14
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Functional Amyloid Signaling via the Inflammasome, Necrosome, and Signalosome: New Therapeutic Targets in Heart Failure

      review-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

          As the most common cause of death and disability, globally, heart disease remains an incompletely understood enigma. A growing number of cardiac diseases are being characterized by the presence of misfolded proteins underlying their pathophysiology, including cardiac amyloidosis and dilated cardiomyopathy (DCM). At least nine precursor proteins have been implicated in the development of cardiac amyloidosis, most commonly caused by multiple myeloma light chain disease and disease-causing mutant or wildtype transthyretin (TTR). Similarly, aggregates with PSEN1 and COFILIN-2 have been identified in up to one-third of idiopathic DCM cases studied, indicating the potential predominance of misfolded proteins in heart failure. In this review, we present recent evidence linking misfolded proteins mechanistically with heart failure and present multiple lines of new therapeutic approaches that target the prevention of misfolded proteins in cardiac TTR amyloid disease. These include multiple small molecule pharmacological chaperones now in clinical trials designed specifically to support TTR folding by rational design, such as tafamidis, and chaperones previously developed for other purposes, such as doxycycline and tauroursodeoxycholic acid. Last, we present newly discovered non-pathological “functional” amyloid structures, such as the inflammasome and necrosome signaling complexes, which can be activated directly by amyloid. These may represent future targets to successfully attenuate amyloid-induced proteotoxicity in heart failure, as the inflammasome, for example, is being therapeutically inhibited experimentally in autoimmune disease. Together, these studies demonstrate multiple novel points in which new therapies may be used to primarily prevent misfolded proteins or to inhibit their downstream amyloid-mediated effectors, such as the inflammasome, to prevent proteotoxicity in heart failure.

          Related collections

          Most cited references67

          • Record: found
          • Abstract: found
          • Article: not found

          Inflammasomes in the CNS.

          Microglia and macrophages in the CNS contain multimolecular complexes termed inflammasomes. Inflammasomes function as intracellular sensors for infectious agents as well as for host-derived danger signals that are associated with neurological diseases, including meningitis, stroke and Alzheimer's disease. Assembly of an inflammasome activates caspase 1 and, subsequently, the proteolysis and release of the cytokines interleukin-1β and interleukin-18, as well as pyroptotic cell death. Since the discovery of inflammasomes in 2002, there has been burgeoning recognition of their complexities and functions. Here, we review the current understanding of the functions of different inflammasomes in the CNS and their roles in neurological diseases.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Protein folding and modification in the mammalian endoplasmic reticulum.

            Analysis of the human genome reveals that approximately a third of all open reading frames code for proteins that enter the endoplasmic reticulum (ER), demonstrating the importance of this organelle for global protein maturation. The path taken by a polypeptide through the secretory pathway starts with its translocation across or into the ER membrane. It then must fold and be modified correctly in the ER before being transported via the Golgi apparatus to the cell surface or another destination. Being physically segregated from the cytosol means that the ER lumen has a distinct folding environment. It contains much of the machinery for fulfilling the task of protein production, including complex pathways for folding, assembly, modification, quality control, and recycling. Importantly, the compartmentalization means that several modifications that do not occur in the cytosol, such as glycosylation and extensive disulfide bond formation, can occur to secreted proteins to enhance their stability before their exposure to the extracellular milieu. How these various machineries interact during the normal pathway of folding and protein secretion is the subject of this review.
              Bookmark
              • Record: found
              • Abstract: not found
              • Article: not found

              The systemic amyloidoses.

                Bookmark

                Author and article information

                Contributors
                Journal
                Front Cardiovasc Med
                Front Cardiovasc Med
                Front. Cardiovasc. Med.
                Frontiers in Cardiovascular Medicine
                Frontiers Media S.A.
                2297-055X
                19 May 2015
                2015
                : 2
                : 25
                Affiliations
                [1] 1McAllister Heart Institute, University of North Carolina , Chapel Hill, NC, USA
                [2] 2Department of Pharmacology, University of North Carolina , Chapel Hill, NC, USA
                [3] 3Section of Cardiology, Department of Medicine, The Institute for CardioScience, Johns Hopkins Medical Institutes , Baltimore, MD, USA
                [4] 4Department of Pathology and Laboratory Medicine, University of North Carolina , Chapel Hill, NC, USA
                Author notes

                Edited by: George W. Booz, University of Mississippi Medical Center, USA

                Reviewed by: Fadi G. Akar, Mount Sinai School of Medicine, USA; Ranganath Mamidi, Case Western Reserve University, USA

                *Correspondence: Monte S. Willis, Department of Pathology and Laboratory Medicine, McAllister Heart Institute, University of North Carolina, 111 Mason Farm Road, MBRB 2340B, Chapel Hill, NC 27599-7525, USA, monte_willis@ 123456med.unc.edu

                Specialty section: This article was submitted to Molecular Cardiology, a section of the journal Frontiers in Cardiovascular Medicine

                Article
                10.3389/fcvm.2015.00025
                4671334
                26664897
                7d932da6-ee42-475e-9503-a903339bd4ba
                Copyright © 2015 Parry, Melehani, Ranek and Willis.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 14 March 2015
                : 28 April 2015
                Page count
                Figures: 5, Tables: 4, Equations: 0, References: 105, Pages: 14, Words: 10744
                Funding
                Funded by: National Institutes of Health
                Award ID: R01HL104129
                Funded by: Leducq Foundation Transatlantic Networks of Excellence Program
                Funded by: Jefferson-Pilot Fellowship in Academic Medicine
                Categories
                Cardiovascular Medicine
                Reviews in Medicine

                functional amyloid,inflammasome,necrosome,signalosome,pharmacological chaperones

                Comments

                Comment on this article