CHF, however little is known about the cellular mechanisms underlying the RTN/BD relationship. Within the RTN, astrocyte‐mediated purinergic signalling modulates respiration, but the potential contribution of RTN astrocytes to BD in CHF has not been explored. Methods Selective neuron and/or astrocyte-targeted interventions using either optogenetic and chemogenetic manipulations in the RTN of CHF rats were used to unveil the contribution of the RTN on the development/maintenance of BD, the role played by astrocytes in BD and the molecular mechanism underpinning these alterations. Findings We showed that episodic photo-stimulation of RTN neurons triggered BD in healthy rats, and that RTN neurons ablation in CHF animals eliminates BD. Also, we found a reduction in astrocytes activity and ATP bioavailability within the RTN of CHF rats, and that chemogenetic restoration of normal RTN astrocyte activity and ATP levels improved breathing regularity in CHF. Importantly, P"X/ P2X7 receptor (P2X7r) expression was reduced in RTN astrocytes from CHF rats and viral vector-mediated delivery of human P2X7 P2X7r into astrocytes increases ATP bioavailability and abolished BD. Interpretation Our results support that RTN astrocytes play a pivotal role on BD generation and maintenance in the setting CHF by a mechanism encompassing P2X7r signalling. Funding This study was funded by the National Research and Development Agency of Chile (ANID).
Collapse
|
6
|
Toledo C, Ortolani D, Ortiz FC, Marcus NJ, Del Rio R. Potential Role of the Retrotrapezoid Nucleus in Mediating Cardio-Respiratory Dysfunction in Heart Failure With Preserved Ejection Fraction. Front Physiol 2022; 13:863963. [PMID: 35492622 PMCID: PMC9039230 DOI: 10.3389/fphys.2022.863963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/29/2022] [Indexed: 01/15/2023] Open
Abstract
A strong association between chemoreflex hypersensitivity, disordered breathing, and elevated sympathetic activity has been shown in experimental and human heart failure (HF). The contribution of chemoreflex hypersensitivity in HF pathophysiology is incompletely understood. There is ample evidence that increased peripheral chemoreflex drive in HF with reduced ejection fraction (HFrEF; EF<40%) leads to pathophysiological changes in autonomic and cardio-respiratory control, but less is known about the neural mechanisms mediating cardio-respiratory disturbances in HF with preserved EF (HFpEF; EF>50%). Importantly, it has been shown that activation of the central chemoreflex worsens autonomic dysfunction in experimental HFpEF, an effect mediated in part by the activation of C1 catecholaminergic neurons neighboring the retrotrapezoid nucleus (RTN), an important region for central chemoreflex control of respiratory and autonomic function. Accordingly, the main purpose of this brief review is to discuss the possible role played by activation of central chemoreflex pathways on autonomic function and its potential role in precipitating disordered breathing in HFpEF. Improving understanding of the contribution of the central chemoreflex to the pathophysiology of HFpEF may help in development of novel interventions intended to improve cardio-respiratory outcomes in HFpEF.
Collapse
Affiliation(s)
- Camilo Toledo
- Laboratory Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Domiziana Ortolani
- Laboratory Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernando C. Ortiz
- Mechanisms of Myelin Formation and Repair Laboratory, Facultad de Ciencias de Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Noah J. Marcus
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA, United States
| | - Rodrigo Del Rio
- Laboratory Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
- Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Rodrigo Del Rio,
| |
Collapse
|
7
|
Schwarz KG, Pereyra KV, Toledo C, Andrade DC, Díaz HS, Díaz-Jara E, Ortolani D, Rios-Gallardo A, Arias P, Las Heras A, Vera I, Ortiz FC, Inestrosa NC, Vio CP, Del Rio R. Effects of enriched-potassium diet on cardiorespiratory outcomes in experimental non-ischemic chronic heart failure. Biol Res 2021; 54:43. [PMID: 34952651 PMCID: PMC8710008 DOI: 10.1186/s40659-021-00365-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022] Open
Abstract
Background Chronic heart failure (CHF) is a global health problem. Increased sympathetic outflow, cardiac arrhythmogenesis and irregular breathing patterns have all been associated with poor outcomes in CHF. Several studies showed that activation of the renin-angiotensin system (RAS) play a key role in CHF pathophysiology. Interestingly, potassium (K+) supplemented diets showed promising results in normalizing RAS axis and autonomic dysfunction in vascular diseases, lowering cardiovascular risk. Whether subtle increases in dietary K+ consumption may exert similar effects in CHF has not been previously tested. Accordingly, we aimed to evaluate the effects of dietary K+ supplementation on cardiorespiratory alterations in rats with CHF. Methods Adult male Sprague–Dawley rats underwent volume overload to induce non-ischemic CHF. Animals were randomly allocated to normal chow diet (CHF group) or supplemented K+ diet (CHF+K+ group) for 6 weeks. Cardiac arrhythmogenesis, sympathetic outflow, baroreflex sensitivity, breathing disorders, chemoreflex function, respiratory–cardiovascular coupling and cardiac function were evaluated. Results Compared to normal chow diet, K+ supplemented diet in CHF significantly reduced arrhythmia incidence (67.8 ± 15.1 vs. 31.0 ± 3.7 events/hour, CHF vs. CHF+K+), decreased cardiac sympathetic tone (ΔHR to propranolol: − 97.4 ± 9.4 vs. − 60.8 ± 8.3 bpm, CHF vs. CHF+K+), restored baroreflex function and attenuated irregular breathing patterns. Additionally, supplementation of the diet with K+ restores normal central respiratory chemoreflex drive and abrogates pathological cardio-respiratory coupling in CHF rats being the outcome an improved cardiac function. Conclusion Our findings support that dietary K+ supplementation in non-ischemic CHF alleviate cardiorespiratory dysfunction. Supplementary Information The online version contains supplementary material available at 10.1186/s40659-021-00365-z.
Collapse
Affiliation(s)
- Karla G Schwarz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katherin V Pereyra
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - David C Andrade
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Fisiología y Medicina de Altura, Departamento Biomédico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Hugo S Díaz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Esteban Díaz-Jara
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Domiziana Ortolani
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angélica Rios-Gallardo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Paulina Arias
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexandra Las Heras
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ignacio Vera
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernando C Ortiz
- Mechanisms of Myelin Formation and Repair Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.,Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos P Vio
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile. .,Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
8
|
Blanken CPS, Gottwald LM, Westenberg JJM, Peper ES, Coolen BF, Strijkers GJ, Nederveen AJ, Planken RN, van Ooij P. Whole-Heart 4D Flow MRI for Evaluation of Normal and Regurgitant Valvular Flow: A Quantitative Comparison Between Pseudo-Spiral Sampling and EPI Readout. J Magn Reson Imaging 2021; 55:1120-1130. [PMID: 34510612 PMCID: PMC9290924 DOI: 10.1002/jmri.27905] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 01/23/2023] Open
Abstract
Background Pseudo‐spiral Cartesian sampling with compressed sensing reconstruction has facilitated highly accelerated 4D flow magnetic resonance imaging (MRI) in various cardiovascular structures. However, unlike echo planar imaging (EPI)‐accelerated 4D flow MRI, it has not been validated in whole‐heart applications. Hypothesis Pseudo‐spiral 4D flow MRI (PROUD [PROspective Undersampling in multiple Dimensions]) is comparable to EPI in robustness of valvular flow measurements and remains comparable as the undersampling factor is increased and scan time reduced. Study Type Prospective. Population Twelve healthy subjects and eight patients with valvular regurgitation. Field Strength/Sequence 3.0 T; PROUD and EPI 4D flow sequences, 2D flow and balanced steady‐state free precession sequences. Assessment Valvular blood flow was quantified using valve tracking. PROUD‐ and EPI‐based measurements of aortic (AV) and pulmonary (PV) flow volumes and left and right ventricular stroke volumes were tested for agreement with 2D MRI‐based measurements. PROUD reconstructions with undersampling factors (R) of 9, 14, 28, and 56 were tested for intervalve consistency (per valve, compared to the other valves) and preservation of peak velocities and E/A ratios. Statistical Tests We used repeated measures ANOVA, Bland‐Altman, Wilcoxon signed rank, and intraclass correlation coefficients. P < 0.05 was considered statistically significant. Results PROUD and EPI intervalve consistencies were not significantly different both in healthy subjects (valve‐averaged mean difference [limits of agreement width]: 3.2 ± 0.8 [8.7 ± 1.1] mL/beat for PROUD, 5.5 ± 2.9 [13.7 ± 2.3] mL/beat for EPI, P = 0.07) and in patients with valvular regurgitation (2.3 ± 1.2 [15.3 ± 5.9] mL/beat for PROUD, 0.6 ± 0.6 [19.3 ± 2.9] mL/beat for EPI, P = 0.47). Agreement between EPI and PROUD was higher than between 4D flow (EPI or PROUD) and 2D MRI for forward flow, stroke volumes, and regurgitant volumes. Up to R = 28 in healthy subjects and R = 14 in patients with valvular regurgitation, PROUD intervalve consistency remained comparable to that of EPI. Peak velocities and E/A ratios were preserved up to R = 9. Conclusion PROUD is comparable to EPI in terms of intervalve consistency and may be used with higher undersampling factors to shorten scan times further. Level of Evidence 1 Technical Efficacy Stage 2
Collapse
Affiliation(s)
- Carmen P S Blanken
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Lukas M Gottwald
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | | | - Eva S Peper
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Bram F Coolen
- Department of Biomedical Engineering and Physics, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Gustav J Strijkers
- Department of Biomedical Engineering and Physics, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Aart J Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - R Nils Planken
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Pim van Ooij
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Díaz-Jara E, Díaz HS, Rios-Gallardo A, Ortolani D, Andrade DC, Toledo C, Pereyra KV, Schwarz K, Ramirez G, Ortiz FC, Andía ME, Del Rio R. Exercise training reduces brainstem oxidative stress and restores normal breathing function in heart failure. Free Radic Biol Med 2021; 172:470-481. [PMID: 34216779 DOI: 10.1016/j.freeradbiomed.2021.06.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 10/21/2022]
Abstract
Enhanced central chemoreflex drive and irregular breathing are both hallmarks in heart failure (HF) and closely related to disease progression. Central chemoreceptor neurons located within the retrotrapezoid nucleus (RTN) are known to play a role in breathing alterations in HF. It has been shown that exercise (EX) effectively reduced reactive oxygen species (ROS) in HF rats. However, the link between EX and ROS, particularly at the RTN, with breathing alterations in HF has not been previously addressed. Accordingly, we aimed to determine: i) ROS levels in the RTN in HF and its association with chemoreflex drive, ii) whether EX improves chemoreflex/breathing function by reducing ROS levels, and iii) determine molecular alterations associated with ROS generation within the RTN of HF rats and study EX effects on these pathways. Adult male Sprague-Dawley rats were allocated into 3 experimental groups: Sham (n = 5), volume overloaded HF (n = 6) and HF (n = 8) rats that underwent EX training for 6 weeks (60 min/day, 25 m/min, 10% inclination). At 8 weeks post-HF induction, breathing patterns and chemoreflex function were analyzed by unrestrained plethysmography. ROS levels and anti/pro-oxidant enzymes gene expression were analyzed in the RTN. Our results showed that HF rats have high ROS levels in the RTN which were closely linked to the enhanced central chemoreflex and breathing disorders. Also, HF rats displayed decreased expression of antioxidant genes in the RTN compared with control rats. EX training increases antioxidant defense in the RTN, reduces ROS formation and restores normal central chemoreflex drive and breathing regularity in HF rats. This study provides evidence for a role of ROS in central chemoreception in the setting of HF and support the use of EX to reduce ROS in the brainstem of HF animals and reveal its potential as an effective mean to normalize chemoreflex and breathing function in HF.
Collapse
Affiliation(s)
- Esteban Díaz-Jara
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Hugo S Díaz
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Angélica Rios-Gallardo
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, 621-0427, Punta Arenas, Chile.
| | - Domiziana Ortolani
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - David C Andrade
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile; Centro de Fisiología y Medicina de Altura, Facultad de Ciencias de la Salud, Universidad de Antofagasta, 1270300, Antofagasta, Chile.
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, 621-0427, Punta Arenas, Chile.
| | - Katherin V Pereyra
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Karla Schwarz
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Gigliola Ramirez
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Fernando C Ortiz
- Centro de Investigación Biomédica, Universidad Autónoma de Chile, Santiago, Chile.
| | - Marcelo E Andía
- Radiology Department & ANID - Millennium Nucleus for Cardiovascular Magnetic Resonance, 8331150, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, 621-0427, Punta Arenas, Chile; Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| |
Collapse
|
10
|
Pereyra KV, Schwarz KG, Andrade DC, Toledo C, Rios-Gallardo A, Díaz-Jara E, Bastías SS, Ortiz FC, Ortolani D, Del Rio R. Paraquat herbicide diminishes chemoreflex sensitivity, induces cardiac autonomic imbalance and impair cardiac function in rats. Am J Physiol Heart Circ Physiol 2021; 320:H1498-H1509. [PMID: 33513085 DOI: 10.1152/ajpheart.00710.2020] [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: 08/28/2020] [Accepted: 01/21/2021] [Indexed: 11/22/2022]
Abstract
Paraquat (PQT) herbicide is widely used in agricultural practices despite being highly toxic to humans. It has been proposed that PQT exposure may promote cardiorespiratory impairment. However, the physiological mechanisms involved in cardiorespiratory dysfunction following PQT exposure are poorly known. We aimed to determine the effects of PQT on ventilatory chemoreflex control, cardiac autonomic control, and cardiac function in rats. Male Sprague-Dawley rats received two injections/week of PQT (5 mg·kg-1 ip) for 4 wk. Cardiac function was assessed through echocardiography and pressure-volume loops. Ventilatory function was evaluated using whole body plethysmography. Autonomic control was indirectly evaluated by heart rate variability (HRV). Cardiac electrophysiology (EKG) and exercise capacity were also measured. Four weeks of PQT administration markedly enlarged the heart as evidenced by increases in ventricular volumes and induced cardiac diastolic dysfunction. Indeed, end-diastolic pressure was significantly higher in PQT rats compared with control (2.42 ± 0.90 vs. 4.01 ± 0.92 mmHg, PQT vs. control, P < 0.05). In addition, PQT significantly reduced both the hypercapnic and hypoxic ventilatory chemoreflex response and induced irregular breathing. Also, PQT induced autonomic imbalance and reductions in the amplitude of EKG waves. Finally, PQT administration impaired exercise capacity in rats as evidenced by a ∼2-fold decrease in times-to-fatigue compared with control rats. Our results showed that 4 wk of PQT treatment induces cardiorespiratory dysfunction in rats and suggests that repetitive exposure to PQT may induce harmful mid/long-term cardiovascular, respiratory, and cardiac consequences.NEW & NOREWORTHY Paraquat herbicide is still employed in agricultural practices in several countries. Here, we showed for the first time that 1 mo paraquat administration results in cardiac adverse remodeling, blunts ventilatory chemoreflex drive, and promotes irregular breathing at rest in previously healthy rats. In addition, paraquat exposure induced cardiac autonomic imbalance and cardiac electrophysiology alterations. Lastly, cardiac diastolic dysfunction was overt in rats following 1 mo of paraquat treatment.
Collapse
Affiliation(s)
- Katherin V Pereyra
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karla G Schwarz
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Envejecimiento y Regeneración, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - David C Andrade
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Fisiología y Medicina de Altura, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes, Universidad de Magallanes, Punta Arenas, Chile
| | - Angélica Rios-Gallardo
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Esteban Díaz-Jara
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sussy S Bastías
- Centro de Excelencia en Biomedicina de Magallanes, Universidad de Magallanes, Punta Arenas, Chile
| | - Fernando C Ortiz
- Mechanism of Myelin Formation and Repair Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Domiziana Ortolani
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Envejecimiento y Regeneración, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes, Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
11
|
Díaz HS, Andrade DC, Toledo C, Schwarz KG, Pereyra KV, Díaz-Jara E, Marcus NJ, Del Rio R. Inhibition of Brainstem Endoplasmic Reticulum Stress Rescues Cardiorespiratory Dysfunction in High Output Heart Failure. Hypertension 2020; 77:718-728. [PMID: 33307852 DOI: 10.1161/hypertensionaha.120.16056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recent evidence shows that chronic activation of catecholaminergic neurons of the rostral ventrolateral medulla is crucial in promoting autonomic imbalance and cardiorespiratory dysfunction in high output heart failure (HF). Brainstem endoplasmic reticulum stress (ERS) is known to promote cardiovascular dysfunction; however, no studies have addressed the potential role of brainstem ERS in cardiorespiratory dysfunction in high output HF. In this study, we assessed the presence of brainstem ERS and its potential role in cardiorespiratory dysfunction in an experimental model of HF induced by volume overload. High output HF was surgically induced via creation of an arterio-venous fistula in adult male Sprague-Dawley rats. Tauroursodeoxycholic acid (TUDCA), an inhibitor of ERS, or vehicle was administered intracerebroventricularly for 4 weeks post-HF induction. Compared with vehicle treatment, TUDCA improved cardiac autonomic balance (LFHRV/HFHRV ratio, 3.02±0.29 versus 1.14±0.24), reduced cardiac arrhythmia incidence (141.5±26.7 versus 35.67±12.5 events/h), and reduced abnormal respiratory patterns (Apneas: 11.83±2.26 versus 4.33±1.80 events/h). TUDCA administration (HF+Veh versus HF+TUDCA, P<0.05) attenuated cardiac hypertrophy (HW/BW 4.4±0.3 versus 4.0±0.1 mg/g) and diastolic dysfunction. Analysis of rostral ventrolateral medulla gene expression confirmed the presence of ERS, inflammation, and activation of renin-angiotensin system pathways in high output HF and showed that TUDCA treatment completely abolished ERS and ERS-related signaling. Taken together, these results support the notion that ERS plays a role in cardiorespiratory dysfunction in high output HF and more importantly that reducing brain ERS with TUDCA treatment has a potent salutary effect on cardiac function in this model.
Collapse
Affiliation(s)
- Hugo S Díaz
- From the Laboratory of Cardiorespiratory Control, Department of Physiology (H.S.D., D.C.A., C.T., K.G.S., K.V.P., E.D.-J., R.D.R.), Pontificia Universidad Católica de Chile, Santiago
| | - David C Andrade
- From the Laboratory of Cardiorespiratory Control, Department of Physiology (H.S.D., D.C.A., C.T., K.G.S., K.V.P., E.D.-J., R.D.R.), Pontificia Universidad Católica de Chile, Santiago.,Centro de Fisiología y Medicina de Altura, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile (D.C.A.)
| | - Camilo Toledo
- From the Laboratory of Cardiorespiratory Control, Department of Physiology (H.S.D., D.C.A., C.T., K.G.S., K.V.P., E.D.-J., R.D.R.), Pontificia Universidad Católica de Chile, Santiago
| | - Karla G Schwarz
- From the Laboratory of Cardiorespiratory Control, Department of Physiology (H.S.D., D.C.A., C.T., K.G.S., K.V.P., E.D.-J., R.D.R.), Pontificia Universidad Católica de Chile, Santiago.,Centro de Envejecimiento y Regeneración (CARE) (K.G.S., R.D.R.), Pontificia Universidad Católica de Chile, Santiago
| | - Katherin V Pereyra
- From the Laboratory of Cardiorespiratory Control, Department of Physiology (H.S.D., D.C.A., C.T., K.G.S., K.V.P., E.D.-J., R.D.R.), Pontificia Universidad Católica de Chile, Santiago
| | - Esteban Díaz-Jara
- From the Laboratory of Cardiorespiratory Control, Department of Physiology (H.S.D., D.C.A., C.T., K.G.S., K.V.P., E.D.-J., R.D.R.), Pontificia Universidad Católica de Chile, Santiago
| | - Noah J Marcus
- Department of Physiology and Pharmacology, Des Moines University, IA (N.J.M.)
| | - Rodrigo Del Rio
- From the Laboratory of Cardiorespiratory Control, Department of Physiology (H.S.D., D.C.A., C.T., K.G.S., K.V.P., E.D.-J., R.D.R.), Pontificia Universidad Católica de Chile, Santiago.,Centro de Envejecimiento y Regeneración (CARE) (K.G.S., R.D.R.), Pontificia Universidad Católica de Chile, Santiago.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile (R.D.R.)
| |
Collapse
|
12
|
Koba S, Hanai E, Kumada N, Watanabe T. Sympathoexcitatory input from hypothalamic paraventricular nucleus neurons projecting to rostral ventrolateral medulla is enhanced after myocardial infarction. Am J Physiol Heart Circ Physiol 2020; 319:H1197-H1207. [PMID: 32946261 DOI: 10.1152/ajpheart.00273.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Elevated sympathetic vasomotor tone seen in heart failure (HF) may involve dysfunction of the hypothalamic paraventricular nucleus neurons that project to the rostral ventrolateral medulla (PVN-RVLM neurons). This study aimed to elucidate the role of PVN-RVLM neurons in the maintenance of resting renal sympathetic nerve activity (RSNA) after myocardial infarction (MI). In male rats, the left coronary artery was chronically ligated to induce MI. The rats received PVN microinjections of an adeno-associated viral (AAV) vector encoding archaerhodopsin T (ArchT) with the reporter yellow fluorescence protein (eYFP). The ArchT rats had abundant distributions of eYFP-labeled, PVN-derived axons in the RVLM. In anesthetized ArchT rats with MI (n = 12), optogenetic inhibition of the PVN-RVLM pathway achieved by 532-nm-wavelength laser illumination to the RVLM significantly decreased RSNA. This effect was not found in sham-operated ArchT rats (n = 6). Other rat groups received RVLM microinjections of a retrograde AAV vector encoding the red light-drivable halorhodopsin Jaws (Jaws) with the reporter green fluorescence protein (GFP) and showed expression of GFP-labeled cell bodies and dendrites in the PVN. Laser illumination of the PVN at a 635 nm wavelength elicited significant renal sympathoinhibition in Jaws rats with MI (n = 9) but not in sham-operated Jaws rats (n = 8). These results indicate that sympathoexcitatory input from PVN-RVLM neurons is enhanced after MI, suggesting that this monosynaptic pathway is part of the central nervous system circuitry that plays a critical role in generating an elevated sympathetic vasomotor tone commonly seen with HF.NEW & NOTEWORTHY Using optogenetics in rats, we report that sympathoexcitatory input from hypothalamic paraventricular nucleus neurons that project to the rostral ventrolateral medulla is enhanced after myocardial infarction. It is suggested that this monosynaptic pathway makes up a key part of central nervous system circuitry underlying sympathetic hyperactivation commonly seen in heart failure.
Collapse
Affiliation(s)
- Satoshi Koba
- Division of Integrative Physiology, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Eri Hanai
- Division of Integrative Physiology, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Nao Kumada
- Division of Integrative Physiology, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Tatsuo Watanabe
- Division of Integrative Physiology, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| |
Collapse
|
13
|
Tapa S, Wang L, Francis Stuart SD, Wang Z, Jiang Y, Habecker BA, Ripplinger CM. Adrenergic supersensitivity and impaired neural control of cardiac electrophysiology following regional cardiac sympathetic nerve loss. Sci Rep 2020; 10:18801. [PMID: 33139790 PMCID: PMC7608682 DOI: 10.1038/s41598-020-75903-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
Myocardial infarction (MI) can result in sympathetic nerve loss in the infarct region. However, the contribution of hypo-innervation to electrophysiological remodeling, independent from MI-induced ischemia and fibrosis, has not been comprehensively investigated. We present a novel mouse model of regional cardiac sympathetic hypo-innervation utilizing a targeted-toxin (dopamine beta-hydroxylase antibody conjugated to saporin, DBH-Sap), and measure resulting electrophysiological and Ca2+ handling dynamics. Five days post-surgery, sympathetic nerve density was reduced in the anterior left ventricular epicardium of DBH-Sap hearts compared to control. In Langendorff-perfused hearts, there were no differences in mean action potential duration (APD80) between groups; however, isoproterenol (ISO) significantly shortened APD80 in DBH-Sap but not control hearts, resulting in a significant increase in APD80 dispersion in the DBH-Sap group. ISO also produced spontaneous diastolic Ca2+ elevation in DBH-Sap but not control hearts. In innervated hearts, sympathetic nerve stimulation (SNS) increased heart rate to a lesser degree in DBH-Sap hearts compared to control. Additionally, SNS produced APD80 prolongation in the apex of control but not DBH-Sap hearts. These results suggest that hypo-innervated hearts have regional super-sensitivity to circulating adrenergic stimulation (ISO), while having blunted responses to SNS, providing important insight into the mechanisms of arrhythmogenesis following sympathetic nerve loss.
Collapse
Affiliation(s)
- Srinivas Tapa
- Department of Pharmacology, UC Davis School of Medicine, 2419B Tupper Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Lianguo Wang
- Department of Pharmacology, UC Davis School of Medicine, 2419B Tupper Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Samantha D Francis Stuart
- Department of Pharmacology, UC Davis School of Medicine, 2419B Tupper Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Zhen Wang
- Department of Pharmacology, UC Davis School of Medicine, 2419B Tupper Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Yanyan Jiang
- Department of Pharmacology, UC Davis School of Medicine, 2419B Tupper Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Beth A Habecker
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
| | - Crystal M Ripplinger
- Department of Pharmacology, UC Davis School of Medicine, 2419B Tupper Hall, One Shields Ave, Davis, CA, 95616, USA.
| |
Collapse
|
14
|
Debi RA, Spector SP. Heart to breathe: partial ablation of rostral ventrolateral medulla catecholaminergic neurons mediates disordered breathing in volume overload heart failure rats. J Physiol 2020; 598:447-449. [PMID: 31900936 DOI: 10.1113/jp279368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Ryan Andrew Debi
- York University, Biology, 4700 Keele St., Toronto, Ontario, M3J 1P3, Canada
| | | |
Collapse
|