1
|
Depes D, Mennander A, Immonen P, Mäkinen A, Huhtala H, Paavonen T, Kholová I. The autonomic nerves around the vein of Marshall: a postmortem study with clinical implications. APMIS 2024; 132:430-443. [PMID: 38468591 DOI: 10.1111/apm.13400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 01/31/2024] [Indexed: 03/13/2024]
Abstract
This study aims to analyze the vein of Marshall (VOM) in human autopsy hearts and its correlation with clinical data to elucidate the morphological substrates of atrial fibrillation (AF) and other cardiac diseases. Twenty-three adult autopsy hearts were studied, assessing autonomic nerves by immunohistochemistry with tyrosine hydroxylase (sympathetic nerves), choline acetyltransferase (parasympathetic nerves), growth-associated protein 43 (neural growth), and S100 (general neural marker) antibodies. Interstitial fibrosis was assessed by Masson trichrome staining. Measurements were conducted via morphometric software. The results were correlated with clinical data. Sympathetic innervation was abundant in all VOM-adjacent regions. Subjects with a history of AF, cardiovascular cause of death, and histologically verified myocardial infarction had increased sympathetic innervation and neural growth around the VOM at the mitral isthmus. Interstitial fibrosis increased with age and heart weight was associated with AF and cardiovascular cause of death. This study increases our understanding of the cardiac autonomic innervation in the VOM area in various diseases, offering implications for the development of new therapeutic approaches targeting the autonomic nervous system.
Collapse
Affiliation(s)
- Denis Depes
- Department of Pathology, Fimlab Laboratories, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ari Mennander
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Cardiothoracic Surgery, Tampere University Heart Hospital, Tampere, Finland
| | - Paavo Immonen
- Department of Pathology, Fimlab Laboratories, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Artturi Mäkinen
- Department of Pathology, Fimlab Laboratories, Tampere, Finland
| | - Heini Huhtala
- Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Timo Paavonen
- Department of Pathology, Fimlab Laboratories, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ivana Kholová
- Department of Pathology, Fimlab Laboratories, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
2
|
Kolesova H, Hrabalova P, Bohuslavova R, Abaffy P, Fabriciova V, Sedmera D, Pavlinkova G. Reprogramming of the developing heart by Hif1a-deficient sympathetic system and maternal diabetes exposure. Front Endocrinol (Lausanne) 2024; 15:1344074. [PMID: 38505753 PMCID: PMC10948485 DOI: 10.3389/fendo.2024.1344074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Introduction Maternal diabetes is a recognized risk factor for both short-term and long-term complications in offspring. Beyond the direct teratogenicity of maternal diabetes, the intrauterine environment can influence the offspring's cardiovascular health. Abnormalities in the cardiac sympathetic system are implicated in conditions such as sudden infant death syndrome, cardiac arrhythmic death, heart failure, and certain congenital heart defects in children from diabetic pregnancies. However, the mechanisms by which maternal diabetes affects the development of the cardiac sympathetic system and, consequently, heightens health risks and predisposes to cardiovascular disease remain poorly understood. Methods and results In the mouse model, we performed a comprehensive analysis of the combined impact of a Hif1a-deficient sympathetic system and the maternal diabetes environment on both heart development and the formation of the cardiac sympathetic system. The synergic negative effect of exposure to maternal diabetes and Hif1a deficiency resulted in the most pronounced deficit in cardiac sympathetic innervation and the development of the adrenal medulla. Abnormalities in the cardiac sympathetic system were accompanied by a smaller heart, reduced ventricular wall thickness, and dilated subepicardial veins and coronary arteries in the myocardium, along with anomalies in the branching and connections of the main coronary arteries. Transcriptional profiling by RNA sequencing (RNA-seq) revealed significant transcriptome changes in Hif1a-deficient sympathetic neurons, primarily associated with cell cycle regulation, proliferation, and mitosis, explaining the shrinkage of the sympathetic neuron population. Discussion Our data demonstrate that a failure to adequately activate the HIF-1α regulatory pathway, particularly in the context of maternal diabetes, may contribute to abnormalities in the cardiac sympathetic system. In conclusion, our findings indicate that the interplay between deficiencies in the cardiac sympathetic system and subtle structural alternations in the vasculature, microvasculature, and myocardium during heart development not only increases the risk of cardiovascular disease but also diminishes the adaptability to the stress associated with the transition to extrauterine life, thus increasing the risk of neonatal death.
Collapse
Affiliation(s)
- Hana Kolesova
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
- Department of Developmental Cardiology, Institute of Physiology Czech Academy of Sciences (CAS), Prague, Czechia
| | - Petra Hrabalova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences (CAS), BIOCEV, Vestec, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Romana Bohuslavova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences (CAS), BIOCEV, Vestec, Czechia
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology Czech Academy of Sciences (CAS), BIOCEV, Vestec, Czechia
| | - Valeria Fabriciova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences (CAS), BIOCEV, Vestec, Czechia
| | - David Sedmera
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
- Department of Developmental Cardiology, Institute of Physiology Czech Academy of Sciences (CAS), Prague, Czechia
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences (CAS), BIOCEV, Vestec, Czechia
| |
Collapse
|
3
|
Elia A, Fossati S. Autonomic nervous system and cardiac neuro-signaling pathway modulation in cardiovascular disorders and Alzheimer's disease. Front Physiol 2023; 14:1060666. [PMID: 36798942 PMCID: PMC9926972 DOI: 10.3389/fphys.2023.1060666] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/19/2023] [Indexed: 01/31/2023] Open
Abstract
The heart is a functional syncytium controlled by a delicate and sophisticated balance ensured by the tight coordination of its several cell subpopulations. Accordingly, cardiomyocytes together with the surrounding microenvironment participate in the heart tissue homeostasis. In the right atrium, the sinoatrial nodal cells regulate the cardiac impulse propagation through cardiomyocytes, thus ensuring the maintenance of the electric network in the heart tissue. Notably, the central nervous system (CNS) modulates the cardiac rhythm through the two limbs of the autonomic nervous system (ANS): the parasympathetic and sympathetic compartments. The autonomic nervous system exerts non-voluntary effects on different peripheral organs. The main neuromodulator of the Sympathetic Nervous System (SNS) is norepinephrine, while the principal neurotransmitter of the Parasympathetic Nervous System (PNS) is acetylcholine. Through these two main neurohormones, the ANS can gradually regulate cardiac, vascular, visceral, and glandular functions by turning on one of its two branches (adrenergic and/or cholinergic), which exert opposite effects on targeted organs. Besides these neuromodulators, the cardiac nervous system is ruled by specific neuropeptides (neurotrophic factors) that help to preserve innervation homeostasis through the myocardial layers (from epicardium to endocardium). Interestingly, the dysregulation of this neuro-signaling pathway may expose the cardiac tissue to severe disorders of different etiology and nature. Specifically, a maladaptive remodeling of the cardiac nervous system may culminate in a progressive loss of neurotrophins, thus leading to severe myocardial denervation, as observed in different cardiometabolic and neurodegenerative diseases (myocardial infarction, heart failure, Alzheimer's disease). This review analyzes the current knowledge on the pathophysiological processes involved in cardiac nervous system impairment from the perspectives of both cardiac disorders and a widely diffused and devastating neurodegenerative disorder, Alzheimer's disease, proposing a relationship between neurodegeneration, loss of neurotrophic factors, and cardiac nervous system impairment. This overview is conducive to a more comprehensive understanding of the process of cardiac neuro-signaling dysfunction, while bringing to light potential therapeutic scenarios to correct or delay the adverse cardiovascular remodeling, thus improving the cardiac prognosis and quality of life in patients with heart or neurodegenerative disorders.
Collapse
|
4
|
Depes D, Mennander A, Paavonen T, Kholová I. Autonomic Nerves in Myocardial Sleeves around Caval Veins: Potential Role in Cardiovascular Mortality? Cardiovasc Pathol 2022; 59:107426. [DOI: 10.1016/j.carpath.2022.107426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/24/2022] [Accepted: 03/26/2022] [Indexed: 11/15/2022] Open
|
5
|
Cui Y, Huang H, Ren W, Xu Y, Zha X, Zeng M, Gao Z, Tang S, Yang G, Huang Y, Xu F, Qian H, Zhou W, Ouyang C, Zhang L, Gao X, Zhang J, Wang J, Guo J, Xing C, Wei Y, Wang N. Parathyroidectomy Is Associated with Reversed Nondipping Heart Rate That Impacts Mortality in Chronic Kidney Disease Patients. Endocr Pract 2021; 28:148-158. [PMID: 33610808 DOI: 10.1016/j.eprac.2021.02.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Nondipping heart rate (HR), defined as a night/day HR ratio >0.90, has been associated with increased mortality in epidemiologic studies. However, its prognostic value in stage 5 chronic kidney disease (CKD5) patients and the effects of parathyroidectomy (PTX) on nondipping HR remain unknown. METHODS This case-control study of 162 healthy controls and 502 CKD5 patients was performed between 2011 and 2018, in which CKD5 patients were further divided into non-PTX (n = 186) and severe secondary hyperparathyroidism (SHPT) with PTX (n = 316) subgroups. Each participant underwent 24-hour Holter monitoring for HR ratio. Mortality was followed up in CKD5 patients (median time: 46.0 months). RESULTS The HR ratio in CKD5 patients was higher than in controls (0.92 ± 0.08 vs 0.81 ± 0.08, P <.001), associated with a 44% increase in mortality risk per 0.1 increment (hazard ratio, 1.44; 95% CI: 1.02-2.03; P =.04), and was positively related to serum intact parathyroid hormone levels (P <.001). PTX reversed nondipping HR in SHPT patients (n = 50, median time: 6.3 months, P <.001). Survival probabilities for PTX (n = 294) were better than non-PTX (n = 47) (hazard ratio, 0.31; 95% CI: 0.14-0.67; P <.01) in SHPT patients (serum intact parathyroid hormone >500.0 pg/mL). CONCLUSION CKD5 patients displayed a nondipping HR pattern, which is a prognostic marker of all-cause mortality. PTX for SHPT patients was associated with a reversal in nondipping HR ratio, which may mediate a better outcome.
Collapse
Affiliation(s)
- Ying Cui
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China; Department of Nephrology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Hui Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wenkai Ren
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Ying Xu
- CAM-SU Genomic Resource Center, Soochow University, Suzhou, China
| | - Xiaoming Zha
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Ming Zeng
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Zhanhui Gao
- Department of Nephrology, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Shaowen Tang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Guang Yang
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Yaoyu Huang
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Fangyan Xu
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Hanyang Qian
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Wenbin Zhou
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Chun Ouyang
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China; Department of Nephrology, Liyang Branch, Jiangsu Province Hospital, Liyang People's Hospital, Liyang, China
| | - Lina Zhang
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Xueyan Gao
- Department of General Medicine, Geriatric Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Zhang
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Jing Wang
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Jing Guo
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Changying Xing
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Yongyue Wei
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China; China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China.
| | - Ningning Wang
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China.
| |
Collapse
|
6
|
Burton RAB, Tomek J, Ambrosi CM, Larsen HE, Sharkey AR, Capel RA, Corbett AD, Bilton S, Klimas A, Stephens G, Cremer M, Bose SJ, Li D, Gallone G, Herring N, Mann EO, Kumar A, Kramer H, Entcheva E, Paterson DJ, Bub G. Optical Interrogation of Sympathetic Neuronal Effects on Macroscopic Cardiomyocyte Network Dynamics. iScience 2020; 23:101334. [PMID: 32674058 PMCID: PMC7363704 DOI: 10.1016/j.isci.2020.101334] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 05/12/2020] [Accepted: 06/26/2020] [Indexed: 12/21/2022] Open
Abstract
Cardiac stimulation via sympathetic neurons can potentially trigger arrhythmias. We present approaches to study neuron-cardiomyocyte interactions involving optogenetic selective probing and all-optical electrophysiology to measure activity in an automated fashion. Here we demonstrate the utility of optical interrogation of sympathetic neurons and their effects on macroscopic cardiomyocyte network dynamics to address research targets such as the effects of adrenergic stimulation via the release of neurotransmitters, the effect of neuronal numbers on cardiac behavior, and the applicability of optogenetics in mechanistic in vitro studies. As arrhythmias are emergent behaviors that involve the coordinated activity of millions of cells, we image at macroscopic scales to capture complex dynamics. We show that neurons can both decrease and increase wave stability and re-entrant activity in culture depending on their induced activity-a finding that may help us understand the often conflicting results seen in experimental and clinical studies.
Collapse
Affiliation(s)
- Rebecca-Ann B Burton
- University of Oxford, Department of Pharmacology, Mansfield Road, Oxford OX1 3QT, UK; University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK.
| | - Jakub Tomek
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Christina M Ambrosi
- The George Washington University, Department of Biomedical Engineering, Washington, DC 20052, USA
| | - Hege E Larsen
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Amy R Sharkey
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Rebecca A Capel
- University of Oxford, Department of Pharmacology, Mansfield Road, Oxford OX1 3QT, UK
| | | | - Samuel Bilton
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Aleksandra Klimas
- The George Washington University, Department of Biomedical Engineering, Washington, DC 20052, USA
| | - Guy Stephens
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Maegan Cremer
- University of Oxford, Department of Pharmacology, Mansfield Road, Oxford OX1 3QT, UK
| | - Samuel J Bose
- University of Oxford, Department of Pharmacology, Mansfield Road, Oxford OX1 3QT, UK
| | - Dan Li
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Giuseppe Gallone
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK; Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, Germany
| | - Neil Herring
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Edward O Mann
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Abhinav Kumar
- University of Oxford, Department of Biochemistry, Glycobiology Institute, Oxford, UK
| | - Holger Kramer
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Emilia Entcheva
- The George Washington University, Department of Biomedical Engineering, Washington, DC 20052, USA
| | - David J Paterson
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Gil Bub
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK; McGill University, Department of Physiology, McIntyre Medical Sciences Building, Room 1128, 3655 Promenade Sir William Osler, Montréal, QC H3G 1Y6, Canada.
| |
Collapse
|
7
|
Armando RG, Gómez DLM, Gomez DE. New drugs are not enough‑drug repositioning in oncology: An update. Int J Oncol 2020; 56:651-684. [PMID: 32124955 PMCID: PMC7010222 DOI: 10.3892/ijo.2020.4966] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 12/16/2019] [Indexed: 11/24/2022] Open
Abstract
Drug repositioning refers to the concept of discovering novel clinical benefits of drugs that are already known for use treating other diseases. The advantages of this are that several important drug characteristics are already established (including efficacy, pharmacokinetics, pharmacodynamics and toxicity), making the process of research for a putative drug quicker and less costly. Drug repositioning in oncology has received extensive focus. The present review summarizes the most prominent examples of drug repositioning for the treatment of cancer, taking into consideration their primary use, proposed anticancer mechanisms and current development status.
Collapse
Affiliation(s)
- Romina Gabriela Armando
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| | - Diego Luis Mengual Gómez
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| | - Daniel Eduardo Gomez
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| |
Collapse
|
8
|
Majumder A, Gupta B, Gupta AK. Pharmaceutically active compounds in aqueous environment: A status, toxicity and insights of remediation. ENVIRONMENTAL RESEARCH 2019; 176:108542. [PMID: 31387068 DOI: 10.1016/j.envres.2019.108542] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 05/22/2023]
Abstract
Pharmaceutically active compounds (PhACs) have pernicious effects on all kinds of life forms because of their toxicological effects and are found profoundly in various wastewater treatment plant influents, hospital effluents, and surface waters. The concentrations of different pharmaceuticals were found in alarmingly high concentrations in various parts of the globe, and it was also observed that the concentration of PhACs present in the water could be eventually related to the socio-economic conditions and climate of the region. Drinking water equivalent limit for each PhAC has been calculated and compared with the occurrence data from various continents. Since these compounds are recalcitrant towards conventional treatment methods, while advanced oxidation processes (AOPs) have shown better efficiency in degrading these PhACs. The performance of the AOPs have been evaluated based on percentage removal, time, and electrical energy consumed to degrade different classes of PhACs. Ozone based AOPs were found to be favorable because of their low treatment time, low cost, and high efficiency. However, complete degradation cannot be achieved by these processes, and various transformation products are formed, which may be more toxic than the parent compounds. The various transformation products formed from various PhACs during treatment have been highlighted. Significant stress has been given on the role of various process parameters, water matrix, oxidizing radicals, and the mechanism of degradation. Presence of organic compounds, nitrate, and phosphate usually hinders the degradation process, while chlorine and sulfate showed a positive effect. The role of individual oxidizing radicals, interfering ions, and pH demonstrated dissimilar effects on different groups of PhACs.
Collapse
Affiliation(s)
- Abhradeep Majumder
- School of Environmental Science and Engineering, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India.
| | - Bramha Gupta
- School of Water Resources, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India.
| | - Ashok Kumar Gupta
- Environmental Engineering Division, Department of Civil Engineering, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India.
| |
Collapse
|
9
|
Palagiri RDR, Chatterjee K, Jillella A, Hammond DA. A Case Report of Hypertensive Emergency and Intracranial Hemorrhage Due to Intracavernosal Phenylephrine. Hosp Pharm 2019; 54:186-189. [PMID: 31205330 DOI: 10.1177/0018578718778230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Intracavernosal injection of phenylephrine is a commonly used therapy for ischemic priapism and is typically well tolerated with few severe adverse side effects. We report a case of intracranial hemorrhage related to hypertensive emergency due to intracavernosal phenylephrine. Case Report: A 43-year-old Caucasian man with history of hypertension, diabetes mellitus type I, end-stage renal disease status post a combination kidney-pancreas transplant, and recurrent idiopathic priapism presented to emergency department with an episode of priapism. His home medications were lisinopril, metoprolol tartrate, mycophenolate mofetil, prednisone, sulfamethoxazole-trimethoprim, and tacrolimus. After local injection of 2 rounds (1 hour apart) of 100 µg phenylephrine into each corpus cavernosa, priapism resolved. Within 5 minutes, the patient had headaches, dyspnea, and excruciating chest pain. His blood pressure (BP) was noted to be 240/130 mm Hg but normalized spontaneously within few minutes. During this period, he developed new-onset right arm and leg weakness and found to have intracranial hemorrhage in the midbrain. Conclusion: A careful review for pharmacologic interactions should be performed prior to intracavernosal phenylephrine administration, and close monitoring should occur after its administration.
Collapse
Affiliation(s)
| | | | - Anusha Jillella
- University of Arkansas for Medical Sciences, Little Rock, USA
| | | |
Collapse
|
10
|
Pianca N, Di Bona A, Lazzeri E, Costantini I, Franzoso M, Prando V, Armani A, Rizzo S, Fedrigo M, Angelini A, Basso C, Pavone FS, Rubart M, Sacconi L, Zaglia T, Mongillo M. Cardiac sympathetic innervation network shapes the myocardium by locally controlling cardiomyocyte size through the cellular proteolytic machinery. J Physiol 2019; 597:3639-3656. [DOI: 10.1113/jp276200] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/21/2019] [Indexed: 01/07/2023] Open
Affiliation(s)
- Nicola Pianca
- Veneto Institute of Molecular Medicine Padova Italy
- Department of Biomedical SciencesUniversity of Padova Padova Italy
| | - Anna Di Bona
- Veneto Institute of Molecular Medicine Padova Italy
- Department of Cardiac, Thoracic, Vascular Sciences and Public HealthUniversity of Padova Padova Italy
| | - Erica Lazzeri
- European Laboratory for Non‐linear SpectroscopyUniversity of Florence Florence Italy
| | - Irene Costantini
- European Laboratory for Non‐linear SpectroscopyUniversity of Florence Florence Italy
- National Institute of Optics, National Research CouncilUniversity of Florence Florence Italy
| | - Mauro Franzoso
- Veneto Institute of Molecular Medicine Padova Italy
- Department of Biomedical SciencesUniversity of Padova Padova Italy
| | - Valentina Prando
- Veneto Institute of Molecular Medicine Padova Italy
- Department of Cardiac, Thoracic, Vascular Sciences and Public HealthUniversity of Padova Padova Italy
| | - Andrea Armani
- Veneto Institute of Molecular Medicine Padova Italy
- Department of Biomedical SciencesUniversity of Padova Padova Italy
| | - Stefania Rizzo
- Department of Cardiac, Thoracic, Vascular Sciences and Public HealthUniversity of Padova Padova Italy
| | - Marny Fedrigo
- Department of Cardiac, Thoracic, Vascular Sciences and Public HealthUniversity of Padova Padova Italy
| | - Annalisa Angelini
- Department of Cardiac, Thoracic, Vascular Sciences and Public HealthUniversity of Padova Padova Italy
| | - Cristina Basso
- Department of Cardiac, Thoracic, Vascular Sciences and Public HealthUniversity of Padova Padova Italy
| | - Francesco S. Pavone
- European Laboratory for Non‐linear SpectroscopyUniversity of Florence Florence Italy
- National Institute of Optics, National Research CouncilUniversity of Florence Florence Italy
- Department of Physics and AstronomyUniversity of Florence Florence Italy
| | - Michael Rubart
- Indiana University School of Medicine Indianapolis IN USA
| | - Leonardo Sacconi
- European Laboratory for Non‐linear SpectroscopyUniversity of Florence Florence Italy
- National Institute of Optics, National Research CouncilUniversity of Florence Florence Italy
| | - Tania Zaglia
- Veneto Institute of Molecular Medicine Padova Italy
- Department of Biomedical SciencesUniversity of Padova Padova Italy
- Department of Cardiac, Thoracic, Vascular Sciences and Public HealthUniversity of Padova Padova Italy
| | - Marco Mongillo
- Veneto Institute of Molecular Medicine Padova Italy
- Department of Biomedical SciencesUniversity of Padova Padova Italy
- CNR Institute of Neuroscience Padova Italy
| |
Collapse
|
11
|
Tomek J, Hao G, Tomková M, Lewis A, Carr C, Paterson DJ, Rodriguez B, Bub G, Herring N. β-Adrenergic Receptor Stimulation and Alternans in the Border Zone of a Healed Infarct: An ex vivo Study and Computational Investigation of Arrhythmogenesis. Front Physiol 2019; 10:350. [PMID: 30984029 PMCID: PMC6450465 DOI: 10.3389/fphys.2019.00350] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/14/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Following myocardial infarction (MI), the myocardium is prone to calcium-driven alternans, which typically precedes ventricular tachycardia and fibrillation. MI is also associated with remodeling of the sympathetic innervation in the infarct border zone, although how this influences arrhythmogenesis is controversial. We hypothesize that the border zone is most vulnerable to alternans, that β-adrenergic receptor stimulation can suppresses this, and investigate the consequences in terms of arrhythmogenic mechanisms. Methods and Results: Anterior MI was induced in Sprague-Dawley rats (n = 8) and allowed to heal over 2 months. This resulted in scar formation, significant (p < 0.05) dilation of the left ventricle, and reduction in ejection fraction compared to sham operated rats (n = 4) on 7 T cardiac magnetic resonance imaging. Dual voltage/calcium optical mapping of post-MI Langendorff perfused hearts (using RH-237 and Rhod2) demonstrated that the border zone was significantly more prone to alternans than the surrounding myocardium at longer cycle lengths, predisposing to spatially heterogeneous alternans. β-Adrenergic receptor stimulation with norepinephrine (1 μmol/L) attenuated alternans by 60 [52–65]% [interquartile range] and this was reversed with metoprolol (10 μmol/L, p = 0.008). These results could be reproduced by computer modeling of the border zone based on our knowledge of β-adrenergic receptor signaling pathways and their influence on intracellular calcium handling and ion channels. Simulations also demonstrated that β-adrenergic receptor stimulation in this specific region reduced the formation of conduction block and the probability of premature ventricular activation propagation. Conclusion: While high levels of overall cardiac sympathetic drive are a negative prognostic indicator of mortality following MI and during heart failure, β-adrenergic receptor stimulation in the infarct border zone reduced spatially heterogeneous alternans, and prevented conduction block and propagation of extrasystoles. This may help explain recent clinical imaging studies using meta-iodobenzylguanidine (MIBG) and 11C-meta-hydroxyephedrine positron emission tomography (PET) which demonstrate that border zone denervation is strongly associated with a high risk of future arrhythmia.
Collapse
Affiliation(s)
- Jakub Tomek
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Guoliang Hao
- Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Markéta Tomková
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Andrew Lewis
- Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Carolyn Carr
- Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - David J Paterson
- Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Blanca Rodriguez
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Gil Bub
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Neil Herring
- Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
12
|
Zaglia T, Mongillo M. Cardiac sympathetic innervation, from a different point of (re)view. J Physiol 2018; 595:3919-3930. [PMID: 28240352 DOI: 10.1113/jp273120] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/23/2017] [Indexed: 12/25/2022] Open
Abstract
The audience of basic and clinical scientists is familiar with the notion that the sympathetic nervous system controls heart function during stresses. However, evidence indicates that the neurogenic control of the heart spans from the maintenance of housekeeping functions in resting conditions to the recruitment of maximal performance, in the fight-or-flight responses, across a whole range of intermediate states. To perform such sophisticated functions, sympathetic ganglia integrate both peripheral and central inputs, and transmit information to the heart via 'motor' neurons, directly interacting with target cardiomyocytes. To date, the dynamics and mode of communication between these two cell types, which determine how neuronal information is adequately translated into the wide spectrum of cardiac responses, are still blurry. By combining the anatomical and structural information brought to light by recent imaging technologies and the functional evidence in cellular systems, we focus on the interface between neurons and cardiomyocytes, and advocate the existence of a specific 'neuro-cardiac junction', where sympathetic neurotransmission occurs in a 'quasi-synaptic' way. The properties of such junctional-type communication fit well with those of the physiological responses elicited by the cardiac sympathetic nervous system, and explain its ability to tune heart function with precision, specificity and elevated temporal resolution.
Collapse
Affiliation(s)
- Tania Zaglia
- Department of Cardiac, Thoracic and Vascular Sciences, via Giustiniani 2, 35128, University of Padova, Padova, Italy.,Department of Biomedical Sciences, via Ugo Bassi 58/B, 35131, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, via G.Orus, 2, 35129, Padova, Italy
| | - Marco Mongillo
- Department of Biomedical Sciences, via Ugo Bassi 58/B, 35131, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, via G.Orus, 2, 35129, Padova, Italy.,CNR institute of Neurosciences, viale Colombo 3, 35133, Padova, Italy
| |
Collapse
|
13
|
The neuropeptide galanin promotes an anti-thrombotic phenotype on endocardial endothelial cells from heart failure patients. Auton Neurosci 2017; 206:35-42. [PMID: 28720509 DOI: 10.1016/j.autneu.2017.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 06/06/2017] [Accepted: 07/09/2017] [Indexed: 02/07/2023]
Abstract
Thromboembolic complications are a significant cause of mortality and re-hospitalization in heart failure (HF) patients. One source of thrombi is the ventricular endocardial surface that becomes increasingly pro-thrombotic as HF progresses. Anticoagulation comes with bleeding risks so identifying therapeutic agents for improving cardiac endothelial health are of critical clinical importance. Endocardial endothelial cells are closely apposed to cardiac sympathetic nerves. In HF, cardiac sympathetic nerves are dysregulated and promote disease progression. Whether endocardial endothelial health and function is impacted by sympathetic dysregulation in HF is unknown. Also unexplored is the impact of neuropeptides, such as galanin and neuropeptide Y (NPY), co-released from sympathetic nerve terminals, on endothelial health. In this study we examined the effect of sympathetic nerve-released neurotransmitters and neuropeptides on the procoagulant phenotype of cultured human endocardial endothelial cells from HF patients. As a functional readout of procoagulant state we examined thrombin-mediated von Willebrand factor (vWF) extrusion and multimer expression. We demonstrate that vWF extrusion and multimer expression is promoted by thrombin, that isoproterenol (a beta-adrenergic receptor agonist) augments this effect, whereas co-treatment with the beta-blockers propranolol and carvedilol blocks this effect. We also show that vWF extrusion and multimer expression is attenuated by treatment with the neuropeptide galanin, but not with NPY. Our results are consistent with a protective role of beta-blockers and galanin on endocardial endothelial health in heart failure. Improving endothelial health through galanin therapy is a future clinical application of this study.
Collapse
|
14
|
Tomek J, Rodriguez B, Bub G, Heijman J. β-Adrenergic receptor stimulation inhibits proarrhythmic alternans in postinfarction border zone cardiomyocytes: a computational analysis. Am J Physiol Heart Circ Physiol 2017; 313:H338-H353. [PMID: 28550171 PMCID: PMC5582914 DOI: 10.1152/ajpheart.00094.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/12/2017] [Accepted: 05/23/2017] [Indexed: 01/09/2023]
Abstract
We integrated, for the first time, postmyocardial infarction electrical and autonomic remodeling in a detailed, validated computer model of β-adrenergic stimulation in ventricular cardiomyocytes. Here, we show that β-adrenergic stimulation inhibits alternans and provide novel insights into underlying mechanisms, adding to a recent controversy about pro-/antiarrhythmic effects of postmyocardial infarction hyperinnervation. The border zone (BZ) of the viable myocardium adjacent to an infarct undergoes extensive autonomic and electrical remodeling and is prone to repolarization alternans-induced cardiac arrhythmias. BZ remodeling processes may promote or inhibit Ca2+ and/or repolarization alternans and may differentially affect ventricular arrhythmogenesis. Here, we used a detailed computational model of the canine ventricular cardiomyocyte to study the determinants of alternans in the BZ and their regulation by β-adrenergic receptor (β-AR) stimulation. The BZ model developed Ca2+ transient alternans at slower pacing cycle lengths than the control model, suggesting that the BZ may promote spatially heterogeneous alternans formation in an infarcted heart. β-AR stimulation abolished alternans. By evaluating all combinations of downstream β-AR stimulation targets, we identified both direct (via ryanodine receptor channels) and indirect [via sarcoplasmic reticulum (SR) Ca2+ load] modulation of SR Ca2+ release as critical determinants of Ca2+ transient alternans. These findings were confirmed in a human ventricular cardiomyocyte model. Cell-to-cell coupling indirectly modulated the likelihood of alternans by affecting the action potential upstroke, reducing the trigger for SR Ca2+ release in one-dimensional strand simulations. However, β-AR stimulation inhibited alternans in both single and multicellular simulations. Taken together, these data highlight a potential antiarrhythmic role of sympathetic hyperinnervation in the BZ by reducing the likelihood of alternans and provide new insights into the underlying mechanisms controlling Ca2+ transient and repolarization alternans. NEW & NOTEWORTHY We integrated, for the first time, postmyocardial infarction electrical and autonomic remodeling in a detailed, validated computer model of β-adrenergic stimulation in ventricular cardiomyocytes. Here, we show that β-adrenergic stimulation inhibits alternans and provide novel insights into underlying mechanisms, adding to a recent controversy about pro-/antiarrhythmic effects of postmyocardial infarction hyperinnervation. Listen to this article’s corresponding podcast at http://ajpheart.podbean.com/e/%CE%B2-ar-stimulation-and-alternans-in-border-zone-cardiomyocytes/.
Collapse
Affiliation(s)
- Jakub Tomek
- Life Sciences Interface Doctoral Training Centre, University of Oxford, Oxford, United Kingdom; .,Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Blanca Rodriguez
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Gil Bub
- Department of Physiology, McGill University, Montreal, Quebec, Canada; and
| | - Jordi Heijman
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
15
|
Leitermann RJ, Rostkowski AB, Urban JH. Neuropeptide Y input to the rat basolateral amygdala complex and modulation by conditioned fear. J Comp Neurol 2016; 524:2418-39. [PMID: 26779765 DOI: 10.1002/cne.23960] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 12/30/2015] [Accepted: 01/05/2016] [Indexed: 12/14/2022]
Abstract
Within the basolateral amygdaloid complex (BLA), neuropeptide Y (NPY) buffers against protracted anxiety and fear. Although the importance of NPY's actions in the BLA is well documented, little is known about the source(s) of NPY fibers to this region. The current studies identified sources of NPY projections to the BLA by using a combination of anatomical and neurochemical approaches. NPY innervation of the BLA was assessed in rats by examining the degree of NPY coexpression within interneurons or catecholaminergic fibers with somatostatin and tyrosine hydroxylase (TH) or dopamine β-hydroxylase (DβH), respectively. Numerous NPY(+) /somatostatin(+) and NPY(+) /somatostatin(-) fibers were observed, suggesting at least two populations of NPY fibers within the BLA. No colocalization was noted between NPY and TH or DβH immunoreactivities. Additionally, Fluorogold (FG) retrograde tracing with immunohistochemistry was used to identify the precise origin of NPY projections to the BLA. FG(+) /NPY(+) cells were identified within the amygdalostriatal transition area (AStr) and stria terminalis and scattered throughout the bed nucleus of the stria terminalis. The subpopulation of NPY neurons in the AStr also coexpressed somatostatin. Subjecting animals to a conditioned fear paradigm increased NPY gene expression within the AStr, whereas no changes were observed within the BLA or stria terminalis. Overall, these studies identified limbic regions associated with stress circuits providing NPY input to the BLA and demonstrated that a unique NPY projection from the AStr may participate in the regulation of conditioned fear. J. Comp. Neurol. 524:2418-2439, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Randy J Leitermann
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Amanda B Rostkowski
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Janice H Urban
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| |
Collapse
|
16
|
Current hypotheses regarding the pathophysiology behind the takotsubo syndrome. Int J Cardiol 2014; 177:771-9. [DOI: 10.1016/j.ijcard.2014.10.156] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/23/2014] [Accepted: 10/24/2014] [Indexed: 01/15/2023]
|
17
|
Vitamin D deficiency leads to sensory and sympathetic denervation of the rat synovium. Neuroscience 2014; 279:77-93. [PMID: 25193239 DOI: 10.1016/j.neuroscience.2014.08.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 08/15/2014] [Accepted: 08/21/2014] [Indexed: 12/20/2022]
Abstract
Vitamin D deficiency is associated with increased susceptibility to inflammatory arthritis. Sensory and sympathetic synovial nerves are critical to the development of inflammatory arthritis and spontaneously degenerate in the early phases of disease. These nerves contain vitamin D receptors and vitamin D influences nerve growth and neurotrophin expression. We therefore examined the density of synovial nerves and neurotrophin-containing cells in vitamin D-deficient rats. Seven-week-old Sprague-Dawley rats were fed either control or vitamin D-deficient diets for 4weeks. Knee synovium sections extending from the patella to the meniscus were immunostained for total nerves, myelinated and unmyelinated nerves, sympathetic nerves, peptidergic and non-peptidergic sensory nerves, and neurotrophins and immune cell markers. In control rats, intimal innervation by unmyelinated sensory fibers was denser than subintimal innervation. In contrast, sympathetic innervation was confined to the subintima. Many sensory axons contained markers for both peptidergic and non-peptidergic nerves. Nerve growth factor (NGF) was primarily expressed by intimal CD163-negative type B synoviocytes, while neurturin, a ligand selective for non-peptidergic sensory neurons, was expressed by synovial mast cells. In vitamin D-deficient rats, there were significant reductions in sensory nerves in the intima and sympathetic nerves in the subintima. While there was no significant change in NGF-immunoreactivity, the number of neurturin-expressing mast cells was significantly reduced in the intima, suggesting that intimal reductions in sensory nerves may be related to reductions in neurturin. Vitamin D deficiency therefore may increase susceptibility to inflammatory arthritis by depleting sensory and sympathetic synovial nerves as a result of reduced synovial neurotrophin content.
Collapse
|
18
|
Berg T. β1-Blockers Lower Norepinephrine Release by Inhibiting Presynaptic, Facilitating β1-Adrenoceptors in Normotensive and Hypertensive Rats. Front Neurol 2014; 5:51. [PMID: 24795691 PMCID: PMC3997042 DOI: 10.3389/fneur.2014.00051] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 03/28/2014] [Indexed: 01/23/2023] Open
Abstract
Peripheral norepinephrine release is facilitated by presynaptic β-adrenoceptors, believed to involve the β2-subtype exclusively. However, β1-selective blockers are the most commonly used β-blockers in hypertension. Here the author tested the hypothesis that β1AR may function as presynaptic, release-facilitating auto-receptors. Since β1AR-blockers are injected during myocardial infarction, their influence on the cardiovascular response to acute norepinephrine release was also studied. By a newly established method, using tyramine-stimulated release through the norepinephrine transporter (NET), presynaptic control of catecholamine release was studied in normotensive and spontaneously hypertensive rats. β1AR-selective antagonists (CGP20712A, atenolol, metoprolol) reduced norepinephrine overflow to plasma equally efficient as β2AR-selective (ICI-118551) and β1+2AR (nadolol) antagonists in both strains. Neither antagonist lowered epinephrine secretion. Atenolol, which does not cross the blood–brain barrier, reduced norepinephrine overflow after adrenalectomy (AdrX), AdrX + ganglion blockade, losartan, or nephrectomy. Atenolol and metoprolol reduced resting cardiac work load. During tyramine-stimulated norepinephrine release, they had little effect on work load, and increased the transient rise in total peripheral vascular resistance, particularly atenolol when combined with losartan. In conclusion, β1AR, like β2AR, stimulated norepinephrine but not epinephrine release, independent of adrenal catecholamines, ganglion transmission, or renal renin release/angiotensin AT1 receptor activation. β1AR therefore functioned as a peripheral, presynaptic, facilitating auto-receptor. Like tyramine, hypoxia may induce NET-mediated release. Augmented tyramine-induced vasoconstriction, as observed after injection of β1AR-blocker, particularly atenolol combined with losartan, may hamper organ perfusion, and may have clinical relevance in hypoxic conditions such as myocardial infarction.
Collapse
Affiliation(s)
- Torill Berg
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo , Oslo , Norway
| |
Collapse
|
19
|
Day JS, O'Neill E, Cawley C, Aretz NK, Kilroy D, Gibney SM, Harkin A, Connor TJ. Noradrenaline acting on astrocytic β2-adrenoceptors induces neurite outgrowth in primary cortical neurons. Neuropharmacology 2014; 77:234-48. [DOI: 10.1016/j.neuropharm.2013.09.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 09/17/2013] [Accepted: 09/30/2013] [Indexed: 12/23/2022]
|
20
|
Freeman K, Tao W, Sun H, Soonpaa MH, Rubart M. In situ three-dimensional reconstruction of mouse heart sympathetic innervation by two-photon excitation fluorescence imaging. J Neurosci Methods 2014; 221:48-61. [PMID: 24056230 PMCID: PMC3858460 DOI: 10.1016/j.jneumeth.2013.09.005] [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] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 09/06/2013] [Accepted: 09/08/2013] [Indexed: 12/26/2022]
Abstract
BACKGROUND Sympathetic nerve wiring in the mammalian heart has remained largely unexplored. Resolving the wiring diagram of the cardiac sympathetic network would help establish the structural underpinnings of neurocardiac coupling. NEW METHOD We used two-photon excitation fluorescence microscopy, combined with a computer-assisted 3-D tracking algorithm, to map the local sympathetic circuits in living hearts from adult transgenic mice expressing enhanced green fluorescent protein (EGFP) in peripheral adrenergic neurons. RESULTS Quantitative co-localization analyses confirmed that the intramyocardial EGFP distribution recapitulated the anatomy of the sympathetic arbor. In the left ventricular subepicardium of the uninjured heart, the sympathetic network was composed of multiple subarbors, exhibiting variable branching and looping topology. Axonal branches did not overlap with each other within their respective parental subarbor nor with neurites of annexed subarbors. The sympathetic network in the border zone of a 2-week-old myocardial infarction was characterized by substantive rewiring, which included spatially heterogeneous loss and gain of sympathetic fibers and formation of multiple, predominately nested, axon loops of widely variable circumference and geometry. COMPARISON WITH EXISTING METHODS In contrast to mechanical tissue sectioning methods that may involve deformation of tissue and uncertainty in registration across sections, our approach preserves continuity of structure, which allows tracing of neurites over distances, and thus enables derivation of the three-dimensional and topological morphology of cardiac sympathetic nerves. CONCLUSIONS Our assay should be of general utility to unravel the mechanisms governing sympathetic axon spacing during development and disease.
Collapse
Affiliation(s)
- Kim Freeman
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, Indianapolis, IN 46202
| | - Wen Tao
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, Indianapolis, IN 46202
| | - Hongli Sun
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, Indianapolis, IN 46202
| | - Mark H. Soonpaa
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, Indianapolis, IN 46202
| | - Michael Rubart
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, Indianapolis, IN 46202
| |
Collapse
|
21
|
Hasan W, Smith PG. Decreased adrenoceptor stimulation in heart failure rats reduces NGF expression by cardiac parasympathetic neurons. Auton Neurosci 2013; 181:13-20. [PMID: 24332566 DOI: 10.1016/j.autneu.2013.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/11/2013] [Accepted: 11/26/2013] [Indexed: 10/25/2022]
Abstract
Postganglionic cardiac parasympathetic and sympathetic nerves are physically proximate in atrial cardiac tissue allowing reciprocal inhibition of neurotransmitter release, depending on demands from central cardiovascular centers or reflex pathways. Parasympathetic cardiac ganglion (CG) neurons synthesize and release the sympathetic neurotrophin nerve growth factor (NGF), which may serve to maintain these close connections. In this study we investigated whether NGF synthesis by CG neurons is altered in heart failure, and whether norepinephrine from sympathetic neurons promotes NGF synthesis. NGF and proNGF immunoreactivity in CG neurons in heart failure rats following chronic coronary artery ligation was investigated. NGF immunoreactivity was decreased significantly in heart failure rats compared to sham-operated animals, whereas proNGF expression was unchanged. Changes in neurochemistry of CG neurons included attenuated expression of the cholinergic marker vesicular acetylcholine transporter, and increased expression of the neuropeptide vasoactive intestinal polypeptide. To further investigate norepinephrine's role in promoting NGF synthesis, we cultured CG neurons treated with adrenergic receptor (AR) agonists. An 82% increase in NGF mRNA levels was detected after 1h of isoproterenol (β-AR agonist) treatment, which increased an additional 22% at 24h. Antagonist treatment blocked isoproterenol-induced increases in NGF transcripts. In contrast, the α-AR agonist phenylephrine did not alter NGF mRNA expression. These results are consistent with β-AR mediated maintenance of NGF synthesis in CG neurons. In heart failure, a decrease in NGF synthesis by CG neurons may potentially contribute to reduced connections with adjacent sympathetic nerves.
Collapse
Affiliation(s)
- Wohaib Hasan
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, United States.
| | - Peter G Smith
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, United States
| |
Collapse
|
22
|
Abstract
Autonomic cardiac neurons have a common origin in the neural crest but undergo distinct developmental differentiation as they mature toward their adult phenotype. Progenitor cells respond to repulsive cues during migration, followed by differentiation cues from paracrine sources that promote neurochemistry and differentiation. When autonomic axons start to innervate cardiac tissue, neurotrophic factors from vascular tissue are essential for maintenance of neurons before they reach their targets, upon which target-derived trophic factors take over final maturation, synaptic strength and postnatal survival. Although target-derived neurotrophins have a central role to play in development, alternative sources of neurotrophins may also modulate innervation. Both developing and adult sympathetic neurons express proNGF, and adult parasympathetic cardiac ganglion neurons also synthesize and release NGF. The physiological function of these “non-classical” cardiac sources of neurotrophins remains to be determined, especially in relation to autocrine/paracrine sustenance during development.
Cardiac autonomic nerves are closely spatially associated in cardiac plexuses, ganglia and pacemaker regions and so are sensitive to release of neurotransmitter, neuropeptides and trophic factors from adjacent nerves. As such, in many cardiac pathologies, it is an imbalance within the two arms of the autonomic system that is critical for disease progression. Although this crosstalk between sympathetic and parasympathetic nerves has been well established for adult nerves, it is unclear whether a degree of paracrine regulation occurs across the autonomic limbs during development. Aberrant nerve remodeling is a common occurrence in many adult cardiovascular pathologies, and the mechanisms regulating outgrowth or denervation are disparate. However, autonomic neurons display considerable plasticity in this regard with neurotrophins and inflammatory cytokines having a central regulatory function, including in possible neurotransmitter changes. Certainly, neurotrophins and cytokines regulate transcriptional factors in adult autonomic neurons that have vital differentiation roles in development. Particularly for parasympathetic cardiac ganglion neurons, additional examinations of developmental regulatory mechanisms will potentially aid in understanding attenuated parasympathetic function in a number of conditions, including heart failure.
Collapse
Affiliation(s)
- Wohaib Hasan
- Knight Cardiovascular Institute; Oregon Health & Science University; Portland, OR USA
| |
Collapse
|
23
|
Bone morphogenetic protein 4 mediates estrogen-regulated sensory axon plasticity in the adult female reproductive tract. J Neurosci 2013; 33:1050-61a. [PMID: 23325243 DOI: 10.1523/jneurosci.1704-12.2013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Peripheral axons are structurally plastic even in the adult, and altered axon density is implicated in many disorders and pain syndromes. However, mechanisms responsible for peripheral axon remodeling are poorly understood. Physiological plasticity is characteristic of the female reproductive tract: vaginal sensory innervation density is low under high estrogen conditions, such as term pregnancy, whereas density is high in low-estrogen conditions, such as menopause. We exploited this system in rats to identify factors responsible for adult peripheral neuroplasticity. Calcitonin gene-related peptide-immunoreactive sensory innervation is distributed primarily within the vaginal submucosa. Submucosal smooth muscle cells express bone morphogenetic protein 4 (BMP4). With low estrogen, BMP4 expression was elevated, indicating negative regulation by this hormone. Vaginal smooth muscle cells induced robust neurite outgrowth by cocultured dorsal root ganglion neurons, which was prevented by neutralizing BMP4 with noggin or anti-BMP4. Estrogen also prevented axon outgrowth, and this was reversed by exogenous BMP4. Nuclear accumulation of phosphorylated Smad1, a primary transcription factor for BMP4 signaling, was high in vagina-projecting sensory neurons after ovariectomy and reduced by estrogen. BMP4 regulation of innervation was confirmed in vivo using lentiviral transduction to overexpress BMP4 in an estrogen-independent manner. Submucosal regions with high virally induced BMP4 expression had high innervation density despite elevated estrogen. These findings show that BMP4, an important factor in early nervous system development and regeneration after injury, is a critical mediator of adult physiological plasticity as well. Altered BMP4 expression may therefore contribute to sensory hyperinnervation, a hallmark of several pain disorders, including vulvodynia.
Collapse
|
24
|
Li F, Ohtani A, Senzaki K, Shiga T. Receptor-dependent regulation of dendrite formation of noradrenaline and dopamine in non-GABAergic cerebral cortical neurons. Dev Neurobiol 2012; 73:370-83. [PMID: 23135899 DOI: 10.1002/dneu.22065] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 09/18/2012] [Accepted: 10/31/2012] [Indexed: 12/12/2022]
Abstract
The present study characterized the receptor-dependent regulation of dendrite formation of noradrenaline (NA) and dopamine (DA) in cultured neurons obtained from embryonic day 16 rat cerebral cortex. Morphological diversity of cortical dendrites was analyzed on various features: dendrite initiation, dendrite outgrowth, and dendrite branching. Using a combination of immunocytochemical markers of dendrites and GABAergic neurons, we focused on the dendrite morphology of non-GABAergic neurons. Our results showed that (1) NA inhibited the dendrite branching, (2) β adrenergic receptor (β-AR) agonist inhibited the dendrite initiation, while promoted the dendrite outgrowth, (3) β1-AR and β2-AR were present in all the cultured neurons, and both agonists inhibited the dendrite initiation, while only β1-AR agonist induced the dendrite branching; (4) DA inhibited the dendrite outgrowth, (5) D1 receptor agonist inhibited the dendrite initiation, while promoted the dendrite branching. In conclusion, this study compared the effects of NA, DA and their receptors and showed that NA and DA regulate different features on the dendrite formation of non-GABAergic cortical neurons, depending on the receptors.
Collapse
Affiliation(s)
- Fei Li
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8577, Ibaraki, Japan
| | | | | | | |
Collapse
|
25
|
Newsome TE, Zewe JW, Olesik SV. Electrospun nanofibrous solid-phase microextraction coatings for preconcentration of pharmaceuticals prior to liquid chromatographic separations. J Chromatogr A 2012; 1262:1-7. [DOI: 10.1016/j.chroma.2012.08.046] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Revised: 08/12/2012] [Accepted: 08/13/2012] [Indexed: 10/28/2022]
|
26
|
Klatt S, Fassold A, Straub RH. Sympathetic nerve fiber repulsion: testing norepinephrine, dopamine, and 17β-estradiol in a primary murine sympathetic neurite outgrowth assay. Ann N Y Acad Sci 2012; 1261:26-33. [PMID: 22823390 DOI: 10.1111/j.1749-6632.2012.06628.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Loss of sympathetic nerve fibers (SNFs) occurs in inflamed tissue; and select semaphorins, upregulated during inflammation, stimulate repulsion/loss of SNFs. However, it is unknown whether other factors released locally in inflamed tissue, such as norepinephrine, dopamine, and 17β-estradiol, are also repellent. In order to study the effects of hormones on SNF repulsion, an SNF outgrowth assay was used. The repellent activity of semaphorins 3C was weaker than of semaphorin 3F. Tumor necrosis factor α (TNF-α) repelled nerve fibers with moderate to strong effects (from 0-100% repulsion). High concentrations of dopamine and norepinephrine (10(-6) M) induced weak but significant nerve fiber repulsion (up to 20%). Norepinephrine at 10(-8) M was comparable with 10(-6) M at inducing nerve fiber outgrowth. Stimulation with low concentrations of 17β-estradiol (10(-10) M, but not 10(-8) M) repelled SNFs. These results demonstrate that not only specific axon guidance molecules, such as semaphorins 3F and 3C, but also hormonal factors and TNF-α influence SNF repulsion and outgrowth.
Collapse
Affiliation(s)
- Susanne Klatt
- Laboratory of Experimental Rheumatology and Neuroendocrine Immunology, Division of Rheumatology, Department of Internal Medicine I, University Hospital, Regensburg, Regensburg Germany
| | | | | |
Collapse
|
27
|
Systemic and topical hormone therapies reduce vaginal innervation density in postmenopausal women. Menopause 2012; 19:630-5. [PMID: 22205148 DOI: 10.1097/gme.0b013e31823b8983] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Menopause is often accompanied by vaginal discomfort including burning, itching, dryness, and spontaneous or provoked pain. Although the direct effects of estrogen withdrawal on vaginal cells are implicated, surgical menopause in rodents causes autonomic and sensory nerves to proliferate, suggesting that indirect effects mediated by changes in vaginal innervation may contribute. We assessed whether postmenopausal women display hormone-dependent changes in vaginal innervation. METHODS Vaginal biopsies from 20 postmenopausal women undergoing surgery for stress urinary incontinence and pelvic organ prolapse were fixed and immunostained for the pan-neuronal marker protein gene product 9.5, sympathetic marker tyrosine hydroxylase, parasympathetic marker vasoactive intestinal polypeptide, and sensory nociceptor marker calcitonin gene-related peptide. Innervation density was measured as an apparent percentage of the section area occupied by immunofluorescent axons. Specimens were grouped according to whether participants received systemic hormone therapy (HT), topical (vaginal) HT, or no HT. RESULTS Women not receiving HT showed relatively high levels of total innervation, with most axons expressing tyrosine hydroxylase or vasoactive intestinal polypeptide immunoreactivity. In women receiving systemic HT, overall innervation was reduced, as were presumptive parasympathetic, sympathetic, and sensory axon populations. Topical HT elicited more dramatic reductions in innervation than in systemic HT. CONCLUSIONS Hormone therapy reduces autonomic and sensory vaginal innervation density, which may, in part, contribute to relief from vaginal discomfort. Moreover, topical therapy is more effective than systemic therapy, which may help explain the greater improvement reported with topical compared with systemic HT.
Collapse
|
28
|
Hardwick JC, Southerland EM, Girasole AE, Ryan SE, Negrotto S, Ardell JL. Remodeling of intrinsic cardiac neurons: effects of β-adrenergic receptor blockade in guinea pig models of chronic heart disease. Am J Physiol Regul Integr Comp Physiol 2012; 303:R950-8. [PMID: 22933026 DOI: 10.1152/ajpregu.00223.2012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chronic heart disease induces remodeling of cardiac tissue and associated neuronal components. Treatment of chronic heart disease often involves pharmacological blockade of adrenergic receptors. This study examined the specific changes in neuronal sensitivity of guinea pig intrinsic cardiac neurons to autonomic modulators in animals with chronic cardiac disease, in the presence or absence of adrenergic blockage. Myocardial infarction (MI) was produced by ligature of the coronary artery and associated vein on the dorsal surface of the heart. Pressure overload (PO) was induced by a banding of the descending dorsal aorta (∼20% constriction). Animals were allowed to recover for 2 wk and then implanted with an osmotic pump (Alzet) containing either timolol (2 mg·kg(-1)·day(-1)) or vehicle, for a total of 6-7 wk of drug treatment. At termination, intracellular recordings from individual neurons in whole mounts of the cardiac plexus were used to assess changes in physiological responses. Timolol treatment did not inhibit the increased sensitivity to norepinephrine seen in both MI and PO animals, but it did inhibit the stimulatory effects of angiotensin II on the norepinephrine-induced increases in neuronal excitability. Timolol treatment also inhibited the increase in synaptically evoked action potentials observed in PO animals with stimulation of fiber tract bundles. These results demonstrate that β-adrenergic blockade can inhibit specific aspects of remodeling within the intrinsic cardiac plexus. In addition, this effect was preferentially observed with active cardiac disease states, indicating that the β-receptors were more influential on remodeling during dynamic disease progression.
Collapse
Affiliation(s)
- Jean C Hardwick
- Department of Biology, Ithaca College, Ithaca, NY 14850, USA.
| | | | | | | | | | | |
Collapse
|
29
|
Lewin A, Lasseter KC, Dong F, Whalen JC. Nebivolol withdrawal results in blood pressure returning toward pretreatment levels, but without rebound symptoms: phase IV randomized trial. ACTA ACUST UNITED AC 2012; 6:228-36. [DOI: 10.1016/j.jash.2012.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 01/30/2012] [Accepted: 02/14/2012] [Indexed: 01/06/2023]
|
30
|
Bateman RJ, Boychuk CR, Philbin KE, Mendelowitz D. β adrenergic receptor modulation of neurotransmission to cardiac vagal neurons in the nucleus ambiguus. Neuroscience 2012; 210:58-66. [PMID: 22425752 DOI: 10.1016/j.neuroscience.2012.02.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 02/15/2012] [Accepted: 02/16/2012] [Indexed: 12/14/2022]
Abstract
β-adrenergic receptors are a class of G protein-coupled receptors that have essential roles in regulating heart rate, blood pressure, and other cardiorespiratory functions. Although the role of β adrenergic receptors in the peripheral nervous system is well characterized, very little is known about their role in the central nervous system despite being localized in many brain regions involved in autonomic activity and regulation. Since parasympathetic activity to the heart is dominated by cardiac vagal neurons (CVNs) originating in the nucleus ambiguus (NA), β adrenergic receptors localized in the NA represent a potential target for modulating cardiac vagal activity and heart rate. This study tests the hypothesis that activation of β adrenergic receptors alters the membrane properties and synaptic neurotransmission to CVNs. CVNs were identified in brainstem slices, and membrane properties and synaptic events were recorded using the whole-cell voltage-clamp technique. The nonselective β agonist isoproterenol significantly decreased inhibitory GABAergic and glycinergic as well as excitatory glutamatergic neurotransmission to CVNs. In addition, the β(1)-selective receptor agonist dobutamine, but not β(2) or β(3) receptor agonists, significantly decreased inhibitory GABAergic and glycinergic and excitatory glutamatergic neurotransmission to CVNs. These decreases in neurotransmission to CVNs persisted in the presence of tetrodotoxin (TTX). These results provide a mechanism by which activation of adrenergic receptors in the brainstem can alter parasympathetic activity to the heart. Likely physiological roles for this adrenergic receptor activation are coordination of parasympathetic-sympathetic activity and β receptor-mediated increases in heart rate upon arousal.
Collapse
Affiliation(s)
- R J Bateman
- Department of Pharmacology and Physiology, The George Washington University, 2300 Eye Street NW, Washington, DC 20037, USA
| | | | | | | |
Collapse
|
31
|
Vitamin D deficiency promotes skeletal muscle hypersensitivity and sensory hyperinnervation. J Neurosci 2011; 31:13728-38. [PMID: 21957236 DOI: 10.1523/jneurosci.3637-11.2011] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Musculoskeletal pain affects nearly half of all adults, most of whom are vitamin D deficient. Previous findings demonstrated that putative nociceptors ("pain-sensing" nerves) express vitamin D receptors (VDRs), suggesting responsiveness to 1,25-dihydroxyvitamin D. In the present study, rats receiving vitamin D-deficient diets for 2-4 weeks showed mechanical deep muscle hypersensitivity, but not cutaneous hypersensitivity. Muscle hypersensitivity was accompanied by balance deficits and occurred before onset of overt muscle or bone pathology. Hypersensitivity was not due to hypocalcemia and was actually accelerated by increased dietary calcium. Morphometry of skeletal muscle innervation showed increased numbers of presumptive nociceptor axons (peripherin-positive axons containing calcitonin gene-related peptide), without changes in sympathetic or skeletal muscle motor innervation. Similarly, there was no change in epidermal innervation. In culture, sensory neurons displayed enriched VDR expression in growth cones, and sprouting was regulated by VDR-mediated rapid response signaling pathways, while sympathetic outgrowth was not affected by different concentrations of 1,25-dihydroxyvitamin D. These findings indicate that vitamin D deficiency can lead to selective alterations in target innervation, resulting in presumptive nociceptor hyperinnervation of skeletal muscle, which in turn is likely to contribute to muscular hypersensitivity and pain.
Collapse
|
32
|
Corti F, Olson KE, Marcus AJ, Levi R. The expression level of ecto-NTP diphosphohydrolase1/CD39 modulates exocytotic and ischemic release of neurotransmitters in a cellular model of sympathetic neurons. J Pharmacol Exp Ther 2011; 337:524-32. [PMID: 21325440 PMCID: PMC3083107 DOI: 10.1124/jpet.111.179994] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Accepted: 02/16/2011] [Indexed: 11/22/2022] Open
Abstract
Once released, norepinephrine is removed from cardiac synapses via reuptake into sympathetic nerves, whereas transmitter ATP is catabolized by ecto-NTP diphosphohydrolase 1 (E-NTPDase1)/CD39, an ecto-ATPase. Because ATP is known to modulate neurotransmitter release at prejunctional sites, we questioned whether this action may be ultimately controlled by the expression of E-NTPDase1/CD39 at sympathetic nerve terminals. Accordingly, we silenced E-NTPDase1/CD39 expression in nerve growth factor-differentiated PC12 cells, a cellular model of sympathetic neuron, in which dopamine is the predominant catecholamine. We report that E-NTPDase1/CD39 deletion markedly increases depolarization-induced exocytosis of ATP and dopamine and increases ATP-induced dopamine release. Moreover, overexpression of E-NTPDase1/CD39 resulted in enhanced removal of exogenous ATP, a marked decrease in exocytosis of ATP and dopamine, and a large decrease in ATP-induced dopamine release. Administration of a recombinant form of E-NTPDase1/CD39 reproduced the effects of E-NTPDase1/CD39 overexpression. Exposure of PC12 cells to simulated ischemia elicited a release of ATP and dopamine that was markedly increased in E-NTPDase1/CD39-silenced cells and decreased in E-NTPDase1/CD39-overexpressing cells. Therefore, transmitter ATP acts in an autocrine manner to promote its own release and that of dopamine, an action that is controlled by the level of E-NTPDase1/CD39 expression. Because ATP availability greatly increases in myocardial ischemia, recombinant E-NTPDase1/CD39 therapeutically used may offer a novel approach to reduce cardiac dysfunctions caused by excessive catecholamine release.
Collapse
Affiliation(s)
- Federico Corti
- Department of Pharmacology, Weill Cornell Medical College, 1300 York Ave., New York, NY 10065-4896, USA
| | | | | | | |
Collapse
|
33
|
Daly CJ, McGrath JC. Previously unsuspected widespread cellular and tissue distribution of β-adrenoceptors and its relevance to drug action. Trends Pharmacol Sci 2011; 32:219-26. [PMID: 21429599 DOI: 10.1016/j.tips.2011.02.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 02/11/2011] [Accepted: 02/11/2011] [Indexed: 01/05/2023]
Abstract
The discovery of β-adrenoceptors in previously unsuspected cell types is contributing to the rethinking of new drug targets. Recent developments in β-adrenoceptor pharmacology might have excited and surprised James Black, given his interest in developing drugs based on the selective manipulation of receptors to alter physiological responses. β-adrenoceptors continue to generate surprises at molecular and pharmacological levels that often require knowledge of receptor location to interpret. In this review, we emphasize the use of fluorescent ligands as the most selective means of demonstrating receptor localization. Fluorescent ligand binding in live tissues can provide quantitative pharmacological data, under carefully controlled conditions, relevant to other signalling parameters. Consideration of the role of β-adrenoceptors in many cell types (previously ignored) is needed to understand the actions of drugs at β-adrenoceptors throughout the body, particularly in the lung epithelium, vascular endothelium, immune cells and other 'structural' and 'restorative' cell types.
Collapse
Affiliation(s)
- C J Daly
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, West Medical Building, University of Glasgow, Glasgow G12 8QQ, UK.
| | | |
Collapse
|