3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      α7 Nicotinic acetylcholine receptor mediates right ventricular fibrosis and diastolic dysfunction in pulmonary hypertension

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Right ventricular (RV) fibrosis is a key feature of maladaptive RV hypertrophy and dysfunction and is associated with poor outcomes in pulmonary hypertension (PH). However, mechanisms and therapeutic strategies to mitigate RV fibrosis remain unrealized. Previously, we identified that cardiac fibroblast α7 nicotinic acetylcholine receptor (α7 nAChR) drives smoking-induced RV fibrosis. Here, we sought to define the role of α7 nAChR in RV dysfunction and fibrosis in the settings of RV pressure overload as seen in PH. We show that RV tissue from PH patients has increased collagen content and ACh expression. Using an experimental rat model of PH, we demonstrate that RV fibrosis and dysfunction are associated with increases in ACh and α7 nAChR expression in the RV but not in the left ventricle (LV). In vitro studies show that α7 nAChR activation leads to an increase in adult ventricular fibroblast proliferation and collagen content mediated by a Ca 2+/epidermal growth factor receptor (EGFR) signaling mechanism. Pharmacological antagonism of nAChR decreases RV collagen content and improves RV function in the PH model. Furthermore, mice lacking α7 nAChR exhibit improved RV diastolic function and have lower RV collagen content in response to persistently increased RV afterload, compared with WT controls. These finding indicate that enhanced α7 nAChR signaling is an important mechanism underlying RV fibrosis and dysfunction, and targeted inhibition of α7 nAChR is a potentially novel therapeutic strategy in the setting of increased RV afterload.

          Abstract

          Related collections

          Most cited references69

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Haemodynamic definitions and updated clinical classification of pulmonary hypertension

          Since the 1st World Symposium on Pulmonary Hypertension (WSPH) in 1973, pulmonary hypertension (PH) has been arbitrarily defined as mean pulmonary arterial pressure (mPAP) ≥25 mmHg at rest, measured by right heart catheterisation. Recent data from normal subjects has shown that normal mPAP was 14.0±3.3 mmHg. Two standard deviations above this mean value would suggest mPAP >20 mmHg as above the upper limit of normal (above the 97.5th percentile). This definition is no longer arbitrary, but based on a scientific approach. However, this abnormal elevation of mPAP is not sufficient to define pulmonary vascular disease as it can be due to an increase in cardiac output or pulmonary arterial wedge pressure. Thus, this 6th WSPH Task Force proposes to include pulmonary vascular resistance ≥3 Wood Units in the definition of all forms of pre-capillary PH associated with mPAP >20 mmHg. Prospective trials are required to determine whether this PH population might benefit from specific management. Regarding clinical classification, the main Task Force changes were the inclusion in group 1 of a subgroup “pulmonary arterial hypertension (PAH) long-term responders to calcium channel blockers”, due to the specific prognostic and management of these patients, and a subgroup “PAH with overt features of venous/capillaries (pulmonary veno-occlusive disease/pulmonary capillary haemangiomatosis) involvement”, due to evidence suggesting a continuum between arterial, capillary and vein involvement in PAH.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Cardiac Fibrosis: The Fibroblast Awakens.

            Myocardial fibrosis is a significant global health problem associated with nearly all forms of heart disease. Cardiac fibroblasts comprise an essential cell type in the heart that is responsible for the homeostasis of the extracellular matrix; however, upon injury, these cells transform to a myofibroblast phenotype and contribute to cardiac fibrosis. This remodeling involves pathological changes that include chamber dilation, cardiomyocyte hypertrophy and apoptosis, and ultimately leads to the progression to heart failure. Despite the critical importance of fibrosis in cardiovascular disease, our limited understanding of the cardiac fibroblast impedes the development of potential therapies that effectively target this cell type and its pathological contribution to disease progression. This review summarizes current knowledge regarding the origins and roles of fibroblasts, mediators and signaling pathways known to influence fibroblast function after myocardial injury, as well as novel therapeutic strategies under investigation to attenuate cardiac fibrosis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Mammalian nicotinic acetylcholine receptors: from structure to function.

              The classical studies of nicotine by Langley at the turn of the 20th century introduced the concept of a "receptive substance," from which the idea of a "receptor" came to light. Subsequent studies aided by the Torpedo electric organ, a rich source of muscle-type nicotinic receptors (nAChRs), and the discovery of alpha-bungarotoxin, a snake toxin that binds pseudo-irreversibly to the muscle nAChR, resulted in the muscle nAChR being the best characterized ligand-gated ion channel hitherto. With the advancement of functional and genetic studies in the late 1980s, the existence of nAChRs in the mammalian brain was confirmed and the realization that the numerous nAChR subtypes contribute to the psychoactive properties of nicotine and other drugs of abuse and to the neuropathology of various diseases, including Alzheimer's, Parkinson's, and schizophrenia, has since emerged. This review provides a comprehensive overview of these findings and the more recent revelations of the impact that the rich diversity in function and expression of this receptor family has on neuronal and nonneuronal cells throughout the body. Despite these numerous developments, our understanding of the contributions of specific neuronal nAChR subtypes to the many facets of physiology throughout the body remains in its infancy.
                Bookmark

                Author and article information

                Contributors
                Journal
                JCI Insight
                JCI Insight
                JCI Insight
                JCI Insight
                American Society for Clinical Investigation
                2379-3708
                22 June 2021
                22 June 2021
                22 June 2021
                : 6
                : 12
                : e142945
                Affiliations
                [1 ]Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island, USA.
                [2 ]Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island, USA.
                [3 ]Biomedical & Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, USA.
                [4 ]Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA.
                [5 ]Department of Molecular Pharmacology, Physiology, and Biotechnology, Alpert Medical School of Brown University, Providence, Rhode Island, USA.
                [6 ]Cardiovascular Research Center, Lifespan Cardiovascular Institute, Rhode Island Hospital, Providence, Rhode Island, USA.
                Author notes
                Address correspondence to: Gaurav Choudhary, Providence VA Medical Center, 830 Chalkstone Avenue, Building 35, Providence, Rhode Island 02908, USA. Phone: 401.273.7100, ext. 12029; Email: gaurav_choudhary@ 123456brown.edu .

                Authorship note: AV, DDSGB, and AFN contributed equally to this work.

                Author information
                http://orcid.org/0000-0001-5691-781X
                http://orcid.org/0000-0002-2412-7669
                http://orcid.org/0000-0001-9916-9962
                http://orcid.org/0000-0002-7333-6555
                http://orcid.org/0000-0001-9343-5481
                Article
                142945
                10.1172/jci.insight.142945
                8262476
                33974567
                68f74966-795a-441e-876e-ee3c68404559
                © 2021 Vang et al.

                This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 10 August 2020
                : 6 May 2021
                Funding
                Funded by: National Heart, Lung, and Blood Institute, https://doi.org/10.13039/100000050;
                Award ID: R01HL128661,R01HL148727,R01HL139795,R01HL136757,R01HL114784,R01HL135236
                Funded by: National Institute of General Medical Sciences, https://doi.org/10.13039/100000057;
                Award ID: P20GM103652,P30GM1114750
                Funded by: U.S. Department of Veterans Affairs, https://doi.org/10.13039/100000738;
                Award ID: I01CX001892
                Funded by: American Heart Association, https://doi.org/10.13039/100000968;
                Award ID: 18CDA34110091
                Categories
                Research Article

                cardiology,pulmonology,fibrosis,heart failure,ion channels
                cardiology, pulmonology, fibrosis, heart failure, ion channels

                Comments

                Comment on this article