1
|
Althammer F, Roy RK, Kirchner MK, Podpecan Y, Helen J, McGrath S, Lira EC, Stern JE. Angiotensin-II drives changes in microglia-vascular interactions in rats with heart failure. Commun Biol 2024; 7:1537. [PMID: 39562706 PMCID: PMC11577102 DOI: 10.1038/s42003-024-07229-8] [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: 12/22/2023] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Activation of microglia, the resident immune cells of the central nervous system, leading to the subsequent release of pro-inflammatory cytokines, has been linked to cardiac remodeling, autonomic disbalance, and cognitive deficits in heart failure (HF). While previous studies emphasized the role of hippocampal Angiotensin II (AngII) signaling in HF-induced microglial activation, unanswered mechanistic questions persist. Evidence suggests significant interactions between microglia and local microvasculature, potentially affecting blood-brain barrier integrity and cerebral blood flow regulation. Still, whether the microglial-vascular interface is affected in the brain during HF remains unknown. Using a well-established ischemic HF rat model, we demonstrate the increased abundance of vessel-associated microglia (VAM) in HF rat hippocampi, along with an increased expression of AngII AT1a receptors. Acute AngII administration to sham rats induced microglia recruitment to brain capillaries, along with increased expression of TNFα. Conversely, administering an AT1aR blocker to HF rats prevented the recruitment of microglia to blood vessels, normalizing their levels to those in healthy rats. These results highlight the critical importance of a rather understudied phenomenon (i.e., microglia-vascular interactions in the brain) in the context of the pathophysiology of a highly prevalent cardiovascular disease, and unveil novel potential therapeutic avenues aimed at mitigating neuroinflammation in cardiovascular diseases.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Ranjan K Roy
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Matthew K Kirchner
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Yuval Podpecan
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Jemima Helen
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Shaina McGrath
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Elba Campos Lira
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Javier E Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA.
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
2
|
Yin Y, Li X, Zhang X, Yuan X, You X, Wu J. Inhibition of Extracellular Signal-Regulated Kinase Activity Improves Cognitive Function in Mice Subjected to Myocardial Infarction. Cardiovasc Toxicol 2024; 24:766-775. [PMID: 38850470 DOI: 10.1007/s12012-024-09877-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
Cognitive impairment is a commonly observed complication following myocardial infarction; however, the underlying mechanisms are still not well understood. The most recent research suggests that extracellular signal-regulated kinase (ERK) plays a critical role in the development and occurrence of cognitive dysfunction-related diseases. This study aims to explore whether the ERK inhibitor U0126 targets the ERK/Signal Transducer and Activator of Transcription 1 (STAT1) pathway to ameliorate cognitive impairment after myocardial infarction. To establish a mouse model of myocardial infarction, we utilized various techniques including Echocardiography, Hematoxylin-eosin (HE) staining, Elisa, Open field test, Elevated plus maze test, and Western blot analysis to assess mouse cardiac function, cognitive function, and signal transduction pathways. For further investigation into the mechanisms of cognitive function and signal transduction, we administered the ERK inhibitor U0126 via intraperitoneal injection. Reduced total distance and activity range were observed in mice subjected to myocardial infarction during the open field test, along with decreased exploration of the open arms in the elevated plus maze test. However, U0126 treatment exhibited a significant improvement in cognitive decline, indicating a protective effect through the inhibition of the ERK/STAT1 signaling pathway. Hence, this study highlights the involvement of the ERK/STAT1 pathway in regulating cognitive dysfunction following myocardial infarction and establishes U0126 as a promising therapeutic target.
Collapse
Affiliation(s)
- Yibo Yin
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, No. 241 West Huaihai Rd., Shanghai, China
| | - Xin Li
- School of Medical Instrument and Food Engineering USST, University of Shanghai for Science and Technology, Shanghai, China
| | - Xiaoxua Zhang
- School of Medicine, Shanghai University, No. 99 Shangda Road, Baoshan District, Shanghai, 200444, China
| | - Xinru Yuan
- School of Medicine, Shanghai University, No. 99 Shangda Road, Baoshan District, Shanghai, 200444, China
| | - Xingji You
- School of Medicine, Shanghai University, No. 99 Shangda Road, Baoshan District, Shanghai, 200444, China.
| | - Jingxiang Wu
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, No. 241 West Huaihai Rd., Shanghai, China.
- School of Medical Instrument and Food Engineering USST, University of Shanghai for Science and Technology, Shanghai, China.
| |
Collapse
|
3
|
Althammer F, Roy RK, Kirchner MK, McGrath S, Lira EC, Stern JE. Angiotensin-II drives changes in microglia-vascular interactions in rats with heart failure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.22.573045. [PMID: 38187537 PMCID: PMC10769361 DOI: 10.1101/2023.12.22.573045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Activation of microglia, the resident immune cells of the central nervous system, leading to the subsequent release of pro-inflammatory cytokines, has been linked to cardiac remodeling, autonomic disbalance, and cognitive deficits in heart failure (HF). While previous studies emphasized the role of hippocampal Angiotensin II (AngII) signaling in HF-induced microglial activation, unanswered mechanistic questions persist. Evidence suggests significant interactions between microglia and local microvasculature, potentially affecting blood-brain barrier integrity and cerebral blood flow regulation. Still, whether the microglial-vascular interface is affected in the brain during HF remains unknow. Using a well-established ischemic HF rat model, we demonstrate increased vessel-associated microglia (VAM) in HF rat hippocampi, which showed heightened expression of AngII AT1a receptors. Acute AngII administration to sham rats induced microglia recruitment to the perivascular space, along with increased expression of TNFa. Conversely, administering an AT1aR blocker to HF rats prevented the recruitment of microglia to the perivascular space, normalizing their levels to those in healthy rats. These results highlight the critical importance of a rather understudied phenomenon (i.e., microglia-vascular interactions in the brain) in the context of the pathophysiology of a highly prevalent cardiovascular disease, and unveil novel potential therapeutic avenues aimed at mitigating neuroinflammation in cardiovascular diseases.
Collapse
|
4
|
Althammer F, Roy RK, Kirchner MK, Campos-Lira E, Whitley KE, Davis S, Montanez J, Ferreira-Neto HC, Danh J, Feresin R, Biancardi VC, Zafar U, Parent MB, Stern JE. Angiotensin II-Mediated Neuroinflammation in the Hippocampus Contributes to Neuronal Deficits and Cognitive Impairment in Heart Failure Rats. Hypertension 2023; 80:1258-1273. [PMID: 37035922 PMCID: PMC10192104 DOI: 10.1161/hypertensionaha.123.21070] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/22/2023] [Indexed: 04/11/2023]
Abstract
BACKGROUND Heart failure (HF) is a debilitating disease affecting >64 million people worldwide. In addition to impaired cardiovascular performance and associated systemic complications, most patients with HF suffer from depression and substantial cognitive decline. Although neuroinflammation and brain hypoperfusion occur in humans and rodents with HF, the underlying neuronal substrates, mechanisms, and their relative contribution to cognitive deficits in HF remains unknown. METHODS To address this critical gap in our knowledge, we used a well-established HF rat model that mimics clinical outcomes observed in the human population, along with a multidisciplinary approach combining behavioral, electrophysiological, neuroanatomical, molecular and systemic physiological approaches. RESULTS Our studies support neuroinflammation, hypoperfusion/hypoxia, and neuronal deficits in the hippocampus of HF rats, which correlated with the progression and severity of the disease. An increased expression of AT1aRs (Ang II [angiotensin II] receptor type 1a) in hippocampal microglia preceded the onset of neuroinflammation. Importantly, blockade of AT1Rs with a clinically used therapeutic drug (Losartan), and delivered in a clinically relevant manner, efficiently reversed neuroinflammatory end points (but not hypoxia ones), resulting in turn in improved cognitive performance in HF rats. Finally, we show than circulating Ang II can leak and access the hippocampal parenchyma in HF rats, constituting a possible source of Ang II initiating the neuroinflammatory signaling cascade in HF. CONCLUSIONS In this study, we identified a neuronal substrate (hippocampus), a mechanism (Ang II-driven neuroinflammation) and a potential neuroprotective therapeutic target (AT1aRs) for the treatment of cognitive deficits in HF.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Center for Neuroinflammation and Cardiometabolic Diseases,
Georgia State University, GA, USA
| | - Ranjan K. Roy
- Center for Neuroinflammation and Cardiometabolic Diseases,
Georgia State University, GA, USA
| | - Matthew K. Kirchner
- Center for Neuroinflammation and Cardiometabolic Diseases,
Georgia State University, GA, USA
| | - Elba Campos-Lira
- Center for Neuroinflammation and Cardiometabolic Diseases,
Georgia State University, GA, USA
- Neuroscience Institute, Georgia State University, GA,
USA
| | | | - Steven Davis
- Neuroscience Institute, Georgia State University, GA,
USA
| | - Juliana Montanez
- Center for Neuroinflammation and Cardiometabolic Diseases,
Georgia State University, GA, USA
| | | | - Jessica Danh
- Department of Nutrition, Georgia State University, Atlanta,
GA 30302, USA
| | - Rafaela Feresin
- Department of Nutrition, Georgia State University, Atlanta,
GA 30302, USA
| | - Vinicia Campana Biancardi
- Anatomy, Physiology, & Pharmacology, College of
Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Usama Zafar
- Center for Neuroinflammation and Cardiometabolic Diseases,
Georgia State University, GA, USA
- Neuroscience Institute, Georgia State University, GA,
USA
| | - Marise B. Parent
- Center for Neuroinflammation and Cardiometabolic Diseases,
Georgia State University, GA, USA
- Neuroscience Institute, Georgia State University, GA,
USA
- Department of Psychology, Georgia State University,
Atlanta, GA 30302, USA
| | - Javier E. Stern
- Center for Neuroinflammation and Cardiometabolic Diseases,
Georgia State University, GA, USA
- Neuroscience Institute, Georgia State University, GA,
USA
| |
Collapse
|
5
|
Althammer F, Roy RK, Lefevre A, Najjar RS, Schoenig K, Bartsch D, Eliava M, Feresin RG, Hammock EA, Murphy AZ, Charlet A, Grinevich V, Stern JE. Altered PVN-to-CA2 hippocampal oxytocin pathway and reduced number of oxytocin-receptor expressing astrocytes in heart failure rats. J Neuroendocrinol 2022; 34:e13166. [PMID: 35657290 PMCID: PMC9495289 DOI: 10.1111/jne.13166] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 11/28/2022]
Abstract
Oxytocinergic actions within the hippocampal CA2 are important for neuromodulation, memory processing and social recognition. However, the source of the OTergic innervation, the cellular targets expressing the OT receptors (OTRs) and whether the PVN-to-CA2 OTergic system is altered during heart failure (HF), a condition recently associated with cognitive and mood decline, remains unknown. Using immunohistochemistry along with retrograde monosynaptic tracing, RNAscope and a novel OTR-Cre rat line, we show that the PVN (but not the supraoptic nucleus) is an important source of OTergic innervation to the CA2. These OTergic fibers were found in many instances in close apposition to OTR expressing cells within the CA2. Interestingly, while only a small proportion of neurons were found to express OTRs (~15%), this expression was much more abundant in CA2 astrocytes (~40%), an even higher proportion that was recently reported for astrocytes in the central amygdala. Using an established ischemic rat heart failure (HF) model, we found that HF resulted in robust changes in the PVN-to-CA2 OTergic system, both at the source and target levels. Within the PVN, we found an increased OT immunoreactivity, along with a diminished OTR expression in PVN neurons. Within the CA2 of HF rats, we observed a blunted OTergic innervation, along with a diminished OTR expression, which appeared to be restricted to CA2 astrocytes. Taken together, our studies highlight astrocytes as key cellular targets mediating OTergic PVN inputs to the CA2 hippocampal region. Moreover, they provide the first evidence for an altered PVN-to-CA2 OTergic system in HF rats, which could potentially contribute to previously reported cognitive and mood impairments in this animal model.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
| | - Ranjan K. Roy
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
| | - Arthur Lefevre
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, University of Heidelberg, Mannheim 68159, Germany
| | - Rami S. Najjar
- Department of Nutrition, Georgia State University, Atlanta, GA 30302, USA
| | - Kai Schoenig
- Department of Molecular Biology Central Institute of Mental Health J5 68159 Mannheim Germany
| | - Dusan Bartsch
- Department of Molecular Biology Central Institute of Mental Health J5 68159 Mannheim Germany
| | - Marina Eliava
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, University of Heidelberg, Mannheim 68159, Germany
| | - Rafaela G. Feresin
- Department of Nutrition, Georgia State University, Atlanta, GA 30302, USA
| | - Elizabeth A.D. Hammock
- Department of Psychology and Program in Neuroscience, The Florida State University, Tallahassee, FL 32306, USA
| | - Anne Z. Murphy
- Neuroscience Institute, Georgia State University, Atlanta, USA
| | - Alexandre Charlet
- Centre National de la Recherche Scientifique and University of Strasbourg, Institute of Cellular and Integrative Neuroscience, 67000 Strasbourg, France
| | - Valery Grinevich
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, University of Heidelberg, Mannheim 68159, Germany
| | - Javier E. Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
6
|
Roy RK, Ferreira-Neto HC, Felder RB, Stern JE. Angiotensin II inhibits the A-type K + current of hypothalamic paraventricular nucleus neurons in rats with heart failure: role of MAPK-ERK1/2 signaling. Am J Physiol Regul Integr Comp Physiol 2022; 322:R526-R534. [PMID: 35319903 PMCID: PMC9076419 DOI: 10.1152/ajpregu.00308.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/03/2022] [Accepted: 03/18/2022] [Indexed: 11/22/2022]
Abstract
Angiotensin II (ANG II)-mediated sympathohumoral activation constitutes a pathophysiological mechanism in heart failure (HF). Although the hypothalamic paraventricular nucleus (PVN) is a major site mediating ANG II effects in HF, the precise mechanisms by which ANG II influences sympathohumoral outflow from the PVN remain unknown. ANG II activates the ubiquitous intracellular MAPK signaling cascades, and recent studies revealed a key role for ERK1/2 MAPK signaling in ANG II-mediated sympathoexcitation in HF rats. Importantly, ERK1/2 was reported to inhibit the transient outward potassium current (IA) in hippocampal neurons. Given that IA is a critical determinant of the PVN neuronal excitability, and that downregulation of IA in the brain has been reported in cardiovascular disease states, including HF, we investigated here whether ANG II modulates IA in PVN neurons via the MAPK-ERK pathway, and, whether these effects are altered in HF rats. Patch-clamp recordings from identified magnocellular neurosecretory neurons (MNNs) and presympathetic (PS) PVN neurons revealed that ANG II inhibited IA in both PVN neuronal types, both in sham and HF rats. Importantly, ANG II effects were blocked by inhibiting MAPK-ERK signaling as well as by inhibiting epidermal growth factor receptor (EGFR), a gateway to MAPK-ERK signaling. Although no differences in basal IA magnitude were found between sham and HF rats under normal conditions, MAPK-ERK blockade resulted in significantly larger IA in both PVN neuronal types in HF rats. Taken together, our studies show that ANG II-induced ERK1/2 activity inhibits IA, an effect expected to increase the excitability of presympathetic and neuroendocrine PVN neurons, contributing in turn to the neurohumoral overactivity that promotes progression of the HF syndrome.
Collapse
Affiliation(s)
- Ranjan K Roy
- Neuroscience Institute, Georgia State University, Atlanta, Georgia
| | | | - Robert B Felder
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Javier E Stern
- Neuroscience Institute, Georgia State University, Atlanta, Georgia
| |
Collapse
|
7
|
Tran N, Garcia T, Aniqa M, Ali S, Ally A, Nauli SM. Endothelial Nitric Oxide Synthase (eNOS) and the Cardiovascular System: in Physiology and in Disease States. AMERICAN JOURNAL OF BIOMEDICAL SCIENCE & RESEARCH 2022; 15:153-177. [PMID: 35072089 PMCID: PMC8774925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Endothelial nitric oxide synthase (eNOS) plays a critical role in regulating and maintaining a healthy cardiovascular system. The importance of eNOS can be emphasized from the genetic polymorphisms of the eNOS gene, uncoupling of eNOS dimerization, and its numerous signaling regulations. The activity of eNOS on the cardiac myocytes, vasculature, and the central nervous system are discussed. The effects of eNOS on the sympathetic autonomic nervous system (SANS) and the parasympathetic autonomic nervous system (PANS), both of which profoundly influence the cardiovascular system, will be elaborated. The relationship between the eNOS protein with cardiovascular autonomic reflexes such as the baroreflex and the Exercise Pressor Reflex will be discussed. For example, the effects of endogenous nitric oxide (NO) are shown to be mediated by the eNOS protein and that eNOS-derived endothelial NO is most effective in regulating blood pressure oscillations via modulating the baroreflex mechanisms. The protective action of eNOS on the CVS is emphasized here because dysfunction of the eNOS enzyme is intricately correlated with the pathogenesis of several cardiovascular diseases such as hypertension, arteriosclerosis, myocardial infarction, and stroke. Overall, our current understanding of the eNOS protein with a focus on its role in the modulation, regulation, and control of the cardiovascular system in a normal physiological state and in cardiovascular diseases are discussed.
Collapse
Affiliation(s)
- N Tran
- Arkansas College of Osteopathic Medicine, Fort Smith, AR, USA
| | - T Garcia
- Arkansas College of Osteopathic Medicine, Fort Smith, AR, USA
| | - M Aniqa
- Arkansas College of Osteopathic Medicine, Fort Smith, AR, USA
| | - S Ali
- Arkansas College of Osteopathic Medicine, Fort Smith, AR, USA
| | - A Ally
- Arkansas College of Osteopathic Medicine, Fort Smith, AR, USA,Corresponding author: Surya M Nauli, Chapman University and University of California, Irvine, CA, USA
| | - SM Nauli
- Chapman University and University of California, Irvine, CA, USA,Corresponding author: Surya M Nauli, Chapman University and University of California, Irvine, CA, USA
| |
Collapse
|
8
|
Grinevich V, Ludwig M. The multiple faces of the oxytocin and vasopressin systems in the brain. J Neuroendocrinol 2021; 33:e13004. [PMID: 34218479 DOI: 10.1111/jne.13004] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/17/2021] [Accepted: 06/10/2021] [Indexed: 11/30/2022]
Abstract
Classically, hypothalamic neuroendocrine cells that synthesise oxytocin and vasopressin were categorised in two major cell types: the magnocellular and parvocellular neurones. It was assumed that magnocellular neurones project exclusively to the pituitary gland where they release oxytocin and vasopressin into the systemic circulation. The parvocellular neurones, on the other hand, project within the brain to regulate discrete brain circuitries and behaviours. Within the last few years, it has become evident that the classical view of these projections is outdated. It is now clear that oxytocin and vasopressin in the brain are released extrasynaptically from dendrites and from varicosities in distant axons. The peptides act principally to modulate information transfer through conventional synapses (such as glutamate synapses) by actions at respective receptors that may be preferentially localised to synaptic regions (on either side of the synapse) to alter the 'gain' of conventional synapses.
Collapse
Affiliation(s)
- Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
- Centre for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
| | - Mike Ludwig
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Centre for Neuroendocrinology, Department of Immunology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
9
|
Parent MB, Ferreira-Neto HC, Kruemmel AR, Althammer F, Patel AA, Keo S, Whitley KE, Cox DN, Stern JE. Heart failure impairs mood and memory in male rats and down-regulates the expression of numerous genes important for synaptic plasticity in related brain regions. Behav Brain Res 2021; 414:113452. [PMID: 34274373 DOI: 10.1016/j.bbr.2021.113452] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 05/21/2021] [Accepted: 07/08/2021] [Indexed: 12/01/2022]
Abstract
Chronic heart failure (HF) is a serious disorder that afflicts more than 26 million patients worldwide. HF is comorbid with depression, anxiety and memory deficits that have serious implications for quality of life and self-care in patients who have HF. Still, there are few studies that have assessed the effects of severely reduced ejection fraction (≤40 %) on cognition in non-human animal models. Moreover, limited information is available regarding the effects of HF on genetic markers of synaptic plasticity in brain areas critical for memory and mood regulation. We induced HF in male rats and tested mood and anxiety (sucrose preference and elevated plus maze) and memory (spontaneous alternation and inhibitory avoidance) and measured the simultaneous expression of 84 synaptic plasticity-associated genes in dorsal (DH) and ventral hippocampus (VH), basolateral (BLA) and central amygdala (CeA) and prefrontal cortex (PFC). We also included the hypothalamic paraventricular nucleus (PVN), which is implicated in neurohumoral activation in HF. Our results show that rats with severely reduced ejection fraction recapitulate behavioral symptoms seen in patients with chronic HF including, increased anxiety and impaired memory in both tasks. HF also downregulated several synaptic-plasticity genes in PFC and PVN, moderate decreases in DH and CeA and minimal effects in BLA and VH. Collectively, these findings identify candidate brain areas and molecular mechanisms underlying HF-induced disturbances in mood and memory.
Collapse
Affiliation(s)
- Marise B Parent
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | | | | | | | - Atit A Patel
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Sreinick Keo
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | | | - Daniel N Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Javier E Stern
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
10
|
Althammer F, Ferreira-Neto HC, Rubaharan M, Roy RK, Patel AA, Murphy A, Cox DN, Stern JE. Three-dimensional morphometric analysis reveals time-dependent structural changes in microglia and astrocytes in the central amygdala and hypothalamic paraventricular nucleus of heart failure rats. J Neuroinflammation 2020; 17:221. [PMID: 32703230 PMCID: PMC7379770 DOI: 10.1186/s12974-020-01892-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cardiovascular diseases, including heart failure, are the most common cause of death globally. Recent studies support a high degree of comorbidity between heart failure and cognitive and mood disorders resulting in memory loss, depression, and anxiety. While neuroinflammation in the hypothalamic paraventricular nucleus contributes to autonomic and cardiovascular dysregulation in heart failure, mechanisms underlying cognitive and mood disorders in this disease remain elusive. The goal of this study was to quantitatively assess markers of neuroinflammation (glial morphology, cytokines, and A1 astrocyte markers) in the central amygdala, a critical forebrain region involved in emotion and cognition, and to determine its time course and correlation to disease severity during the progression of heart failure. METHODS We developed and implemented a comprehensive microglial/astrocyte profiler for precise three-dimensional morphometric analysis of individual microglia and astrocytes in specific brain nuclei at different time points during the progression of heart failure. To this end, we used a well-established ischemic heart failure rat model. Morphometric studies were complemented with quantification of various pro-inflammatory cytokines and A1/A2 astrocyte markers via qPCR. RESULTS We report structural remodeling of central amygdala microglia and astrocytes during heart failure that affected cell volume, surface area, filament length, and glial branches, resulting overall in somatic swelling and deramification, indicative of a change in glial state. These changes occurred in a time-dependent manner, correlated with the severity of heart failure, and were delayed compared to changes in the hypothalamic paraventricular nucleus. Morphometric changes correlated with elevated mRNA levels of pro-inflammatory cytokines and markers of reactive A1-type astrocytes in the paraventricular nucleus and central amygdala during heart failure. CONCLUSION We provide evidence that in addition to the previously described hypothalamic neuroinflammation implicated in sympathohumoral activation during heart failure, microglia, and astrocytes within the central amygdala also undergo structural remodeling indicative of glial shifts towards pro-inflammatory phenotypes. Thus, our studies suggest that neuroinflammation in the amygdala stands as a novel pathophysiological mechanism and potential therapeutic target that could be associated with emotional and cognitive deficits commonly observed at later stages during the course of heart failure.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, USA
| | | | | | - Ranjan K Roy
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, USA
| | - Atit A Patel
- Neuroscience Institute, Georgia State University, Atlanta, USA
| | - Anne Murphy
- Neuroscience Institute, Georgia State University, Atlanta, USA
| | - Daniel N Cox
- Neuroscience Institute, Georgia State University, Atlanta, USA
| | - Javier E Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, USA.
| |
Collapse
|
11
|
Chen J, Yin D, He X, Gao M, Choi Y, Luo G, Wang H, Qu X. Modulation of activated astrocytes in the hypothalamus paraventricular nucleus to prevent ventricular arrhythmia complicating acute myocardial infarction. Int J Cardiol 2020; 308:33-41. [PMID: 31987663 DOI: 10.1016/j.ijcard.2020.01.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/06/2020] [Accepted: 01/15/2020] [Indexed: 02/06/2023]
|
12
|
Brown CH, Ludwig M, Tasker JG, Stern JE. Somato-dendritic vasopressin and oxytocin secretion in endocrine and autonomic regulation. J Neuroendocrinol 2020; 32:e12856. [PMID: 32406599 PMCID: PMC9134751 DOI: 10.1111/jne.12856] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/29/2020] [Accepted: 04/11/2020] [Indexed: 12/29/2022]
Abstract
Somato-dendritic secretion was first demonstrated over 30 years ago. However, although its existence has become widely accepted, the function of somato-dendritic secretion is still not completely understood. Hypothalamic magnocellular neurosecretory cells were among the first neuronal phenotypes in which somato-dendritic secretion was demonstrated and are among the neurones for which the functions of somato-dendritic secretion are best characterised. These neurones secrete the neuropeptides, vasopressin and oxytocin, in an orthograde manner from their axons in the posterior pituitary gland into the blood circulation to regulate body fluid balance and reproductive physiology. Retrograde somato-dendritic secretion of vasopressin and oxytocin modulates the activity of the neurones from which they are secreted, as well as the activity of neighbouring populations of neurones, to provide intra- and inter-population signals that coordinate the endocrine and autonomic responses for the control of peripheral physiology. Somato-dendritic vasopressin and oxytocin have also been proposed to act as hormone-like signals in the brain. There is some evidence that somato-dendritic secretion from magnocellular neurosecretory cells modulates the activity of neurones beyond their local environment where there are no vasopressin- or oxytocin-containing axons but, to date, there is no conclusive evidence for, or against, hormone-like signalling throughout the brain, although it is difficult to imagine that the levels of vasopressin found throughout the brain could be underpinned by release from relatively sparse axon terminal fields. The generation of data to resolve this issue remains a priority for the field.
Collapse
Affiliation(s)
- Colin H. Brown
- Department of Physiology, Brain Health Research Centre, Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - Mike Ludwig
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Department of Immunology, Centre for Neuroendocrinology, University of Pretoria, Pretoria, South Africa
| | - Jeffrey G. Tasker
- Department of Cell and Molecular Biology, Brain Institute, Tulane University, New Orleans, LA, USA
| | - Javier E. Stern
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
13
|
Ferreira-Neto HC, Stern JE. Functional coupling between NMDA receptors and SK channels in rat hypothalamic magnocellular neurons: altered mechanisms during heart failure. J Physiol 2019; 599:507-520. [PMID: 31667845 DOI: 10.1113/jp278910] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Glutamatergic NMDA receptors (NMDARs) and small conductance Ca2+ -activated K+ (SK) channels are critical synaptic and intrinsic mechanisms, respectively, that regulate the activity of hypothalamic magnocellular neurosecretory neurons (MNNs). In this work, we investigated whether NMDARs and SK channels in MNNs are functionally coupled, and whether an altered coupling may contribute to exacerbated neuronal activity in this condition. We report that NMDARs and SK channels form a functional Ca2+ -dependent negative feedback loop that restrains the excitatory effect on membrane potential and firing activity evoked by NMDAR activation. The negative feedback loop between NMDARs and SK channels was blunted or absent in MNNs of heart failure (HF) rats. These results help us better understand how synaptic and intrinsic mechanisms regulate hypothalamic neuronal activity, as well as how changes in the interaction among these disparate mechanisms contribute to altered neuronal activity during prevalent neurogenic cardiovascular diseases. ABSTRACT Glutamatergic NMDA receptors (NMDARs) and small conductance Ca2+ -activated K+ (SK) channels are critical synaptic and intrinsic mechanisms, respectively, that regulate the activity of hypothalamic magnocellular neurosecretory neurons (MNNs), both under physiological and pathological states, such as lactation and heart failure (HF). However, whether NMDARs and SK channels in MNNs are functionally coupled, and whether changes in this coupling contribute to exacerbated neuronal activity during HF is at present unknown. In the present study, we addressed these questions using patch-clamp electrophysiology and confocal Ca2+ imaging in a rat model of ischaemic HF. We found that in MNNs of sham rats, blockade of SK channels with apamin (200 nM) significantly increased the magnitude of an NMDAR-evoked current (INMDA ). We also observed that blockade of SK channels potentiated NMDAR-evoked firing, and abolished spike frequency adaptation in MNNs from sham, but not HF rats. Importantly, a larger INMDA -ΔCa2+ response was observed under basal conditions in HF compared to sham rats. Finally, we found that dialysing recorded cells with the Ca2+ chelator BAPTA (10 mM) increased the magnitude of INMDA in MNNs from both sham and HF rats, and occluded the effects of apamin in the former. Together our studies demonstrate that in MNNs, NMDARs and SK channels are functionally coupled, forming a local negative feedback loop that restrains the excitatory effect evoked by NMDAR activation. Moreover, our studies also support a blunted NMDAR-SK channel coupling in MNNs of HF rats, establishing it as a pathophysiological mechanism contributing to exacerbated hypothalamic neuronal activity during this prevalent neurogenic cardiovascular disease.
Collapse
Affiliation(s)
| | - Javier E Stern
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
14
|
Affiliation(s)
- Costantino Iadecola
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York.
| |
Collapse
|
15
|
McBryde FD, Liu BH, Roloff EV, Kasparov S, Paton JFR. Hypothalamic paraventricular nucleus neuronal nitric oxide synthase activity is a major determinant of renal sympathetic discharge in conscious Wistar rats. Exp Physiol 2018; 103:419-428. [PMID: 29215757 DOI: 10.1113/ep086744] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 11/30/2017] [Indexed: 12/17/2022]
Abstract
NEW FINDINGS What is the central question of this study? Does chronic reduction of neuronally generated nitric oxide in the hypothalamic paraventricular nucleus affect the set-point regulation of blood pressure and sympathetic activity destined to the kidneys? What is the main finding and its importance? Within the hypothalamic paraventricular nucleus, nitric oxide generated by neuronal nitric oxide synthase plays a major constitutive role in suppressing long term the levels of both ongoing renal sympathetic activity and arterial pressure in conscious Wistar rats. This finding unequivocally demonstrates a mechanism by which the diencephalon exerts a tonic influence on sympathetic discharge to the kidney and may provide the basis for both blood volume and osmolality homeostasis. ABSTRACT The paraventricular nucleus (PVN) of the hypothalamus plays a crucial role in cardiovascular and neuroendocrine regulation. Application of nitric oxide donors to the PVN stimulates GABAergic transmission, and may suppress sympathetic nerve activity (SNA) to lower arterial pressure. However, the role of endogenous nitric oxide within the PVN in regulating renal SNA chronically remains to be established in conscious animals. To address this, we used our previously established lentiviral vectors to knock down neuronal nitric oxide synthase (nNOS) selectively in the PVN of conscious Wistar rats. Blood pressure and renal SNA were monitored simultaneously and continuously for 21 days (n = 14) using radio-telemetry. Renal SNA was normalized to maximal evoked discharge and expressed as a percentage change from baseline. The PVN was microinjected bilaterally with a neurone-specific tetracycline-controllable lentiviral vector, expressing a short hairpin miRNA30 interference system targeting nNOS (n = 7) or expressing a mis-sense as control (n = 7). Recordings continued for a further 18 days. The vectors also expressed green fluorescent protein, and successful expression in the PVN and nNOS knockdown were confirmed histologically post hoc. Knockdown of nNOS expression in the PVN resulted in a sustained increase in blood pressure (from 95 ± 2 to 104 ± 3 mmHg, P < 0.05), with robust concurrent sustained activation of renal SNA (>70%, P < 0.05). The study reveals a major role for nNOS-derived nitric oxide within the PVN in chronic set-point regulation of cardiovascular autonomic activity in the conscious, normotensive rat.
Collapse
Affiliation(s)
- F D McBryde
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, UK.,Cardiovascular Autonomic Research Cluster, Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - B H Liu
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, UK
| | - E V Roloff
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, UK
| | - S Kasparov
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, UK
| | - J F R Paton
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, UK.,Cardiovascular Autonomic Research Cluster, Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
16
|
Ferreira‐Neto HC, Biancardi VC, Stern JE. A reduction in SK channels contributes to increased activity of hypothalamic magnocellular neurons during heart failure. J Physiol 2017; 595:6429-6442. [PMID: 28714070 PMCID: PMC5638886 DOI: 10.1113/jp274730] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/03/2017] [Indexed: 01/18/2023] Open
Abstract
KEY POINTS Small conductance Ca2+ -activated K+ (SK) channels play an important role in regulating the excitability of magnocellular neurosecretory cells (MNCs). Although an increased SK channel function contributes to adaptive physiological responses, it remains unknown whether changes in SK channel function/expression contribute to exacerbated MNC activity under disease conditions. We show that the input-output function of MNCs in heart failure (HF) rats is enhanced. Moreover, the SK channel blocker apamin enhanced the input-output function in sham, although not in HF rats. We found that both the after-hyperpolarizing potential magnitude and the underlying apamin-sensitive IAHP are blunted in MNCs from HF rats. The magnitude of spike-induced increases in intracellular Ca2+ levels was not affected in MNCs of HF rats. We found a diminished expression of SK2/SK3 channel subunit mRNA expression in the supraoptic nucleus of HF rats. Our studies suggest that a reduction in SK channel expression, but not changes in Ca2+ -mediated activation of SK channels, contributes to exacerbated MNC activity in HF rats. ABSTRACT Small conductance Ca2+ -activated K+ channels (SK) play an important role in regulating the activity of magnocellular neurosecretory cells (MNCs) and hormone release from the posterior pituitary. Moreover, enhanced SK activity contributes to the adaptive responses of MNCs to physiological challenge, such as lactation. Nevertheless, whether changes in SK function/expression contribute to exacerbated MNC activity during diseases such as heart failure (HF) remains unknown. In the present study, we used a combination of patch clamp electrophysiology, confocal Ca2+ imaging and molecular biology in a rat model of ischaemic HF. We found that the input-output function of MNCs was enhanced in HF compared to sham rats. Moreover, although the SK blocker apamin (200 nm) strengthened the input-output function in sham rats, it failed to have an effect in HF rats. The magnitude of the after-hyperpolarizing potential (AHP) following a train of spikes and the underlying apamin-sensitive IAHP were blunted in MNCs from HF rats. However, spike-induced increases in intracellular Ca2+ were not affected in the MNCs of HF rats. Real-time PCR measurements of SK channel subunits mRNA in supraoptic nucleus punches revealed a diminished expression of SK2/SK3 subunits in HF compared to sham rats. Together, our studies demonstrate that MNCs from HF rats exhibit increased membrane excitability and an enhanced input-output function, and also that a reduction in SK channel-mediated, apamin-sensitive AHP is a critical contributing mechanism. Moreover, our results suggest that the reduced AHP is related to a down-regulation of SK2/SK3 channel subunit expression but not the result of a blunted activity-dependent intracellular Ca2+ increase following a burst of action potentials.
Collapse
|
17
|
Sharma NM, Patel KP. Post-translational regulation of neuronal nitric oxide synthase: implications for sympathoexcitatory states. Expert Opin Ther Targets 2017; 21:11-22. [PMID: 27885874 PMCID: PMC5488701 DOI: 10.1080/14728222.2017.1265505] [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: 03/14/2016] [Accepted: 11/23/2016] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Nitric oxide (NO) synthesized via neuronal nitric oxide synthase (nNOS) plays a significant role in regulation/modulation of autonomic control of circulation. Various pathological states are associated with diminished nNOS expression and blunted autonomic effects of NO in the central nervous system (CNS) including heart failure, hypertension, diabetes mellitus, chronic renal failure etc. Therefore, elucidation of the molecular mechanism/s involved in dysregulation of nNOS is essential to understand the pathogenesis of increased sympathoexcitation in these diseased states. Areas covered: nNOS is a highly regulated enzyme, being regulated at transcriptional and posttranslational levels via protein-protein interactions and modifications viz. phosphorylation, ubiquitination, and sumoylation. The enzyme activity of nNOS also depends on the optimal concentration of substrate, cofactors and association with regulatory proteins. This review focuses on the posttranslational regulation of nNOS in the context of normal and diseased states within the CNS. Expert opinion: Gaining insight into the mechanism/s involved in the regulation of nNOS would provide novel strategies for manipulating nNOS directed therapeutic modalities in the future, including catalytically active dimer stabilization and protein-protein interactions with intracellular protein effectors. Ultimately, this is expected to provide tools to improve autonomic dysregulation in various diseases such as heart failure, hypertension, and diabetes.
Collapse
Affiliation(s)
- Neeru M Sharma
- a Department of Cellular & Integrative Physiology , University of Nebraska Medical Center , Omaha , NE , USA
| | - Kaushik P Patel
- a Department of Cellular & Integrative Physiology , University of Nebraska Medical Center , Omaha , NE , USA
| |
Collapse
|
18
|
Stern JE, Son S, Biancardi VC, Zheng H, Sharma N, Patel KP. Astrocytes Contribute to Angiotensin II Stimulation of Hypothalamic Neuronal Activity and Sympathetic Outflow. Hypertension 2016; 68:1483-1493. [PMID: 27698069 DOI: 10.1161/hypertensionaha.116.07747] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 05/17/2016] [Accepted: 08/24/2016] [Indexed: 02/07/2023]
Abstract
Angiotensin II (AngII) is a key neuropeptide that acting within the brain hypothalamic paraventricular nucleus regulates neurohumoral outflow to the circulation. Moreover, an exacerbated AngII action within the paraventricular nucleus contributes to neurohumoral activation in hypertension. Although AngII effects involve changes in paraventricular nucleus neuronal activity, the precise underlying mechanisms, cellular targets, and distribution of AngII receptors within the paraventricular nucleus remain largely unknown. Thus, whether AngII effects involve direct actions on paraventricular neurons, or whether it acts via intermediary cells, such as astrocytes, is still controversial. To address this important gap in our knowledge, we used a multidisciplinary approach combining patch-clamp electrophysiology in presympathetic paraventricular neurons and astrocytes, along with in vivo sympathetic nerve recordings and astrocyte-targeted gene manipulations. We present evidence for a novel mechanism underlying central AngII actions, which involves astrocytes as major intermediary cellular targets. We found that AngII type 1 receptor mRNA is expressed in paraventricular astrocytes. Moreover, we report that AngII inhibited glutamate transporter function, increasing in turn extracellular glutamate levels. This resulted in the activation of neuronal extrasynaptic NMDA (N-methyl-d-aspartate) receptors, increased presympathetic neuronal activity, enhanced sympathoexcitatory outflow, and increased blood pressure. Together, our studies support astrocytes as critical intermediary cell types mediating brain AngII regulation of the circulation and indicate that AngII-mediated neuronal and sympathoexcitatory effects are dependent on a unique neuroglial signaling modality involving nonsynaptic glutamate transmission.
Collapse
Affiliation(s)
- Javier E Stern
- From the Department of Physiology, Augusta University, GA (J.E.S., S.S., V.C.B.); and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (H.Z., N.S., K.P.P.).
| | - Sookjin Son
- From the Department of Physiology, Augusta University, GA (J.E.S., S.S., V.C.B.); and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (H.Z., N.S., K.P.P.)
| | - Vinicia C Biancardi
- From the Department of Physiology, Augusta University, GA (J.E.S., S.S., V.C.B.); and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (H.Z., N.S., K.P.P.)
| | - Hong Zheng
- From the Department of Physiology, Augusta University, GA (J.E.S., S.S., V.C.B.); and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (H.Z., N.S., K.P.P.)
| | - Neeru Sharma
- From the Department of Physiology, Augusta University, GA (J.E.S., S.S., V.C.B.); and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (H.Z., N.S., K.P.P.)
| | - Kaushik P Patel
- From the Department of Physiology, Augusta University, GA (J.E.S., S.S., V.C.B.); and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (H.Z., N.S., K.P.P.)
| |
Collapse
|
19
|
Reis WL, Biancardi VC, Zhou Y, Stern JE. A Functional Coupling Between Carbon Monoxide and Nitric Oxide Contributes to Increased Vasopressin Neuronal Activity in Heart Failure rats. Endocrinology 2016; 157:2052-66. [PMID: 26982634 PMCID: PMC4870874 DOI: 10.1210/en.2015-1958] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite the pathophysiological importance of neurohumoral activation in patients with heart failure (HF), the precise underlying mechanisms contributing to elevated vasopressin (VP) activation in HF remains unknown. Carbon monoxide (CO) is a gaseous neurotransmitter in the central nervous system that stimulates VP neuronal firing activity. Recently, we showed that the excitatory effect of CO on VP neurons in the hypothalamic paraventricular nucleus (PVN) was mediated by inhibition of nitric oxide (NO). Given that previous studies showed that VP neuronal activity is enhanced, whereas NO inhibitory signaling is blunted in HF rats, we tested whether an enhanced endogenous CO availability within the PVN contributes to elevated VP neuronal activity and blunted NO signaling in HF rats. We found that both haeme-oxygenase 1 (the CO-synthesizing enzyme) protein and mRNA expression levels were enhanced in the PVN of HF compared with sham rats (∼18% and ∼38%, respectively). We report that in sham rats, bath application of a CO donor (tricarbonyldichlororuthenium dimer) increased the firing activity of identified PVN VP neurons (P < .05), whereas inhibition of endogenous CO production (Tin-protoporphyrin IX [SnPP]) failed to affect neuronal activity. In HF rats, however, SnPP decreased VP activity (P < .05), an effect that was occluded by previous NO synathase blockade NG-nitro-larginine methyl ester. Finally, we found that SnPP increased the mean frequency of γ-aminobutyric acid inhibitory postsynaptic currents in VP neurons in HF (P < .05) but not sham rats. Our results support an enhanced endogenous CO excitatory signaling in VP neurons, which likely contributes to blunted NO and γ-aminobutyric acid inhibitory function in HF rats.
Collapse
Affiliation(s)
- Wagner L Reis
- Department of Physiology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912
| | - Vinicia C Biancardi
- Department of Physiology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912
| | - Yiqiang Zhou
- Department of Physiology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912
| | - Javier E Stern
- Department of Physiology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912
| |
Collapse
|
20
|
Rajapakse NW, Head GA, Kaye DM. Say NO to Obesity-Related Hypertension: Role of the L-Arginine-Nitric Oxide Pathway. Hypertension 2016; 67:813-9. [PMID: 27021014 DOI: 10.1161/hypertensionaha.116.06778] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Niwanthi W Rajapakse
- From the Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (N.W.R., G.A.H., D.M.K.); Department of Medicine, Monash University, Melbourne, VIC, Australia (D.M.K.); and Department of Physiology, Monash University, Melbourne, VIC, Australia (N.W.R.).
| | - Geoffrey A Head
- From the Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (N.W.R., G.A.H., D.M.K.); Department of Medicine, Monash University, Melbourne, VIC, Australia (D.M.K.); and Department of Physiology, Monash University, Melbourne, VIC, Australia (N.W.R.)
| | - David M Kaye
- From the Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (N.W.R., G.A.H., D.M.K.); Department of Medicine, Monash University, Melbourne, VIC, Australia (D.M.K.); and Department of Physiology, Monash University, Melbourne, VIC, Australia (N.W.R.)
| |
Collapse
|
21
|
Biancardi VC, Stranahan AM, Krause EG, de Kloet AD, Stern JE. Cross talk between AT1 receptors and Toll-like receptor 4 in microglia contributes to angiotensin II-derived ROS production in the hypothalamic paraventricular nucleus. Am J Physiol Heart Circ Physiol 2016; 310:H404-15. [PMID: 26637556 PMCID: PMC4796625 DOI: 10.1152/ajpheart.00247.2015] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 11/14/2015] [Indexed: 02/07/2023]
Abstract
ANG II is thought to increase sympathetic outflow by increasing oxidative stress and promoting local inflammation in the paraventricular nucleus (PVN) of the hypothalamus. However, the relative contributions of inflammation and oxidative stress to sympathetic drive remain poorly understood, and the underlying cellular and molecular targets have yet to be examined. ANG II has been shown to enhance Toll-like receptor (TLR)4-mediated signaling on microglia. Thus, in the present study, we aimed to determine whether ANG II-mediated activation of microglial TLR4 signaling is a key molecular target initiating local oxidative stress in the PVN. We found TLR4 and ANG II type 1 (AT1) receptor mRNA expression in hypothalamic microglia, providing molecular evidence for the potential interaction between these two receptors. In hypothalamic slices, ANG II induced microglial activation within the PVN (∼65% increase, P < 0.001), an effect that was blunted in the absence of functional TLR4. ANG II increased ROS production, as indicated by dihydroethidium fluorescence, within the PVN of rats and mice (P < 0.0001 in both cases), effects that were also dependent on the presence of functional TLR4. The microglial inhibitor minocycline attenuated ANG II-mediated ROS production, yet ANG II effects persisted in PVN single-minded 1-AT1a knockout mice, supporting the contribution of a non-neuronal source (likely microglia) to ANG II-driven ROS production in the PVN. Taken together, these results support functional interactions between AT1 receptors and TLR4 in mediating ANG II-dependent microglial activation and oxidative stress within the PVN. More broadly, our results support a functional interaction between the central renin-angiotensin system and innate immunity in the regulation of neurohumoral outflows from the PVN.
Collapse
Affiliation(s)
| | - Alexis M Stranahan
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| | - Eric G Krause
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Annette D de Kloet
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Javier E Stern
- Department of Physiology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| |
Collapse
|
22
|
Cauley E, Wang X, Dyavanapalli J, Sun K, Garrott K, Kuzmiak-Glancy S, Kay MW, Mendelowitz D. Neurotransmission to parasympathetic cardiac vagal neurons in the brain stem is altered with left ventricular hypertrophy-induced heart failure. Am J Physiol Heart Circ Physiol 2015; 309:H1281-7. [PMID: 26371169 DOI: 10.1152/ajpheart.00445.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/03/2015] [Indexed: 01/23/2023]
Abstract
Hypertension, cardiac hypertrophy, and heart failure (HF) are widespread and debilitating cardiovascular diseases that affect nearly 23 million people worldwide. A distinctive hallmark of these cardiovascular diseases is autonomic imbalance, with increased sympathetic activity and decreased parasympathetic vagal tone. Recent device-based approaches, such as implantable vagal stimulators that stimulate a multitude of visceral sensory and motor fibers in the vagus nerve, are being evaluated as new therapeutic approaches for these and other diseases. However, little is known about how parasympathetic activity to the heart is altered with these diseases, and this lack of knowledge is an obstacle in the goal of devising selective interventions that can target and selectively restore parasympathetic activity to the heart. To identify the changes that occur within the brain stem to diminish the parasympathetic cardiac activity, left ventricular hypertrophy was elicited in rats by aortic pressure overload using a transaortic constriction approach. Cardiac vagal neurons (CVNs) in the brain stem that generate parasympathetic activity to the heart were identified with a retrograde tracer and studied using patch-clamp electrophysiological recordings in vitro. Animals with left cardiac hypertrophy had diminished excitation of CVNs, which was mediated both by an augmented frequency of spontaneous inhibitory GABAergic neurotransmission (with no alteration of inhibitory glycinergic activity) as well as a diminished amplitude and frequency of excitatory neurotransmission to CVNs. Opportunities to alter these network pathways and neurotransmitter receptors provide future targets of intervention in the goal to restore parasympathetic activity and autonomic balance to the heart in cardiac hypertrophy and other cardiovascular diseases.
Collapse
Affiliation(s)
- Edmund Cauley
- Department of Pharmacology and Physiology, The George Washington University, Washington, District of Columbia; and
| | - Xin Wang
- Department of Pharmacology and Physiology, The George Washington University, Washington, District of Columbia; and
| | - Jhansi Dyavanapalli
- Department of Pharmacology and Physiology, The George Washington University, Washington, District of Columbia; and
| | - Ke Sun
- Department of Pharmacology and Physiology, The George Washington University, Washington, District of Columbia; and
| | - Kara Garrott
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - Sarah Kuzmiak-Glancy
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - Matthew W Kay
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - David Mendelowitz
- Department of Pharmacology and Physiology, The George Washington University, Washington, District of Columbia; and
| |
Collapse
|
23
|
de Souza Mecawi A, Ruginsk SG, Elias LLK, Varanda WA, Antunes‐Rodrigues J. Neuroendocrine Regulation of Hydromineral Homeostasis. Compr Physiol 2015; 5:1465-516. [DOI: 10.1002/cphy.c140031] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
24
|
Stern JE. Neuroendocrine-autonomic integration in the paraventricular nucleus: novel roles for dendritically released neuropeptides. J Neuroendocrinol 2015; 27:487-97. [PMID: 25546497 PMCID: PMC4447596 DOI: 10.1111/jne.12252] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 12/09/2014] [Accepted: 12/18/2014] [Indexed: 12/21/2022]
Abstract
Communication between pairs of neurones in the central nervous system typically involves classical 'hard-wired' synaptic transmission, characterised by high temporal and spatial precision. Over the last two decades, however, knowledge regarding the repertoire of communication modalities used in the brain has notably expanded to include less conventional forms, characterised by a diffuse and less temporally precise transfer of information. These forms are best suited to mediate communication among entire neuronal populations, now recognised to be a fundamental process in the brain for the generation of complex behaviours. In response to an osmotic stressor, the hypothalamic paraventricular nucleus (PVN) generates a multimodal homeostatic response that involves orchestrated neuroendocrine (i.e. systemic release of vasopressin) and autonomic (i.e. sympathetic outflow to the kidneys) components. The precise mechanisms that underlie interpopulation cross-talk between these two distinct neuronal populations, however, remain largely unknown. The present review summarises and discusses a series of recent studies that have identified the dendritic release of neuropeptides as a novel interpopulation signalling modality in the PVN. A current working model is described in which it is proposed that the activity-dependent dendritic release of vasopressin from neurosecretory neurones in the PVN acts in a diffusible manner to increase the activity of distant presympathetic neurones, resulting in an integrated sympathoexcitatory population response, particularly within the context of a hyperosmotic challenge. The cellular mechanism underlying this novel form of intercellular communication, as well as its physiological and pathophysiological implications, is discussed.
Collapse
Affiliation(s)
- J E Stern
- Department of Physiology, Georgia Regents University, Augusta, GA, USA
| |
Collapse
|
25
|
Rajapakse NW, Nanayakkara S, Kaye DM. Pathogenesis and treatment of the cardiorenal syndrome: Implications of L-arginine-nitric oxide pathway impairment. Pharmacol Ther 2015; 154:1-12. [PMID: 25989232 DOI: 10.1016/j.pharmthera.2015.05.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 01/11/2023]
Abstract
A highly complex interplay exists between the heart and kidney in the setting of both normal and abnormal physiology. In the context of heart failure, a pathophysiological condition termed the cardiorenal syndrome (CRS) exists whereby dysfunction in the heart or kidney can accelerate pathology in the other organ. The mechanisms that underpin CRS are complex, and include neuro-hormonal activation, oxidative stress and endothelial dysfunction. The endothelium plays a central role in the regulation of both cardiac and renal function, and as such impairments in endothelial function can lead to dysfunction of both these organs. In particular, reduced bioavailability of nitric oxide (NO) is a key pathophysiologic component of endothelial dysfunction. The synthesis of NO by the endothelium is critically dependent on the plasmalemmal transport of its substrate, L-arginine, via the cationic amino acid transporter-1 (CAT1). Impaired L-arginine-NO pathway activity has been demonstrated individually in heart and renal failure. Recent findings suggest abnormalities of the L-arginine-NO pathway also play a role in the pathogenesis of CRS and thus this pathway may represent a potential new target for the treatment of heart and renal failure.
Collapse
Affiliation(s)
- Niwanthi W Rajapakse
- Heart Failure Research Group, Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Physiology, Monash University, Melbourne, Australia.
| | | | - David M Kaye
- Heart Failure Research Group, Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne Australia; Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia
| |
Collapse
|
26
|
Reis WL, Biancardi VC, Son S, Antunes-Rodrigues J, Stern JE. Carbon monoxide and nitric oxide interactions in magnocellular neurosecretory neurones during water deprivation. J Neuroendocrinol 2015; 27:111-22. [PMID: 25494574 DOI: 10.1111/jne.12245] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 11/13/2014] [Accepted: 12/07/2014] [Indexed: 12/21/2022]
Abstract
Nitric oxide (NO) and carbon monoxide (CO) are diffusible gas messengers in the brain. Previously, we have shown their independent involvement in central fluid/electrolyte homeostasis control. In the present study, we investigated a possible functional interaction between NO/CO in the regulation of vasopressin (VP) and oxytocin (OT) magnocellular neurosecretory cells (MNCs) activity in euhydrated (EU) and dehydrated [48-h water-deprived (48WD)] rats. Using brain slices from EU and 48WD rats, we measured, by immunohistochemistry, the expression of neuronal NO synthase (nNOS, which synthesises NO) and haeme-oxygenase (HO-1, which synthesises CO) in the hypothalamic supraoptic nucleus (SON). In addition, we used patch-clamp electrophysiology to investigate whether regulation of SON MNC firing activity by endogenous CO was dependent on NO bioavailability and GABAergic inhibitory synaptic function. We found a proportion of OT and VP SON MNCs in EU rats to co-express both of HO-1 and nNOS (33.2 ± 2.9% and 15.3 ± 1.4%, respectively), which was increased in 48WD rats (55.5 ± 0.9% and 21.0 ± 1.7%, respectively, P < 0.05 for both). Inhibition of endogenous HO activity [chromium mesoporphyrin IX chloride (CrMP) 20 μm] induced MNC membrane hyperpolarisation and decreased firing activity, and these effects were blunted by previous blockade of endogenous NOS activity (l-NAME, 2 mm) or blockade of inhibitory GABA function [Picrotoxin (Sigma-Aldrich, St Louis, MO, USA), 50 μm]. No significant changes in SON NO bioavailability (4,5 diaminofluorescein diacetate fluorescence) were observed after CrMP treatment. Taken together, our results support a state-dependent functional inter-relationship between NO and CO in MNCs, in which CO acts as an excitatory gas molecule, whose effects are largely dependent on interactions with the inhibitory SON signals NO and GABA.
Collapse
Affiliation(s)
- W L Reis
- Department of Physiology, Georgia Regents University, Augusta, GA, USA; Department of Physiology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | | | | | | | | |
Collapse
|
27
|
Zhao YJ, Liu FQ, Xiu CH, Jiang J, Wang JH, Xu YS, Fu SY, Huang Q. The Effects of High Thoracic Epidural Anesthesia on Sympathetic Activity and Apoptosis in Experimentally Induced Congestive Heart Failure. J Cardiothorac Vasc Anesth 2014; 28:317-22. [DOI: 10.1053/j.jvca.2013.05.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Indexed: 01/17/2023]
|
28
|
Abstract
SIGNIFICANCE There is now compelling evidence to substantiate the notion that by depressing baroreflex regulation of blood pressure and augmenting central sympathetic outflow through their actions on the nucleus tractus solitarii (NTS) and rostral ventrolateral medulla (RVLM), brain stem nitric oxide synthase (NOS) and reactive oxygen species (ROS) are important contributing factors to neural mechanisms of hypertension. This review summarizes our contemporary views on the impact of NOS and ROS in the NTS and RVLM on neurogenic hypertension, and presents potential antihypertensive strategies that target brain stem NOS/ROS signaling. RECENT ADVANCES NO signaling in the brain stem may be pro- or antihypertensive depending on the NOS isoform that generates this gaseous moiety and the site of action. Elevation of the ROS level when its production overbalances its degradation in the NTS and RVLM underlies neurogenic hypertension. Interventional strategies with emphases on alleviating the adverse actions of these molecules on blood pressure regulation have been investigated. CRITICAL ISSUES The pathological roles of NOS in the RVLM and NTS in neural mechanisms of hypertension are highly complex. Likewise, multiple signaling pathways underlie the deleterious roles of brain-stem ROS in neurogenic hypertension. There are recent indications that interactions between brain stem ROS and NOS may play a contributory role. FUTURE DIRECTIONS Given the complicity of action mechanisms of brain-stem NOS and ROS in neural mechanisms of hypertension, additional studies are needed to identify the most crucial therapeutic target that is applicable not only in animal models but also in patients suffering from neurogenic hypertension.
Collapse
Affiliation(s)
- Samuel H H Chan
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital , Kaohsiung, Taiwan, Republic of China
| | | |
Collapse
|
29
|
Abstract
Statins (3-hydroxy-3-methylglutaryl-CoA reductase inhibitors) reduce plasma cholesterol and improve endothelium-dependent vasodilation, inflammation and oxidative stress. A ‘pleiotropic’ property of statins receiving less attention is their effect on the autonomic nervous system. Increased central sympathetic outflow and diminished cardiac vagal tone are disturbances characteristic of a range of cardiovascular conditions for which statins are now prescribed routinely to reduce cardiovascular events: following myocardial infarction, and in hypertension, chronic kidney disease, heart failure and diabetes. The purpose of the present review is to synthesize contemporary evidence that statins can improve autonomic circulatory regulation. In experimental preparations, high-dose lipophilic statins have been shown to reduce adrenergic outflow by attenuating oxidative stress in central brain regions involved in sympathetic and parasympathetic discharge induction and modulation. In patients with hypertension, chronic kidney disease and heart failure, lipophilic statins, such as simvastatin or atorvastatin, have been shown to reduce MNSA (muscle sympathetic nerve activity) by 12–30%. Reports concerning the effect of statin therapy on HRV (heart rate variability) are less consistent. Because of their implications for BP (blood pressure) control, insulin sensitivity, arrhythmogenesis and sudden cardiac death, these autonomic nervous system actions should be considered additional mechanisms by which statins lower cardiovascular risk.
Collapse
|
30
|
Sharpe AL, Calderon AS, Andrade MA, Cunningham JT, Mifflin SW, Toney GM. Chronic intermittent hypoxia increases sympathetic control of blood pressure: role of neuronal activity in the hypothalamic paraventricular nucleus. Am J Physiol Heart Circ Physiol 2013; 305:H1772-80. [PMID: 24097432 DOI: 10.1152/ajpheart.00592.2013] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Like humans with sleep apnea, rats exposed to chronic intermittent hypoxia (CIH) experience arterial hypoxemias and develop hypertension characterized by exaggerated sympathetic nerve activity (SNA). To gain insights into the poorly understood mechanisms that initiate sleep apnea/CIH-associated hypertension, experiments were performed in rats exposed to CIH for only 7 days. Compared with sham-treated normoxic control rats, CIH-exposed rats (n = 8 rats/group) had significantly increased hematocrit (P < 0.001) and mean arterial pressure (MAP; P < 0.05). Blockade of ganglionic transmission caused a significantly (P < 0.05) greater reduction of MAP in rats exposed to CIH than control rats (n = 8 rats/group), indicating a greater contribution of SNA in the support of MAP even at this early stage of CIH hypertension. Chemical inhibition of neuronal discharge in the hypothalamic paraventricular nucleus (PVN) (100 pmol muscimol) had no effect on renal SNA but reduced lumbar SNA (P < 0.005) and MAP (P < 0.05) more in CIH-exposed rats (n = 8) than control rats (n = 7), indicating that CIH increased the contribution of PVN neuronal activity in the support of lumbar SNA and MAP. Because CIH activates brain regions controlling body fluid homeostasis, the effects of internal carotid artery injection of hypertonic saline were tested and determined to increase lumbar SNA more (P < 0.05) in CIH-exposed rats than in control rats (n = 9 rats/group). We conclude that neurogenic mechanisms are activated early in the development of CIH hypertension such that elevated MAP relies on increased sympathetic tonus and ongoing PVN neuronal activity. The increased sensitivity of Na(+)/osmosensitive circuitry in CIH-exposed rats suggests that early neuroadaptive responses among body fluid regulatory neurons could contribute to the initiation of CIH hypertension.
Collapse
Affiliation(s)
- Amanda L Sharpe
- Department of Pharmaceutical Sciences, Feik School of Pharmacy, University of the Incarnate Word, San Antonio, Texas
| | | | | | | | | | | |
Collapse
|
31
|
Son SJ, Filosa JA, Potapenko ES, Biancardi VC, Zheng H, Patel KP, Tobin VA, Ludwig M, Stern JE. Dendritic peptide release mediates interpopulation crosstalk between neurosecretory and preautonomic networks. Neuron 2013; 78:1036-49. [PMID: 23791197 DOI: 10.1016/j.neuron.2013.04.025] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2013] [Indexed: 01/08/2023]
Abstract
Although communication between neurons is considered a function of the synapse, neurons also release neurotransmitter from their dendrites. We found that dendritic transmitter release coordinates activity across distinct neuronal populations to generate integrative homeostatic responses. We show that activity-dependent vasopressin release from hypothalamic neuroendocrine neurons in the paraventricular nucleus stimulates neighboring (~100 μm soma-to-soma) presympathetic neurons, resulting in a sympathoexcitatory population response. This interpopulation crosstalk was engaged by an NMDA-mediated increase in dendritic Ca(2+), influenced by vasopressin's ability to diffuse in the extracellular space, and involved activation of CAN channels at the target neurons. Furthermore, we demonstrate that this interpopulation crosstalk plays a pivotal role in the generation of a systemic, polymodal neurohumoral response to a hyperosmotic challenge. Because dendritic release is emerging as a widespread process, our results suggest that a similar mechanism could mediate interpopulation crosstalk in other brain systems, particularly those involved in generating complex behaviors.
Collapse
Affiliation(s)
- Sook Jin Son
- Department of Physiology, Georgia Regents University, Augusta, GA 30912, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Stern JE, Potapenko ES. Enhanced NMDA receptor-mediated intracellular calcium signaling in magnocellular neurosecretory neurons in heart failure rats. Am J Physiol Regul Integr Comp Physiol 2013; 305:R414-22. [PMID: 23785079 DOI: 10.1152/ajpregu.00160.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
An enhanced glutamate excitatory function within the hypothalamic supraoptic and paraventricluar nuclei is known to contribute to increased neurosecretory and presympathetic neuronal activity, and hence, neurohumoral activation, during heart failure (HF). Still, the precise mechanisms underlying enhanced glutamate-driven neuronal activity in HF remain to be elucidated. Here, we performed simultaneous electrophysiology and fast confocal Ca²⁺ imaging to determine whether altered N-methyl-d-aspartate (NMDA) receptor-mediated changes in intracellular Ca²⁺ levels (NMDA-ΔCa²⁺) occurred in hypothalamic magnocellular neurosecretory cells (MNCs) in HF rats. We found that activation of NMDA receptors resulted in a larger ΔCa²⁺ in MNCs from HF when compared with sham rats. The enhanced NMDA-ΔCa²⁺ was neither dependent on the magnitude of the NMDA-mediated current (voltage clamp) nor on the degree of membrane depolarization or firing activity evoked by NMDA (current clamp). Differently from NMDA receptor activation, firing activity evoked by direct membrane depolarization resulted in similar changes in intracellular Ca²⁺ in sham and HF rats. Taken together, our results support a relatively selective alteration of intracellular Ca²⁺ homeostasis and signaling following activation of NMDA receptors in MNCs during HF. The downstream functional consequences of such altered ΔCa²⁺ signaling during HF are discussed.
Collapse
Affiliation(s)
- Javier E Stern
- Department of Physiology, Georgia Regents University, Augusta, Georgia
| | | |
Collapse
|
33
|
Astrocytes modulate a postsynaptic NMDA-GABAA-receptor crosstalk in hypothalamic neurosecretory neurons. J Neurosci 2013; 33:631-40. [PMID: 23303942 DOI: 10.1523/jneurosci.3936-12.2013] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
A dynamic balance between the excitatory and inhibitory neurotransmitters glutamate and GABA is critical for maintaining proper neuronal activity in the brain. This balance is partly achieved via presynaptic interactions between glutamatergic and GABA(A)ergic synapses converging into the same targets. Here, we show that in hypothalamic magnocellular neurosecretory neurons (MNCs), a direct crosstalk between postsynaptic NMDA receptors (NMDARs) and GABA(A) receptors (GABA(A)Rs) contributes to the excitatory/inhibitory balance in this system. We found that activation of NMDARs by endogenous glutamate levels controlled by astrocyte glutamate transporters, evokes a transient and reversible potentiation of postsynaptic GABA(A)Rs. This inter-receptor crosstalk is calcium-dependent and involves a kinase-dependent phosphorylation mechanism, but does not require nitric oxide as an intermediary signal. Finally, we found the NMDAR-GABA(A)R crosstalk to be blunted in rats with heart failure, a pathological condition in which the hypothalamic glutamate-GABA balance is tipped toward an excitatory predominance. Together, our findings support a novel form of glutamate-GABA interactions in MNCs, which involves crosstalk between NMDA and GABA(A) postsynaptic receptors, whose strength is controlled by the activity of local astrocytes. We propose this inter-receptor crosstalk to act as a compensatory, counterbalancing mechanism to dampen glutamate-mediated overexcitation. Finally, we propose that an uncoupling between NMDARs and GABA(A)Rs may contribute to exacerbated neuronal activity and, consequently, sympathohumoral activation in such disease conditions as heart failure.
Collapse
|
34
|
Bidirectional neuro-glial signaling modalities in the hypothalamus: role in neurohumoral regulation. Auton Neurosci 2013; 175:51-60. [PMID: 23375650 DOI: 10.1016/j.autneu.2012.12.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 12/17/2012] [Accepted: 12/26/2012] [Indexed: 12/20/2022]
Abstract
Maintenance of bodily homeostasis requires concerted interactions between the neuroendocrine and the autonomic nervous systems, which generate adaptive neurohumoral outflows in response to a variety of sensory inputs. Moreover, an exacerbated neurohumoral activation is recognized to be a critical component in numerous disease conditions, including hypertension, heart failure, stress, and the metabolic syndrome. Thus, the study of neurohumoral regulation in the brain is of critical physiological and pathological relevance. Most of the work in the field over the last decades has been centered on elucidating neuronal mechanisms and pathways involved in neurohumoral control. More recently however, it has become increasingly clear that non-neuronal cell types, particularly astrocytes and microglial cells, actively participate in information processing in areas of the brain involved in neuroendocrine and autonomic control. Thus, in this work, we review recent advances in our understanding of neuro-glial interactions within the hypothalamic supraoptic and paraventricular nuclei, and their impact on neurohumoral integration in these nuclei. Major topics reviewed include anatomical and functional properties of the neuro-glial microenvironment, neuron-to-astrocyte signaling, gliotransmitters, and astrocyte regulation of signaling molecules in the extracellular space. We aimed in this review to highlight the importance of neuro-glial bidirectional interactions in information processing within major hypothalamic networks involved in neurohumoral integration.
Collapse
|
35
|
Potapenko ES, Biancardi VC, Zhou Y, Stern JE. Altered astrocyte glutamate transporter regulation of hypothalamic neurosecretory neurons in heart failure rats. Am J Physiol Regul Integr Comp Physiol 2012; 303:R291-300. [PMID: 22696576 DOI: 10.1152/ajpregu.00056.2012] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neurohumoral activation, which includes augmented plasma levels of the neurohormone vasopressin (VP), is a common finding in heart failure (HF) that contributes to morbidity and mortality in this disease. While an increased activation of magnocellular neurosecretory cells (MNCs) and enhanced glutamate function in HF is well documented, the precise underlying mechanisms remain to be elucidated. Here, we combined electrophysiology and protein measurements to determine whether altered glial glutamate transporter function and/or expression occurs in the hypothalamic supraoptic nucleus (SON) during HF. Patch-clamp recordings obtained from MNCs in brain slices show that pharmacological blockade of astrocyte glutamate transporter 1 (GLT1) function [500 μM dihydrokainate (DHK)], resulted in a persistent N-methyl-D-aspartate receptor (NMDAR)-mediated inward current (tonic I(NMDA)) in sham rats, an effect that was significantly smaller in MNCs from HF rats. In addition, we found a diminished GLT1 protein content in plasma membrane (but not cytosolic) fractions of SON punches in HF rats. Conversely, astrocyte GLAST expression was significantly higher in the SON of HF rats, while nonselective blockade of glutamate transport activity (100 μM TBOA) evoked an enhanced tonic I(NMDA) activation in HF rats. Steady-state activation of NMDARs by extracellular glutamate levels was diminished during HF. Taken together, these results support a shift in the relative expression and function of two major glial glutamate transporters (from GLT1 to GLAST predominance) during HF. This shift may act as a compensatory mechanism to preserve an adequate basal glutamate uptake level in the face of an enhanced glutamatergic afferent activity in HF rats.
Collapse
Affiliation(s)
- Evgeniy S Potapenko
- Department of Physiology, Georgia Health Sciences University, Augusta, 30912, USA
| | | | | | | |
Collapse
|
36
|
Gui L, LaGrange LP, Larson RA, Gu M, Zhu J, Chen QH. Role of small conductance calcium-activated potassium channels expressed in PVN in regulating sympathetic nerve activity and arterial blood pressure in rats. Am J Physiol Regul Integr Comp Physiol 2012; 303:R301-10. [PMID: 22647293 DOI: 10.1152/ajpregu.00114.2012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Small conductance Ca(2+)-activated K(+) (SK) channels regulate membrane properties of rostral ventrolateral medulla (RVLM) projecting hypothalamic paraventricular nucleus (PVN) neurons and inhibition of SK channels increases in vitro excitability. Here, we determined in vivo the role of PVN SK channels in regulating sympathetic nerve activity (SNA) and mean arterial pressure (MAP). In anesthetized rats, bilateral PVN microinjection of SK channel blocker with peptide apamin (0, 0.125, 1.25, 3.75, 12.5, and 25 pmol) increased splanchnic SNA (SSNA), renal SNA (RSNA), MAP, and heart rate (HR) in a dose-dependent manner. Maximum increases in SSNA, RSNA, MAP, and HR elicited by apamin (12.5 pmol, n = 7) were 330 ± 40% (P < 0.01), 271 ± 40% (P < 0.01), 29 ± 4 mmHg (P < 0.01), and 34 ± 9 beats/min (P < 0.01), respectively. PVN injection of the nonpeptide SK channel blocker UCL1684 (250 pmol, n = 7) significantly increased SSNA (P < 0.05), RSNA (P < 0.05), MAP (P < 0.05), and HR (P < 0.05). Neither apamin injected outside the PVN (12.5 pmol, n = 6) nor peripheral administration of the same dose of apamin (12.5 pmol, n = 5) evoked any significant changes in the recorded variables. PVN-injected SK channel enhancer 5,6-dichloro-1-ethyl-1,3-dihydro-2H-benzimidazol-2-one (DCEBIO, 5 nmol, n = 4) or N-cyclohexyl-N-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-4-pyrimidin]amine (CyPPA, 5 nmol, n = 6) did not significantly alter the SSNA, RSNA, MAP, and HR. Western blot and RT-PCR analysis of punched PVN tissue showed abundant expression of SK1-3 channels. We conclude that SK channels expressed in the PVN play an important role in the regulation of sympathetic outflow and cardiovascular function.
Collapse
Affiliation(s)
- Le Gui
- Department of Cardiology, Affiliated Hospital of Nantong University, Jiangsu, Peoples Republic of China
| | | | | | | | | | | |
Collapse
|
37
|
Reis WL, Biancardi VC, Son S, Antunes-Rodrigues J, Stern JE. Enhanced expression of heme oxygenase-1 and carbon monoxide excitatory effects in oxytocin and vasopressin neurones during water deprivation. J Neuroendocrinol 2012; 24:653-63. [PMID: 22060896 PMCID: PMC3314108 DOI: 10.1111/j.1365-2826.2011.02249.x] [Citation(s) in RCA: 22] [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] [Indexed: 11/30/2022]
Abstract
A growing body of evidence indiates that carbon monoxide (CO) acts as a gas neurotransmitter within the central nervous system. Although CO has been shown to affect neurohypophyseal hormone release in response to osmotic stimuli, the precise sources, targets and mechanisms underlying the actions of CO within the magnocellular neurosecretory system remain largely unknown. In the present study, we combined immunohistochemistry and patch-clamp electrophysiology to study the cellular distribution of the CO-synthase enzyme heme oxygenase type 1 (HO-1), as well as the actions of CO on oxytocin (OT) and vasopressin (VP) magnocellular neurosecretory cells (MNCs), in euhydrated (EU) and 48-h water-deprived rats (48WD). Our results show the expression of HO-1 immunoreactivity both in OT and VP neurones, as well as in a small proportion of astrocytes, both in supraoptic (SON) and paraventricular (PVN) nuclei. HO-1 expression, and its colocalisation with OT and VP neurones within the SON and PVN, was significantly enhanced in 48WD rats. Inhibition of HO activity with chromium mesoporphyrin IX chloride (CrMP; 20 μm) resulted in a slight membrane hyperpolarisation in SON neurones from EU rats, without significantly affecting their firing activity. In 48WD rats, on the other hand, CrMP resulted in a more robust membrane hyperpolarisation, significantly decreasing neuronal firing discharge. Taken together, our results indicate that magnocellular SON and PVN neurones express HO-1, and that CO acts as an excitatory gas neurotransmitter in this system. Moreover, we found that the expression and actions of CO were enhanced in water-deprived rats, suggesting that the state-dependent up-regulation of the HO-1/CO signalling pathway contributes to enhance MNCs firing activity during an osmotic challenge.
Collapse
Affiliation(s)
- W L Reis
- Department of Physiology, Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | | | | | |
Collapse
|