1
|
Wang X, Lang Z, Yan Z, Xu J, Zhang J, Jiao L, Zhang H. Dilated cardiomyopathy: from genes and molecules to potential treatments. Mol Cell Biochem 2025:10.1007/s11010-025-05269-0. [PMID: 40155570 DOI: 10.1007/s11010-025-05269-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
Dilated cardiomyopathy is a myocardial condition marked by the enlargement of the heart's ventricular chambers and the gradual decline in systolic function, frequently resulting in congestive heart failure. Dilated cardiomyopathy has obvious familial characteristics, and mutations in related pathogenic genes can account for about 50% of patients with dilated cardiomyopathy. The most common genes related to dilated cardiomyopathy include TTN, LMNA, MYH7, etc. With more and more research on these genes, it will undoubtedly provide more potential targets and therapeutic pathways for the treatment of dilated cardiomyopathy. In addition, myocardial inflammation, myocardial metabolism abnormalities and cardiomyocyte apoptosis all have an important impact on the pathogenesis of dilated cardiomyopathy. Approximately half of sudden deaths among children and adolescents, along with the majority of patients undergoing heart transplantation, stem from cardiomyopathy. Therefore, precise and prompt clinical diagnosis holds paramount importance. Currently, diagnosis primarily hinges on the patient's medical background and imaging tests, with the significance of genetic testing steadily gaining prominence. The primary treatment for dilated cardiomyopathy remains heart transplantation. However, the scarcity of donors and the risk of severe immune rejection underscore the pressing need for novel therapies. Presently, research is actively exploring preclinical treatments like stem cell therapy as potential solutions.
Collapse
Affiliation(s)
- Xiumei Wang
- Department of Anesthesiology and Operating Theater, The First Hospital of Lanzhou University, Lanzhou, 730000, The People's Republic of China
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Zekun Lang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Zeyi Yan
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Jing Xu
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Jinyuan Zhang
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Lianhang Jiao
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Haijun Zhang
- Department of Anesthesiology and Operating Theater, The First Hospital of Lanzhou University, Lanzhou, 730000, The People's Republic of China.
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China.
| |
Collapse
|
2
|
Qu HQ, Hakonarson H. BAG3's dual roles in Parkinson's disease and cardiomyopathy: benefit or liability? Acta Neuropathol 2024; 148:71. [PMID: 39585448 DOI: 10.1007/s00401-024-02837-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Affiliation(s)
- Hui-Qi Qu
- The Center for Applied Genomics, Children's Hospital of Philadelphia, 3615 Civic Center Blvd, Abramson Building, Philadelphia, PA, 19104, USA
| | - Hakon Hakonarson
- The Center for Applied Genomics, Children's Hospital of Philadelphia, 3615 Civic Center Blvd, Abramson Building, Philadelphia, PA, 19104, USA.
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
- Division of Pulmonary Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland.
| |
Collapse
|
3
|
Qu HQ, Wang JF, Rosa-Campos A, Hakonarson H, Feldman AM. The Role of BAG3 Protein Interactions in Cardiomyopathies. Int J Mol Sci 2024; 25:11308. [PMID: 39457090 PMCID: PMC11605229 DOI: 10.3390/ijms252011308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Bcl-2-associated athanogene 3 (BAG3) plays an important function in cellular protein quality control (PQC) maintaining proteome stability. Mutations in the BAG3 gene result in cardiomyopathies. Due to its roles in cardiomyopathies and the complexity of BAG3-protein interactions, it is important to understand these protein interactions given the importance of the multifunctional cochaperone BAG3 in cardiomyocytes, using an in vitro cardiomyocyte model. The experimental assay was conducted using high pressure liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) in the human AC16 cardiomyocyte cell line with BioID technology. Proteins with BAG3-interaction were identified in all the 28 hallmark gene sets enriched in idiopathic cardiomyopathies and/or ischemic disease. Among the 24 hallmark gene sets enriched in both idiopathic cardiomyopathies and ischemic disease, 15 gene sets had at least 3 proteins with BAG3-interaction. This study highlights BAG3 protein interactions, unveiling the key gene sets affected in cardiomyopathies, which help to explain the molecular mechanisms of the cardioprotective effects of BAG3. In addition, this study also highlighted the complexity of proteins with BAG3 interactions, implying unwanted effects of BAG3.
Collapse
Affiliation(s)
- Hui-Qi Qu
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Ju-Fang Wang
- Department of Medicine, Division of Cardiology, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (J.-F.W.); (A.M.F.)
| | - Alexandre Rosa-Campos
- Proteomics Facility, Sanford-Burnham-Presby Medical Discovery Institute, La Jolla, CA 92037, USA;
| | - Hakon Hakonarson
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- Division of Human Genetics, Division of Pulmonary Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Faculty of Medicine, University of Iceland, 102 Reykjavík, Iceland
| | - Arthur M. Feldman
- Department of Medicine, Division of Cardiology, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (J.-F.W.); (A.M.F.)
| |
Collapse
|
4
|
Maroli G, Schänzer A, Günther S, Garcia-Gonzalez C, Rupp S, Schlierbach H, Chen Y, Graumann J, Wietelmann A, Kim J, Braun T. Inhibition of autophagy prevents cardiac dysfunction at early stages of cardiomyopathy in Bag3-deficient hearts. J Mol Cell Cardiol 2024; 193:53-66. [PMID: 38838815 DOI: 10.1016/j.yjmcc.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
The HSP70 co-chaperone BAG3 targets unfolded proteins to degradation via chaperone assisted selective autophagy (CASA), thereby playing pivotal roles in the proteostasis of adult cardiomyocytes (CMs). However, the complex functions of BAG3 for regulating autophagy in cardiac disease are not completely understood. Here, we demonstrate that conditional inactivation of Bag3 in murine CMs leads to age-dependent dysregulation of autophagy, associated with progressive cardiomyopathy. Surprisingly, Bag3-deficient CMs show increased canonical and non-canonical autophagic flux in the juvenile period when first signs of cardiac dysfunction appear, but reduced autophagy during later stages of the disease. Juvenile Bag3-deficient CMs are characterized by decreased levels of soluble proteins involved in synchronous contraction of the heart, including the gap junction protein Connexin 43 (CX43). Reiterative administration of chloroquine (CQ), an inhibitor of canonical and non-canonical autophagy, but not inactivation of Atg5, restores normal concentrations of soluble cardiac proteins in juvenile Bag3-deficient CMs without an increase of detergent-insoluble proteins, leading to complete recovery of early-stage cardiac dysfunction in Bag3-deficient mice. We conclude that loss of Bag3 in CMs leads to age-dependent differences in autophagy and cardiac dysfunction. Increased non-canonical autophagic flux in the juvenile period removes soluble proteins involved in cardiac contraction, leading to early-stage cardiomyopathy, which is prevented by reiterative CQ treatment.
Collapse
Affiliation(s)
- Giovanni Maroli
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.; Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany..
| | - Anne Schänzer
- Institute of Neuropathology, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Stefan Günther
- Max Planck Institute for Heart and Lung Research, Bioinformatics and deep sequencing platform, Ludwigstr. 43., 61231 Bad Nauheim, Germany
| | - Claudia Garcia-Gonzalez
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.; Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain
| | - Stefan Rupp
- Department of Pediatric Cardiology and Congenital Heart Disease, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Hannah Schlierbach
- Institute of Neuropathology, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Yanpu Chen
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Johannes Graumann
- Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.; The German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main
| | - Astrid Wietelmann
- Magnetic Resonance Imaging Group, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Johnny Kim
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.; The German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main..
| |
Collapse
|
5
|
Chi K, Liu J, Li X, Wang H, Li Y, Liu Q, Zhou Y, Ge Y. Biomarkers of heart failure: advances in omics studies. Mol Omics 2024; 20:169-183. [PMID: 38224222 DOI: 10.1039/d3mo00173c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Heart failure is a complex syndrome characterized by progressive circulatory dysfunction, manifesting clinically as pulmonary and systemic venous congestion, alongside inadequate tissue perfusion. The early identification of HF, particularly at the mild and moderate stages (stages B and C), presents a clinical challenge due to the overlap of signs, symptoms, and natriuretic peptide levels with other cardiorespiratory pathologies. Nonetheless, early detection coupled with timely pharmacological intervention is imperative for enhancing patient outcomes. Advances in high-throughput omics technologies have enabled researchers to analyze patient-derived biofluids and tissues, discovering biomarkers that are sensitive and specific for HF diagnosis. Due to the diversity of HF etiology, it is insufficient to study the diagnostic data of early HF using a single omics technology. This study reviewed the latest progress in genomics, transcriptomics, proteomics, and metabolomics for the identification of HF biomarkers, offering novel insights into the early clinical diagnosis of HF. However, the validity of biomarkers depends on the disease status, intervention time, genetic diversity and comorbidities of the subjects. Moreover, biomarkers lack generalizability in different clinical settings. Hence, it is imperative to conduct multi-center, large-scale and standardized clinical trials to enhance the diagnostic accuracy and utility of HF biomarkers.
Collapse
Affiliation(s)
- Kuo Chi
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Jing Liu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Xinghua Li
- Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China.
| | - He Wang
- Department of Cardiovascular Disease II, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Yanliang Li
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Qingnan Liu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Yabin Zhou
- Department of Cardiovascular Disease II, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Yuan Ge
- Department of Cardiovascular Disease II, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| |
Collapse
|
6
|
Taube N, Kabir R, Ebenebe OV, Garbus H, Alam El Din SM, Illingworth E, Fitch M, Wang N, Kohr MJ. Prenatal arsenite exposure alters maternal cardiac remodeling during late pregnancy. Toxicol Appl Pharmacol 2024; 483:116833. [PMID: 38266874 PMCID: PMC10922692 DOI: 10.1016/j.taap.2024.116833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/08/2024] [Accepted: 01/21/2024] [Indexed: 01/26/2024]
Abstract
Exposure to inorganic arsenic through drinking water is widespread and has been linked to many chronic diseases, including cardiovascular disease. Arsenic exposure has been shown to alter hypertrophic signaling in the adult heart, as well as in utero offspring development. However, the effect of arsenic on maternal cardiac remodeling during pregnancy has not been studied. As such, there is a need to understand how environmental exposure contributes to adverse pregnancy-related cardiovascular events. This study seeks to understand the impact of trivalent inorganic arsenic exposure during gestation on maternal cardiac remodeling in late pregnancy, as well as offspring outcomes. C57BL/6 J mice were exposed to 0 (control), 100 or 1000 μg/L sodium arsenite (NaAsO2) beginning at embryonic day (E) 2.5 and continuing through E17.5. Maternal heart function and size were assessed via transthoracic echocardiography, gravimetric measurement, and histology. Transcript levels of hypertrophic markers were probed via qRT-PCR and confirmed by western blot. Offspring outcomes were assessed through echocardiography and gravimetric measurement. We found that maternal heart size was smaller and transcript levels of Esr1 (estrogen receptor alpha), Pgrmc1 (progesterone receptor membrane component 1) and Pgrmc2 (progesterone receptor membrane component 2) reduced during late pregnancy with exposure to 1000 μg/L iAs vs. non-exposed pregnant controls. Both 100 and 1000 μg/L iAs also reduced transcription of Nppa (atrial natriuretic peptide). Akt protein expression was also significantly reduced after 1000 μg/L iAs exposure in the maternal heart with no change in activating phosphorylation. This significant abrogation of maternal cardiac hypertrophy suggests that arsenic exposure during pregnancy can potentially contribute to cardiovascular disease. Taken together, our findings further underscore the importance of reducing arsenic exposure during pregnancy and indicate that more research is needed to assess the impact of arsenic and other environmental exposures on the maternal heart and adverse pregnancy events.
Collapse
Affiliation(s)
- Nicole Taube
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Raihan Kabir
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Obialunanma V Ebenebe
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Haley Garbus
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Sarah-Marie Alam El Din
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Emily Illingworth
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Michael Fitch
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Nadan Wang
- Cardiology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mark J Kohr
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States.
| |
Collapse
|
7
|
Wang J, Tomar D, Martin TG, Dubey S, Dubey PK, Song J, Landesberg G, McCormick MG, Myers VD, Merali S, Merali C, Lemster B, McTiernan CF, Khalili K, Madesh M, Cheung JY, Kirk JA, Feldman AM. Bag3 Regulates Mitochondrial Function and the Inflammasome Through Canonical and Noncanonical Pathways in the Heart. JACC Basic Transl Sci 2023; 8:820-839. [PMID: 37547075 PMCID: PMC10401293 DOI: 10.1016/j.jacbts.2022.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/14/2022] [Accepted: 12/29/2022] [Indexed: 08/08/2023]
Abstract
B-cell lymphoma 2-associated athanogene-3 (Bag3) is expressed in all animal species, with Bag3 levels being most prominent in the heart, the skeletal muscle, the central nervous system, and in many cancers. Preclinical studies of Bag3 biology have focused on animals that have developed compromised cardiac function; however, the present studies were performed to identify the pathways perturbed in the heart even before the occurrence of clinical signs of dilatation and failure of the heart. These studies show that hearts carrying variants that knockout one allele of BAG3 have significant alterations in multiple cellular pathways including apoptosis, autophagy, mitochondrial homeostasis, and the inflammasome.
Collapse
Affiliation(s)
- JuFang Wang
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
- Center for Neurovirology and Gene Editing, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Dhadendra Tomar
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Thomas G. Martin
- Department of Cell and Molecular Physiology, Loyola University Strich School of Medicine, Maywood, Illinois, USA
| | - Shubham Dubey
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Praveen K. Dubey
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Jianliang Song
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
- Center for Neurovirology and Gene Editing, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Gavin Landesberg
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
- Center for Neurovirology and Gene Editing, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Michael G. McCormick
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | | | - Salim Merali
- Temple University School of Pharmacy, Philadelphia, Pennsylvania, USA
| | - Carmen Merali
- Temple University School of Pharmacy, Philadelphia, Pennsylvania, USA
| | - Bonnie Lemster
- Department of Medicine, Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Charles F. McTiernan
- Department of Medicine, Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kamel Khalili
- Center for Neurovirology and Gene Editing, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Muniswamy Madesh
- Department of Medicine, Center for Precision Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Joseph Y. Cheung
- Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan A. Kirk
- Department of Cell and Molecular Physiology, Loyola University Strich School of Medicine, Maywood, Illinois, USA
| | - Arthur M. Feldman
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
- Center for Neurovirology and Gene Editing, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Tedesco B, Vendredy L, Timmerman V, Poletti A. The chaperone-assisted selective autophagy complex dynamics and dysfunctions. Autophagy 2023:1-23. [PMID: 36594740 DOI: 10.1080/15548627.2022.2160564] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Each protein must be synthesized with the correct amino acid sequence, folded into its native structure, and transported to a relevant subcellular location and protein complex. If any of these steps fail, the cell has the capacity to break down aberrant proteins to maintain protein homeostasis (also called proteostasis). All cells possess a set of well-characterized protein quality control systems to minimize protein misfolding and the damage it might cause. Autophagy, a conserved pathway for the degradation of long-lived proteins, aggregates, and damaged organelles, was initially characterized as a bulk degradation pathway. However, it is now clear that autophagy also contributes to intracellular homeostasis by selectively degrading cargo material. One of the pathways involved in the selective removal of damaged and misfolded proteins is chaperone-assisted selective autophagy (CASA). The CASA complex is composed of three main proteins (HSPA, HSPB8 and BAG3), essential to maintain protein homeostasis in muscle and neuronal cells. A failure in the CASA complex, caused by mutations in the respective coding genes, can lead to (cardio)myopathies and neurodegenerative diseases. Here, we summarize our current understanding of the CASA complex and its dynamics. We also briefly discuss how CASA complex proteins are involved in disease and may represent an interesting therapeutic target.Abbreviation ALP: autophagy lysosomal pathway; ALS: amyotrophic lateral sclerosis; AMOTL1: angiomotin like 1; ARP2/3: actin related protein 2/3; BAG: BAG cochaperone; BAG3: BAG cochaperone 3; CASA: chaperone-assisted selective autophagy; CMA: chaperone-mediated autophagy; DNAJ/HSP40: DnaJ heat shock protein family (Hsp40); DRiPs: defective ribosomal products; EIF2A/eIF2α: eukaryotic translation initiation factor 2A; EIF2AK1/HRI: eukaryotic translation initiation factor 2 alpha kinase 1; GABARAP: GABA type A receptor-associated protein; HDAC6: histone deacetylase 6; HSP: heat shock protein; HSPA/HSP70: heat shock protein family A (Hsp70); HSP90: heat shock protein 90; HSPB8: heat shock protein family B (small) member 8; IPV: isoleucine-proline-valine; ISR: integrated stress response; KEAP1: kelch like ECH associated protein 1; LAMP2A: lysosomal associated membrane protein 2A; LATS1: large tumor suppressor kinase 1; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOC: microtubule organizing center; MTOR: mechanistic target of rapamycin kinase; NFKB/NF-κB: nuclear factor kappa B; NFE2L2: NFE2 like bZIP transcription factor 2; PLCG/PLCγ: phospholipase C gamma; polyQ: polyglutamine; PQC: protein quality control; PxxP: proline-rich; RAN translation: repeat-associated non-AUG translation; SG: stress granule; SOD1: superoxide dismutase 1; SQSTM1/p62: sequestosome 1; STUB1/CHIP: STIP1 homology and U-box containing protein 1; STK: serine/threonine kinase; SYNPO: synaptopodin; TBP: TATA-box binding protein; TARDBP/TDP-43: TAR DNA binding protein; TFEB: transcription factor EB; TPR: tetratricopeptide repeats; TSC1: TSC complex subunit 1; UBA: ubiquitin associated; UPS: ubiquitin-proteasome system; WW: tryptophan-tryptophan; WWTR1: WW domain containing transcription regulator 1; YAP1: Yes1 associated transcriptional regulator.
Collapse
Affiliation(s)
- Barbara Tedesco
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari, Dipartimento di Eccellenza 2018-2027, Università degli studi di Milano, Milan, Italy.,Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Leen Vendredy
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, Institute Born Bunge, University of Antwerp, Antwerpen, Belgium
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, Institute Born Bunge, University of Antwerp, Antwerpen, Belgium
| | - Angelo Poletti
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari, Dipartimento di Eccellenza 2018-2027, Università degli studi di Milano, Milan, Italy
| |
Collapse
|
9
|
Qu H, Feldman AM, Hakonarson H. Genetics of BAG3: A Paradigm for Developing Precision Therapies for Dilated Cardiomyopathies. J Am Heart Assoc 2022; 11:e027373. [PMID: 36382946 PMCID: PMC9851466 DOI: 10.1161/jaha.122.027373] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022]
Abstract
Nonischemic dilated cardiomyopathy is a common form of heart muscle disease in which genetic factors play a critical etiological role. In this regard, both rare disease-causing mutations and common disease-susceptible variants, in the Bcl-2-associated athanogene 3 (BAG3) gene have been reported, highlighting the critical role of BAG3 in cardiomyocytes and in the development of dilated cardiomyopathy. The phenotypic effects of the BAG3 mutations help investigators understand the structure and function of the BAG3 gene. Indeed, we report herein that all of the known pathogenic/likely pathogenic variants affect at least 1 of 3 protein functional domains, ie, the WW domain, the second IPV (Ile-Pro-Val) domain, or the BAG domain, whereas none of the missense nontruncating pathogenic/likely pathogenic variants affect the proline-rich repeat (PXXP) domain. A common variant, p.Cys151Arg, associated with reduced susceptibility to dilated cardiomyopathy demonstrated a significant difference in allele frequencies among diverse human populations, suggesting evolutionary selective pressure. As BAG3-related therapies for heart failure move from the laboratory to the clinic, the ability to provide precision medicine will depend in large part on having a thorough understanding of the potential effects of both common and uncommon genetic variants on these target proteins. The current review article provides a roadmap that investigators can utilize to determine the potential interactions between a patient's genotype, their phenotype, and their response to therapeutic interventions with both gene delivery and small molecules.
Collapse
Affiliation(s)
- Hui‐Qi Qu
- The Center for Applied Genomics, Children’s Hospital of PhiladelphiaPhiladelphiaPA
| | - Arthur M. Feldman
- Department of Medicine, Division of CardiologyThe Lewis Katz School of Medicine at Temple UniversityPhiladelphiaPA
- The Center for Neurovirology and Gene EditingThe Lewis Katz School of Medicine at Temple UniversityPhiladelphiaPA
| | - Hakon Hakonarson
- The Center for Applied Genomics, Children’s Hospital of PhiladelphiaPhiladelphiaPA
- Department of Pediatrics, The Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA
- Division of Human GeneticsChildren’s Hospital of PhiladelphiaPhiladelphiaPA
- Division of Pulmonary MedicineChildren’s Hospital of PhiladelphiaPhiladelphiaPA
- Faculty of MedicineUniversity of IcelandReykjavikIceland
| |
Collapse
|
10
|
Chintanaphol M, Orgil BO, Alberson NR, Towbin JA, Purevjav E. Restrictive cardiomyopathy: from genetics and clinical overview to animal modeling. Rev Cardiovasc Med 2022; 23:108. [PMID: 35345275 DOI: 10.31083/j.rcm2303108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/08/2021] [Accepted: 10/25/2021] [Indexed: 11/06/2022] Open
Abstract
Restrictive cardiomyopathy (RCM), a potentially devastating heart muscle disorder, is characterized by diastolic dysfunction due to abnormal muscle relaxation and myocardial stiffness resulting in restrictive filling of the ventricles. Diastolic dysfunction is often accompanied by left atrial or bi-atrial enlargement and normal ventricular size and systolic function. RCM is the rarest form of cardiomyopathy, accounting for 2-5% of pediatric cardiomyopathy cases, however, survival rates have been reported to be 82%, 80%, and 68% at 1-, 2-, and 5-years after diagnosis, respectively. RCM can be idiopathic, familial, or secondary to a systemic disorder, such as amyloidosis, sarcoidosis, and hereditary hemochromatosis. Approximately 30% of cases are familial RCM, and the genes that have been linked to RCM are cTnT, cTnI, MyBP-C, MYH7, MYL2, MYL3, DES, MYPN, TTN, BAG3, DCBLD2, LNMA, and FLNC. Increased Ca2+ sensitivity, sarcomere disruption, and protein aggregates are some of the few mechanisms of pathogenesis that have been revealed by studies utilizing cell lines and animal models. Additional exploration into the pathogenesis of RCM is necessary to create novel therapeutic strategies to reverse restrictive cardiomyopathic phenotypes.
Collapse
Affiliation(s)
- Michelle Chintanaphol
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Buyan-Ochir Orgil
- Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103, USA
| | - Neely R Alberson
- Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103, USA
| | - Jeffrey A Towbin
- Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103, USA
- Pediatric Cardiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Enkhsaikhan Purevjav
- Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103, USA
| |
Collapse
|
11
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
12
|
Lin H, Koren SA, Cvetojevic G, Girardi P, Johnson GV. The role of BAG3 in health and disease: A "Magic BAG of Tricks". J Cell Biochem 2022; 123:4-21. [PMID: 33987872 PMCID: PMC8590707 DOI: 10.1002/jcb.29952] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/25/2021] [Indexed: 01/03/2023]
Abstract
The multi-domain structure of Bcl-2-associated athanogene 3 (BAG3) facilitates its interaction with many different proteins that participate in regulating a variety of biological pathways. After revisiting the BAG3 literature published over the past ten years with Citespace software, we classified the BAG3 research into several clusters, including cancer, cardiomyopathy, neurodegeneration, and viral propagation. We then highlighted recent key findings in each cluster. To gain greater insight into the roles of BAG3, we analyzed five different published mass spectrometry data sets of proteins that co-immunoprecipitate with BAG3. These data gave us insight into universal, as well as cell-type-specific BAG3 interactors in cancer cells, cardiomyocytes, and neurons. Finally, we mapped variable BAG3 SNPs and also mutation data from previous publications to further explore the link between the domains and function of BAG3. We believe this review will provide a better understanding of BAG3 and direct future studies towards understanding BAG3 function in physiological and pathological conditions.
Collapse
Affiliation(s)
- Heng Lin
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester NY 14642 USA
| | - Shon A. Koren
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester NY 14642 USA
| | - Gregor Cvetojevic
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester NY 14642 USA
| | - Peter Girardi
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester NY 14642 USA
| | - Gail V.W. Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester NY 14642 USA
| |
Collapse
|
13
|
Sun X, Siri S, Hurst A, Qiu H. Heat Shock Protein 22 in Physiological and Pathological Hearts: Small Molecule, Large Potentials. Cells 2021; 11:cells11010114. [PMID: 35011676 PMCID: PMC8750610 DOI: 10.3390/cells11010114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/22/2022] Open
Abstract
Small heat shock protein 22 (HSP22) belongs to the superfamily of heat shock proteins and is predominantly expressed in the heart, brain, skeletal muscle, and different types of cancers. It has been found that HSP22 is involved in variant cellular functions in cardiomyocytes and plays a vital role in cardiac protection against cardiomyocyte injury under diverse stress. This review summarizes the multiple functions of HSP22 in the heart and the underlying molecular mechanisms through modulating gene transcription, post-translational modification, subcellular translocation of its interacting proteins, and protein degradation, facilitating mitochondrial function, cardiac metabolism, autophagy, and ROS production and antiapoptotic effect. We also discuss the association of HSP22 in cardiac pathologies, including human dilated cardiomyopathy, pressure overload-induced heart failure, ischemic heart diseases, and aging-related cardiac metabolism disorder. The collected information would provide insights into the understanding of the HSP22 in heart diseases and lead to discovering the therapeutic targets.
Collapse
|
14
|
With or without You: Co-Chaperones Mediate Health and Disease by Modifying Chaperone Function and Protein Triage. Cells 2021; 10:cells10113121. [PMID: 34831344 PMCID: PMC8619055 DOI: 10.3390/cells10113121] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 01/18/2023] Open
Abstract
Heat shock proteins (HSPs) are a family of molecular chaperones that regulate essential protein refolding and triage decisions to maintain protein homeostasis. Numerous co-chaperone proteins directly interact and modify the function of HSPs, and these interactions impact the outcome of protein triage, impacting everything from structural proteins to cell signaling mediators. The chaperone/co-chaperone machinery protects against various stressors to ensure cellular function in the face of stress. However, coding mutations, expression changes, and post-translational modifications of the chaperone/co-chaperone machinery can alter the cellular stress response. Importantly, these dysfunctions appear to contribute to numerous human diseases. Therapeutic targeting of chaperones is an attractive but challenging approach due to the vast functions of HSPs, likely contributing to the off-target effects of these therapies. Current efforts focus on targeting co-chaperones to develop precise treatments for numerous diseases caused by defects in protein quality control. This review focuses on the recent developments regarding selected HSP70/HSP90 co-chaperones, with a concentration on cardioprotection, neuroprotection, cancer, and autoimmune diseases. We also discuss therapeutic approaches that highlight both the utility and challenges of targeting co-chaperones.
Collapse
|
15
|
Hsp22 Deficiency Induces Age-Dependent Cardiac Dilation and Dysfunction by Impairing Autophagy, Metabolism, and Oxidative Response. Antioxidants (Basel) 2021; 10:antiox10101550. [PMID: 34679684 PMCID: PMC8533440 DOI: 10.3390/antiox10101550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/19/2021] [Accepted: 09/27/2021] [Indexed: 02/04/2023] Open
Abstract
Heat shock protein 22 (Hsp22) is a small heat shock protein predominantly expressed in skeletal and cardiac muscle. Previous studies indicate that Hsp22 plays a vital role in protecting the heart against cardiac stress. However, the essential role of Hsp22 in the heart under physiological conditions remains largely unknown. In this study, we used an Hsp22 knockout (KO) mouse model to determine whether loss of Hsp22 impairs cardiac growth and function with increasing age under physiological conditions. Cardiac structural and functional alterations at baseline were measured using echocardiography and invasive catheterization in Hsp22 KO mice during aging transition compared to their age-matched wild-type (WT) littermates. Our results showed that Hsp22 deletion induced progressive cardiac dilation along with declined function during the aging transition. Mechanistically, the loss of Hsp22 impaired BCL-2-associated athanogene 3 (BAG3) expression and its associated cardiac autophagy, undermined cardiac energy metabolism homeostasis and increased oxidative damage. This study showed that Hsp22 played an essential role in the non-stressed heart during the early stage of aging, which may bring new insight into understanding the pathogenesis of age-related dilated cardiomyopathy.
Collapse
|
16
|
Kirk JA, Cheung JY, Feldman AM. Therapeutic targeting of BAG3: considering its complexity in cancer and heart disease. J Clin Invest 2021; 131:e149415. [PMID: 34396980 DOI: 10.1172/jci149415] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Bcl2-associated athanogene-3 (BAG3) is expressed ubiquitously in humans, but its levels are highest in the heart, the skeletal muscle, and the central nervous system; it is also elevated in many cancers. BAG3's diverse functions are supported by its multiple protein-protein binding domains, which couple with small and large heat shock proteins, members of the Bcl2 family, other antiapoptotic proteins, and various sarcomere proteins. In the heart, BAG3 inhibits apoptosis, promotes autophagy, couples the β-adrenergic receptor with the L-type Ca2+ channel, and maintains the structure of the sarcomere. In cancer cells, BAG3 binds to and supports an identical array of prosurvival proteins, and it may represent a therapeutic target. However, the development of strategies to block BAG3 function in cancer cells may be challenging, as they are likely to interfere with the essential roles of BAG3 in the heart. In this Review, we present the current knowledge regarding the biology of this complex protein in the heart and in cancer and suggest several therapeutic options.
Collapse
Affiliation(s)
- Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, USA
| | - Joseph Y Cheung
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Arthur M Feldman
- Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
17
|
Höhfeld J, Benzing T, Bloch W, Fürst DO, Gehlert S, Hesse M, Hoffmann B, Hoppe T, Huesgen PF, Köhn M, Kolanus W, Merkel R, Niessen CM, Pokrzywa W, Rinschen MM, Wachten D, Warscheid B. Maintaining proteostasis under mechanical stress. EMBO Rep 2021; 22:e52507. [PMID: 34309183 PMCID: PMC8339670 DOI: 10.15252/embr.202152507] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/11/2022] Open
Abstract
Cell survival, tissue integrity and organismal health depend on the ability to maintain functional protein networks even under conditions that threaten protein integrity. Protection against such stress conditions involves the adaptation of folding and degradation machineries, which help to preserve the protein network by facilitating the refolding or disposal of damaged proteins. In multicellular organisms, cells are permanently exposed to stress resulting from mechanical forces. Yet, for long time mechanical stress was not recognized as a primary stressor that perturbs protein structure and threatens proteome integrity. The identification and characterization of protein folding and degradation systems, which handle force-unfolded proteins, marks a turning point in this regard. It has become apparent that mechanical stress protection operates during cell differentiation, adhesion and migration and is essential for maintaining tissues such as skeletal muscle, heart and kidney as well as the immune system. Here, we provide an overview of recent advances in our understanding of mechanical stress protection.
Collapse
Affiliation(s)
- Jörg Höhfeld
- Institute for Cell BiologyRheinische Friedrich‐Wilhelms University BonnBonnGermany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Wilhelm Bloch
- Institute of Cardiovascular Research and Sports MedicineGerman Sport UniversityCologneGermany
| | - Dieter O Fürst
- Institute for Cell BiologyRheinische Friedrich‐Wilhelms University BonnBonnGermany
| | - Sebastian Gehlert
- Institute of Cardiovascular Research and Sports MedicineGerman Sport UniversityCologneGermany
- Department for the Biosciences of SportsInstitute of Sports ScienceUniversity of HildesheimHildesheimGermany
| | - Michael Hesse
- Institute of Physiology I, Life & Brain CenterMedical FacultyRheinische Friedrich‐Wilhelms UniversityBonnGermany
| | - Bernd Hoffmann
- Institute of Biological Information Processing, IBI‐2: MechanobiologyForschungszentrum JülichJülichGermany
| | - Thorsten Hoppe
- Institute for GeneticsCologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD) and CMMCUniversity of CologneCologneGermany
| | - Pitter F Huesgen
- Central Institute for Engineering, Electronics and Analytics, ZEA3Forschungszentrum JülichJülichGermany
- CECADUniversity of CologneCologneGermany
| | - Maja Köhn
- Institute of Biology IIIFaculty of Biology, and Signalling Research Centres BIOSS and CIBSSAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| | - Waldemar Kolanus
- LIMES‐InstituteRheinische Friedrich‐Wilhelms University BonnBonnGermany
| | - Rudolf Merkel
- Institute of Biological Information Processing, IBI‐2: MechanobiologyForschungszentrum JülichJülichGermany
| | - Carien M Niessen
- Department of Dermatology and CECADUniversity of CologneCologneGermany
| | | | - Markus M Rinschen
- Department of Biomedicine and Aarhus Institute of Advanced StudiesAarhus UniversityAarhusDenmark
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Dagmar Wachten
- Institute of Innate ImmunityUniversity Hospital BonnBonnGermany
| | - Bettina Warscheid
- Institute of Biology IIFaculty of Biology, and Signalling Research Centres BIOSS and CIBSSAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| |
Collapse
|
18
|
Jia P, Wu N, Yang H, Guo Y, Guo X, Sun Y. Different roles of BAG3 in cardiac physiological hypertrophy and pathological remodeling. Transl Res 2021; 233:47-61. [PMID: 33578031 DOI: 10.1016/j.trsl.2021.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 12/30/2022]
Abstract
Heart failure is one of the leading causes of death worldwide. A stimulated heart undergoes either adaptive physiological hypertrophy, which can maintain a normal heart function, or maladaptive pathological remodeling, which can deteriorate heart function. These 2 kinds of remodeling often co-occur at the early stages of many heart diseases and have important effects on cardiac function. The Bcl2-associated athanogene 3 (BAG3) protein is highly expressed in the heart and has many functions. However, it is unknown how BAG3 is regulated and what its function is during physiological hypertrophy and pathological remodeling. We generated tamoxifen-induced, heart-specific heterozygous and homozygous BAG3 knockout mouse models (BAG3 protein level decreased by approximately 40% and 80% in the hearts after tamoxifen administration). BAG3 knockout models were subjected to swimming training or phenylephrine (PE) infusion to induce cardiac physiological hypertrophy and pathological remodeling. Neonatal rat ventricular cardiomyocytes (NRVCs) were used to study BAG3 functions and mechanisms in vitro. We found that BAG3 was upregulated in physiological hypertrophy and in pathological remodeling both in vivo and in vitro. Heterozygous or homozygous knockout BAG3 in mouse hearts and knockdown of BAG3 in the NRVCs blunted physiological hypertrophy and aggravated pathological remodeling, while overexpression of BAG3 promoted physiological hypertrophy and inhibited pathological remodeling in NRVCs. Mechanistically, BAG3 overexpression in NRVCs promoted physiological hypertrophy by activating the protein kinase B (AKT)/mammalian (or mechanistic) target of rapamycin (mTOR) pathway. BAG3 knockdown in NRVCs aggravated pathological remodeling through activation of the calcineurin/nuclear factor of activated T cells 2 (NFATc2) pathway. Because BAG3 has a dual role in cardiac remodeling, heart-specific regulation of BAG3 may be an effective therapeutic strategy to protect against deterioration of heart function and heart failure caused by many heart diseases.
Collapse
Affiliation(s)
- Pengyu Jia
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Nan Wu
- The Central Laboratory of the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Huimin Yang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yuxuan Guo
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiaofan Guo
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
19
|
Overexpression of human BAG3 P209L in mice causes restrictive cardiomyopathy. Nat Commun 2021; 12:3575. [PMID: 34117258 PMCID: PMC8196106 DOI: 10.1038/s41467-021-23858-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 05/20/2021] [Indexed: 12/15/2022] Open
Abstract
An amino acid exchange (P209L) in the HSPB8 binding site of the human co-chaperone BAG3 gives rise to severe childhood cardiomyopathy. To phenocopy the disease in mice and gain insight into its mechanisms, we generated humanized transgenic mouse models. Expression of human BAG3P209L-eGFP in mice caused Z-disc disintegration and formation of protein aggregates. This was accompanied by massive fibrosis resulting in early-onset restrictive cardiomyopathy with increased mortality as observed in patients. RNA-Seq and proteomics revealed changes in the protein quality control system and increased autophagy in hearts from hBAG3P209L-eGFP mice. The mutation renders hBAG3P209L less soluble in vivo and induces protein aggregation, but does not abrogate hBAG3 binding properties. In conclusion, we report a mouse model mimicking the human disease. Our data suggest that the disease mechanism is due to accumulation of hBAG3P209L and mouse Bag3, causing sequestering of components of the protein quality control system and autophagy machinery leading to sarcomere disruption. An amino acid exchange (P209L) in the human co-chaperone BAG3 gives rise to severe childhood restrictive cardiomyopathy. Here the authors describe humanized transgenic mouse models which phenocopy the disease and provide insight into the pathogenic mechanisms.
Collapse
|
20
|
Martin TG, Myers VD, Dubey P, Dubey S, Perez E, Moravec CS, Willis MS, Feldman AM, Kirk JA. Cardiomyocyte contractile impairment in heart failure results from reduced BAG3-mediated sarcomeric protein turnover. Nat Commun 2021; 12:2942. [PMID: 34011988 PMCID: PMC8134551 DOI: 10.1038/s41467-021-23272-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 04/22/2021] [Indexed: 12/22/2022] Open
Abstract
The association between reduced myofilament force-generating capacity (Fmax) and heart failure (HF) is clear, however the underlying molecular mechanisms are poorly understood. Here, we show impaired Fmax arises from reduced BAG3-mediated sarcomere turnover. Myofilament BAG3 expression decreases in human HF and positively correlates with Fmax. We confirm this relationship using BAG3 haploinsufficient mice, which display reduced Fmax and increased myofilament ubiquitination, suggesting impaired protein turnover. We show cardiac BAG3 operates via chaperone-assisted selective autophagy (CASA), conserved from skeletal muscle, and confirm sarcomeric CASA complex localization is BAG3/proteotoxic stress-dependent. Using mass spectrometry, we characterize the myofilament CASA interactome in the human heart and identify eight clients of BAG3-mediated turnover. To determine if increasing BAG3 expression in HF can restore sarcomere proteostasis/Fmax, HF mice were treated with rAAV9-BAG3. Gene therapy fully rescued Fmax and CASA protein turnover after four weeks. Our findings indicate BAG3-mediated sarcomere turnover is fundamental for myofilament functional maintenance. Decreased expression of BAG3 in the heart is associated with contractile dysfunction and heart failure. Here the authors show that this is due to decreased BAG3-dependent sarcomere protein turnover, which impairs mechanical function, and that sarcomere force-generating capacity is restored with BAG3 gene therapy.
Collapse
Affiliation(s)
- Thomas G Martin
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL, USA
| | - Valerie D Myers
- Department of Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Praveen Dubey
- Department of Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Shubham Dubey
- Department of Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Edith Perez
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL, USA
| | - Christine S Moravec
- Department of Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA
| | - Monte S Willis
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Arthur M Feldman
- Department of Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL, USA.
| |
Collapse
|
21
|
Huot JR, Pin F, Narasimhan A, Novinger LJ, Keith AS, Zimmers TA, Willis MS, Bonetto A. ACVR2B antagonism as a countermeasure to multi-organ perturbations in metastatic colorectal cancer cachexia. J Cachexia Sarcopenia Muscle 2020; 11:1779-1798. [PMID: 33200567 PMCID: PMC7749603 DOI: 10.1002/jcsm.12642] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/11/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Advanced colorectal cancer (CRC) is often accompanied by the development of liver metastases, as well as cachexia, a multi-organ co-morbidity primarily affecting skeletal (SKM) and cardiac muscles. Activin receptor type 2B (ACVR2B) signalling is known to cause SKM wasting, and its inhibition restores SKM mass and prolongs survival in cancer. Using a recently generated mouse model, here we tested whether ACVR2B blockade could preserve multiple organs, including skeletal and cardiac muscle, in the presence of metastatic CRC. METHODS NSG male mice (8 weeks old) were injected intrasplenically with HCT116 human CRC cells (mHCT116), while sham-operated animals received saline (n = 5-10 per group). Sham and tumour-bearing mice received weekly injections of ACVR2B/Fc, a synthetic peptide inhibitor of ACVR2B. RESULTS mHCT116 hosts displayed losses in fat mass ( - 79%, P < 0.0001), bone mass ( - 39%, P < 0.05), and SKM mass (quadriceps: - 22%, P < 0.001), in line with reduced muscle cross-sectional area ( - 24%, P < 0.01) and plantarflexion force ( - 28%, P < 0.05). Further, despite only moderately affected heart size, cardiac function was significantly impaired (ejection fraction %: - 16%, P < 0.0001; fractional shortening %: - 25%, P < 0.0001) in the mHCT116 hosts. Conversely, ACVR2B/Fc preserved fat mass ( + 238%, P < 0.001), bone mass ( + 124%, P < 0.0001), SKM mass (quadriceps: + 31%, P < 0.0001), size (cross-sectional area: + 43%, P < 0.0001) and plantarflexion force ( + 28%, P < 0.05) in tumour hosts. Cardiac function was also completely preserved in tumour hosts receiving ACVR2B/Fc (ejection fraction %: + 19%, P < 0.0001), despite no effect on heart size. RNA sequencing analysis of heart muscle revealed rescue of genes related to cardiac development and contraction in tumour hosts treated with ACVR2B/Fc. CONCLUSIONS Our metastatic CRC model recapitulates the multi-systemic derangements of cachexia by displaying loss of fat, bone, and SKM along with decreased muscle strength in mHCT116 hosts. Additionally, with evidence of severe cardiac dysfunction, our data support the development of cardiac cachexia in the occurrence of metastatic CRC. Notably, ACVR2B antagonism preserved adipose tissue, bone, and SKM, whereas muscle and cardiac functions were completely maintained upon treatment. Altogether, our observations implicate ACVR2B signalling in the development of multi-organ perturbations in metastatic CRC and further dictate that ACVR2B represents a promising therapeutic target to preserve body composition and functionality in cancer cachexia.
Collapse
Affiliation(s)
- Joshua R Huot
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fabrizio Pin
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ashok Narasimhan
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Leah J Novinger
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Teresa A Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.,Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Monte S Willis
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.,Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrea Bonetto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.,Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
22
|
Xu B, Zang SC, Lang LM, Lian S, Lu J, Li SZ, Yang HM, Zhen L. Down-regulation of miR-383-5p suppresses apoptosis in oxidative stress rat hepatocytes by targeting Bcl2. J Anim Physiol Anim Nutr (Berl) 2020; 104:1948-1959. [PMID: 32090391 DOI: 10.1111/jpn.13328] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/25/2019] [Accepted: 01/25/2020] [Indexed: 12/23/2022]
Abstract
miRNAs are a class of small non-coding RNAs that are involved in various biological processes. In the preliminary work of the laboratory, found that miR-383-5p was down-regulated in the liver tissue of acute cold stress rats and has been shown to be an important regulatory factor in tumour proliferation, but there are very few studies involving the mediation of cold stress in rat liver tissues. Therefore, the purpose of this study was to determine the effect of miR-383-5p on the livers of cold stress rats by simulating the cold stress state of rat liver tissues in vitro using H2 O2 to induce rat hepatocyte oxidative stress. The results showed that MDA content, Caspase 3 and Cyto C protein levels increased significantly; GPx activity and SOD1 protein levels decreased significantly and miR-383-5p expression was significantly down-regulated in rat liver tissues after cold stress. Different concentrations of H2 O2 was added to rat hepatocytes, and the results showed that the expression of miR-383-5p, the ROS level, and the apoptosis rate in rat hepatocytes was increased significantly in a concentration-dependent fashion. Transfection of miR-383-5p inhibitor revealed that the apoptosis rate of rat hepatocytes, and the protein level of apoptosis-related protein Caspase 3 were reduced; the results of the dual-luciferase reporter gene assay showed that miR-383-5p targeted regulation of Bcl2. The results suggested that the expression of miR-383-5p was up-regulated in oxidative stress rat hepatocytes and may aggravate the apoptosis of rat hepatocytes induced by targeting inhibition of Bcl2 translation.
Collapse
Affiliation(s)
- Bin Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shu-Cheng Zang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Li-Min Lang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shuai Lian
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jingjing Lu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shi-Ze Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Huan-Min Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Li Zhen
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| |
Collapse
|
23
|
Abstract
The B cell lymphoma 2-associated anthanogene (BAG3) is an anti-apoptotic co-chaperone protein. Previous reports suggest that mutations in BAG3 are associated with dilated cardiomyopathy. This review aims to summarize the current understanding of the relationship between BAG3 mutations and dilated cardiomyopathy, primarily focusing on the role and protective mechanism of BAG3 in cardiomyocytes from individuals with dilated cardiomyopathy. The results of published studies show that BAG3 is critically important for reducing cardiomyocyte apoptosis, maintaining protein homeostasis, regulating mitochondrial stability, modulating myocardial contraction, and reducing cardiac arrhythmia, which suggests an indispensable protective mechanism of BAG3 in dilated cardiomyopathy. The significant role of BAG3 in protecting cardiomyocytes provides a new direction for the diagnosis and treatment of dilated cardiomyopathy. However, further research is required to explore the molecular mechanisms that regulate BAG3 expression, to identify a novel therapy for patients with dilated cardiomyopathy.
Collapse
|
24
|
Cresci S, Pereira NL, Ahmad F, Byku M, de las Fuentes L, Lanfear DE, Reilly CM, Owens AT, Wolf MJ. Heart Failure in the Era of Precision Medicine: A Scientific Statement From the American Heart Association. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2019; 12:458-485. [PMID: 31510778 DOI: 10.1161/hcg.0000000000000058] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
One of 5 people will develop heart failure over his or her lifetime. Early diagnosis and better understanding of the pathophysiology of this disease are critical to optimal treatment. The "omics"-genomics, pharmacogenomics, epigenomics, proteomics, metabolomics, and microbiomics- of heart failure represent rapidly expanding fields of science that have, to date, not been integrated into a single body of work. The goals of this statement are to provide a comprehensive overview of the current state of these omics as they relate to the development and progression of heart failure and to consider the current and potential future applications of these data for precision medicine with respect to prevention, diagnosis, and therapy.
Collapse
|
25
|
Fang X, Bogomolovas J, Zhou PS, Mu Y, Ma X, Chen Z, Zhang L, Zhu M, Veevers J, Ouyang K, Chen J. P209L mutation in Bag3 does not cause cardiomyopathy in mice. Am J Physiol Heart Circ Physiol 2018; 316:H392-H399. [PMID: 30499714 DOI: 10.1152/ajpheart.00714.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Bcl-2-associated athanogene 3 (BAG3) is a cochaperone protein and a central player of the cellular protein quality control system. BAG3 is prominently expressed in the heart and plays an essential role in cardiac protein homeostasis by interacting with chaperone heat shock proteins (HSPs) in large, functionally distinct multichaperone complexes. The BAG3 mutation of proline 209 to leucine (P209L), which resides in a critical region that mediates the direct interaction between BAG3 and small HSPs (sHSPs), is associated with cardiomyopathy in humans. However, the mechanism by which the BAG3 P209L missense mutation leads to cardiomyopathy remains unknown. To determine the molecular basis underlying the cardiomyopathy caused by the BAG3 P209L mutation, we generated a knockin (KI) mouse model in which the endogenous Bag3 gene was replaced with mutant Bag3 containing the P215L mutation, which is equivalent to the human P209L mutation. We performed physiological, histological, and biochemical analyses of Bag3 P209L KI mice to determine the functional, morphological, and molecular consequences of the P209L mutation. We found that Bag3 P209L KI mice exhibited normal cardiac function and morphology up to 16 mo of age. Western blot analysis further revealed that levels of sHSPs, stress-inducible HSPs, ubiquitinated proteins, and autophagy were unaffected in P209L mutant mouse hearts. In conclusion, the P209L mutation in Bag3 does not cause cardiomyopathy in mice up to 16 mo of age under baseline conditions. NEW & NOTEWORTHY Bcl-2-associated athanogene 3 (BAG3) P209L mutation is associated with human cardiomyopathy. A recent study reported that transgenic mice overexpressing human BAG3 P209L in cardiomyocytes have cardiac dysfunction. In contrast, our P209L mice that express mutant BAG3 at the same level as that of wild-type mice displayed no overt phenotype. Our results suggest that human cardiomyopathy may result from species-specific requirements for the conserved motif that is disrupted by P209L mutation or from genetic background-dependent effects.
Collapse
Affiliation(s)
- Xi Fang
- Department of Medicine, University of California-San Diego , La Jolla, California
| | - Julius Bogomolovas
- Department of Medicine, University of California-San Diego , La Jolla, California.,Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University , Mannheim , Germany
| | - Paul Shichao Zhou
- Department of Medicine, University of California-San Diego , La Jolla, California
| | - Yongxin Mu
- Department of Medicine, University of California-San Diego , La Jolla, California
| | - Xiaolong Ma
- Department of Medicine, University of California-San Diego , La Jolla, California.,Department of Cardiothoracic Surgery, The Second Xiangya Hospital, Central South University , Changsha, Hunan , China
| | - Zee Chen
- Department of Medicine, University of California-San Diego , La Jolla, California.,Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School , Shenzhen , China
| | - Lunfeng Zhang
- Department of Medicine, University of California-San Diego , La Jolla, California
| | - Mason Zhu
- Department of Medicine, University of California-San Diego , La Jolla, California
| | - Jennifer Veevers
- Department of Medicine, University of California-San Diego , La Jolla, California
| | - Kunfu Ouyang
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School , Shenzhen , China
| | - Ju Chen
- Department of Medicine, University of California-San Diego , La Jolla, California
| |
Collapse
|
26
|
Tahrir FG, Langford D, Amini S, Mohseni Ahooyi T, Khalili K. Mitochondrial quality control in cardiac cells: Mechanisms and role in cardiac cell injury and disease. J Cell Physiol 2018; 234:8122-8133. [PMID: 30417391 DOI: 10.1002/jcp.27597] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/19/2018] [Indexed: 12/17/2022]
Abstract
Mitochondria play an important role in maintaining cardiac homeostasis by supplying the major energy required for cardiac excitation-contraction coupling as well as controlling the key intracellular survival and death pathways. Healthy mitochondria generate ATP molecules through an aerobic process known as oxidative phosphorylation (OXPHOS). Mitochondrial injury during myocardial infarction (MI) impairs OXPHOS and results in the excessive production of reactive oxygen species (ROS), bioenergetic insufficiency, and contributes to the development of cardiovascular diseases. Therefore, mitochondrial biogenesis along with proper mitochondrial quality control machinery, which removes unhealthy mitochondria is pivotal for mitochondrial homeostasis and cardiac health. Upon damage to the mitochondrial network, mitochondrial quality control components are recruited to segregate the unhealthy mitochondria and target aberrant mitochondrial proteins for degradation and elimination. Impairment of mitochondrial quality control and accumulation of abnormal mitochondria have been reported in the pathogenesis of various cardiac disorders and heart failure. Here, we provide an overview of the recent studies describing various mechanistic pathways underlying mitochondrial homeostasis with the main focus on cardiac cells. In addition, this review demonstrates the potential effects of mitochondrial quality control dysregulation in the development of cardiovascular disease.
Collapse
Affiliation(s)
- Farzaneh G Tahrir
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Dianne Langford
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Shohreh Amini
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Taha Mohseni Ahooyi
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
27
|
Ranek MJ, Stachowski MJ, Kirk JA, Willis MS. The role of heat shock proteins and co-chaperones in heart failure. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2016.0530. [PMID: 29203715 DOI: 10.1098/rstb.2016.0530] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2017] [Indexed: 12/18/2022] Open
Abstract
The ongoing contractile and metabolic demands of the heart require a tight control over protein quality control, including the maintenance of protein folding, turnover and synthesis. In heart disease, increases in mechanical and oxidative stresses, post-translational modifications (e.g., phosphorylation), for example, decrease protein stability to favour misfolding in myocardial infarction, heart failure or ageing. These misfolded proteins are toxic to cardiomyocytes, directly contributing to the common accumulation found in human heart failure. One of the critical class of proteins involved in protecting the heart against these threats are molecular chaperones, including the heat shock protein70 (HSP70), HSP90 and co-chaperones CHIP (carboxy terminus of Hsp70-interacting protein, encoded by the Stub1 gene) and BAG-3 (BCL2-associated athanogene 3). Here, we review their emerging roles in the maintenance of cardiomyocytes in human and experimental models of heart failure, including their roles in facilitating the removal of misfolded and degraded proteins, inhibiting apoptosis and maintaining the structural integrity of the sarcomere and regulation of nuclear receptors. Furthermore, we discuss emerging evidence of increased expression of extracellular HSP70, HSP90 and BAG-3 in heart failure, with complementary independent roles from intracellular functions with important therapeutic and diagnostic considerations. While our understanding of these major HSPs in heart failure is incomplete, there is a clear potential role for therapeutic modulation of HSPs in heart failure with important contextual considerations to counteract the imbalance of protein damage and endogenous protein quality control systems.This article is part of the theme issue 'Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective'.
Collapse
Affiliation(s)
- Mark J Ranek
- Division of Cardiology, Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD 21205, USA
| | - Marisa J Stachowski
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University, Chicago, IL 60302, USA
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University, Chicago, IL 60302, USA
| | - Monte S Willis
- Department of Pathology and Laboratory Medicine, McAllister Heart Institute, CB#7525, Chapel Hill, NC 27599-7525, USA
| |
Collapse
|
28
|
Mota R, Parry TL, Yates CC, Qiang Z, Eaton SC, Mwiza JM, Tulasi D, Schisler JC, Patterson C, Zaglia T, Sandri M, Willis MS. Increasing Cardiomyocyte Atrogin-1 Reduces Aging-Associated Fibrosis and Regulates Remodeling in Vivo. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1676-1692. [PMID: 29758183 DOI: 10.1016/j.ajpath.2018.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 03/10/2018] [Accepted: 04/03/2018] [Indexed: 12/12/2022]
Abstract
The muscle-specific ubiquitin ligase atrogin-1 (MAFbx) has been identified as a critical regulator of pathologic and physiological cardiac hypertrophy; it regulates these processes by ubiquitinating transcription factors [nuclear factor of activated T-cells and forkhead box O (FoxO) 1/3]. However, the role of atrogin-1 in regulating transcription factors in aging has not previously been described. Atrogin-1 cardiomyocyte-specific transgenic (Tg+) adult mice (α-major histocompatibility complex promoter driven) have normal cardiac function and size. Herein, we demonstrate that 18-month-old atrogin-1 Tg+ hearts exhibit significantly increased anterior wall thickness without functional impairment versus wild-type mice. Histologic analysis at 18 months revealed atrogin-1 Tg+ mice had significantly less fibrosis and significantly greater nuclei and cardiomyocyte cross-sectional analysis. Furthermore, by real-time quantitative PCR, atrogin-1 Tg+ had increased Col 6a4, 6a5, 6a6, matrix metalloproteinase 8 (Mmp8), and Mmp9 mRNA, suggesting a role for atrogin-1 in regulating collagen deposits and MMP-8 and MMP-9. Because atrogin-1 Tg+ mice exhibited significantly less collagen deposition and protein levels, enhanced Mmp8 and Mmp9 mRNA may offer one mechanism by which collagen levels are kept in check in the aged atrogin-1 Tg+ heart. In addition, atrogin-1 Tg+ hearts showed enhanced FoxO1/3 activity. The present study shows a novel link between atrogin-1-mediated regulation of FoxO1/3 activity and reduced collagen deposition and fibrosis in the aged heart. Therefore, targeting FoxO1/3 activity via the muscle-specific atrogin-1 ubiquitin ligase may offer a muscle-specific method to modulate aging-related cardiac fibrosis.
Collapse
Affiliation(s)
- Roberto Mota
- McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina
| | - Traci L Parry
- McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Cecelia C Yates
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zhaoyan Qiang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina; Department of Pharmacology, Tianjin Medical University, Tianjin, China
| | - Samuel C Eaton
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina
| | - Jean Marie Mwiza
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Deepthi Tulasi
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina
| | - Jonathan C Schisler
- McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina; Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina
| | - Cam Patterson
- Presbyterian Hospital/Weill-Cornell Medical Center, New York, New York
| | - Tania Zaglia
- Department of Biomedical Sciences, University of Padova, Padova, Italy; Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy
| | - Marco Sandri
- Department of Biomedical Sciences, University of Padova, Padova, Italy; Dulbecco Telethon Institute, Padova, Italy
| | - Monte S Willis
- McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina; Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina; Indiana Center for Musculoskeletal Health and Department of Pathology and Laboratory Medicine, University of Indiana School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
29
|
Bcl-2-associated athanogene 3 (BAG3) is an enhancer of small heat shock protein turnover via activation of autophagy in the heart. Biochem Biophys Res Commun 2018; 496:1141-1147. [DOI: 10.1016/j.bbrc.2018.01.158] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 01/25/2018] [Indexed: 11/17/2022]
|
30
|
Myers VD, McClung JM, Wang J, Tahrir FG, Gupta MK, Gordon J, Kontos CH, Khalili K, Cheung JY, Feldman AM. The Multifunctional Protein BAG3: A Novel Therapeutic Target in Cardiovascular Disease. JACC Basic Transl Sci 2018; 3:122-131. [PMID: 29938246 PMCID: PMC6013050 DOI: 10.1016/j.jacbts.2017.09.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The B-cell lymphoma 2–associated anthanogene (BAG3) protein is expressed most prominently in the heart, the skeletal muscle, and in many forms of cancer. In the heart, it serves as a co-chaperone with heat shock proteins in facilitating autophagy; binds to B-cell lymphoma 2, resulting in inhibition of apoptosis; attaches actin to the Z disk, providing structural support for the sarcomere; and links the α-adrenergic receptor with the L-type Ca2+ channel. When BAG3 is overexpressed in cancer cells, it facilitates prosurvival pathways that lead to insensitivity to chemotherapy, metastasis, cell migration, and invasiveness. In contrast, in the heart, mutations in BAG3 have been associated with a variety of phenotypes, including both hypertrophic/restrictive and dilated cardiomyopathy. In murine skeletal muscle and vasculature, a mutation in BAG3 leads to critical limb ischemia after femoral artery ligation. An understanding of the biology of BAG3 is relevant because it may provide a therapeutic target in patients with both cardiac and skeletal muscle disease.
Collapse
Affiliation(s)
- Valerie D Myers
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Joseph M McClung
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - JuFang Wang
- Center for Translational Medicine, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Farzaneh G Tahrir
- Department of Neuroscience, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Manish K Gupta
- Department of Neuroscience, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Jennifer Gordon
- Department of Neuroscience, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Christopher H Kontos
- Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, North Carolina
| | - Kamel Khalili
- Department of Neuroscience, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Joseph Y Cheung
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine, Philadelphia, Pennsylvania.,Center for Translational Medicine, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Arthur M Feldman
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
31
|
Jensen BC, Parry TL, Huang W, Beak JY, Ilaiwy A, Bain JR, Newgard CB, Muehlbauer MJ, Patterson C, Johnson GL, Willis MS. Effects of the kinase inhibitor sorafenib on heart, muscle, liver and plasma metabolism in vivo using non-targeted metabolomics analysis. Br J Pharmacol 2017; 174:4797-4811. [PMID: 28977680 PMCID: PMC5727336 DOI: 10.1111/bph.14062] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 09/11/2017] [Accepted: 09/25/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE The human kinome consists of roughly 500 kinases, including 150 that have been proposed as therapeutic targets. Protein kinases regulate an array of signalling pathways that control metabolism, cell cycle progression, cell death, differentiation and survival. It is not surprising, then, that new kinase inhibitors developed to treat cancer, including sorafenib, also exhibit cardiotoxicity. We hypothesized that sorafenib cardiotoxicity is related to its deleterious effects on specific cardiac metabolic pathways given the critical roles of protein kinases in cardiac metabolism. EXPERIMENTAL APPROACH FVB/N mice (10 per group) were challenged with sorafenib or vehicle control daily for 2 weeks. Echocardiographic assessment of the heart identified systolic dysfunction consistent with cardiotoxicity in sorafenib-treated mice compared to vehicle-treated controls. Heart, skeletal muscle, liver and plasma were flash frozen and prepped for non-targeted GC-MS metabolomics analysis. KEY RESULTS Compared to vehicle-treated controls, sorafenib-treated hearts exhibited significant alterations in 11 metabolites, including markedly altered taurine/hypotaurine metabolism (25-fold enrichment), identified by pathway enrichment analysis. CONCLUSIONS AND IMPLICATIONS These studies identified alterations in taurine/hypotaurine metabolism in the hearts and skeletal muscles of mice treated with sorafenib. Interventions that rescue or prevent these sorafenib-induced changes, such as taurine supplementation, may be helpful in attenuating sorafenib-induced cardiac injury.
Collapse
Affiliation(s)
- Brian C Jensen
- McAllister Heart InstituteUniversity of North CarolinaChapel HillNCUSA
- Department of Internal MedicineDivision of Cardiology University of North CarolinaChapel HillNCUSA
- Department of PharmacologyUniversity of North CarolinaChapel HillNCUSA
| | - Traci L Parry
- McAllister Heart InstituteUniversity of North CarolinaChapel HillNCUSA
- Department of Pathology & Laboratory MedicineUniversity of North CarolinaChapel HillNCUSA
| | - Wei Huang
- McAllister Heart InstituteUniversity of North CarolinaChapel HillNCUSA
| | - Ju Youn Beak
- McAllister Heart InstituteUniversity of North CarolinaChapel HillNCUSA
| | - Amro Ilaiwy
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology InstituteDuke University Medical CenterDurhamNCUSA
- Division of Endocrinology, Metabolism, and Nutrition, Department of MedicineDuke University Medical CenterDurhamNCUSA
| | - James R Bain
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology InstituteDuke University Medical CenterDurhamNCUSA
- Division of Endocrinology, Metabolism, and Nutrition, Department of MedicineDuke University Medical CenterDurhamNCUSA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology InstituteDuke University Medical CenterDurhamNCUSA
- Division of Endocrinology, Metabolism, and Nutrition, Department of MedicineDuke University Medical CenterDurhamNCUSA
| | - Michael J Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology InstituteDuke University Medical CenterDurhamNCUSA
| | - Cam Patterson
- Presbyterian Hospital/Weill‐Cornell Medical CenterNew YorkNYUSA
| | - Gary L Johnson
- Department of PharmacologyUniversity of North CarolinaChapel HillNCUSA
| | - Monte S Willis
- McAllister Heart InstituteUniversity of North CarolinaChapel HillNCUSA
- Department of Pathology & Laboratory MedicineUniversity of North CarolinaChapel HillNCUSA
- Department of PharmacologyUniversity of North CarolinaChapel HillNCUSA
| |
Collapse
|
32
|
Meng Q, Bhandary B, Osinska H, James J, Xu N, Shay-Winkler K, Gulick J, Willis MS, Lander C, Robbins J. MMI-0100 Inhibits Cardiac Fibrosis in a Mouse Model Overexpressing Cardiac Myosin Binding Protein C. J Am Heart Assoc 2017; 6:JAHA.117.006590. [PMID: 28871043 PMCID: PMC5634300 DOI: 10.1161/jaha.117.006590] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Background Cardiac stress can trigger production of a 40‐kDa peptide fragment derived from the amino terminus of the cardiac myosin‐binding protein C. Cardiac stress, as well as cMyBP‐C mutations, can trigger production of 1 such truncated protein fragment, a 40‐kDa peptide fragment derived from the amino terminus of cMyBP‐C. Genetic expression of this 40‐kDa fragment in mouse cardiomyocytes (cMyBP‐C40k) leads to cardiac disease, fibrosis, and death within the first year. Fibrosis can occur in many cardiovascular diseases, and mitogen‐activated protein kinase––activated protein kinase‐2 signaling has been implicated in a variety of fibrotic processes. Recent studies demonstrated that mitogen‐activated protein kinase––activated protein kinase‐2 inhibition using the cell‐permeant peptide inhibitor MMI‐0100 is protective in the setting of acute myocardial infarction. We hypothesized that MMI‐0100 might also be protective in a chronic model of fibrosis, produced as a result of cMyBP‐C40k cardiomyocyte expression. Methods and Results Nontransgenic and cMyBP‐C40k inducible transgenic mice were given MMI‐0100 or PBS daily for 30 weeks. In control groups, long‐term MMI‐0100 was benign, with no measurable effects on cardiac anatomy, function, cell viability, hypertrophy, or probability of survival. In the inducible transgenic group, MMI‐0100 treatment reduced cardiac fibrosis, decreased cardiac hypertrophy, and prolonged survival. Conclusions Pharmaceutical inhibition of mitogen‐activated protein kinase––activated protein kinase‐2 signaling via MMI‐0100 treatment is beneficial in the context of fibrotic cMyBPC40k disease.
Collapse
Affiliation(s)
- Qinghang Meng
- Division of Molecular Cardiovascular Biology, The Heart Institute Cincinnati Children's Hospital, Cincinnati, OH
| | - Bidur Bhandary
- Division of Molecular Cardiovascular Biology, The Heart Institute Cincinnati Children's Hospital, Cincinnati, OH
| | - Hanna Osinska
- Division of Molecular Cardiovascular Biology, The Heart Institute Cincinnati Children's Hospital, Cincinnati, OH
| | - Jeanne James
- Children's Hospital of Wisconsin-Milwaukee Campus, Milwaukee, WI
| | - Na Xu
- Division of Molecular Cardiovascular Biology, The Heart Institute Cincinnati Children's Hospital, Cincinnati, OH
| | - Kritton Shay-Winkler
- Division of Molecular Cardiovascular Biology, The Heart Institute Cincinnati Children's Hospital, Cincinnati, OH
| | - James Gulick
- Division of Molecular Cardiovascular Biology, The Heart Institute Cincinnati Children's Hospital, Cincinnati, OH
| | - Monte S Willis
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC
| | | | - Jeffrey Robbins
- Division of Molecular Cardiovascular Biology, The Heart Institute Cincinnati Children's Hospital, Cincinnati, OH
| |
Collapse
|
33
|
Judge LM, Perez-Bermejo JA, Truong A, Ribeiro AJ, Yoo JC, Jensen CL, Mandegar MA, Huebsch N, Kaake RM, So PL, Srivastava D, Pruitt BL, Krogan NJ, Conklin BR. A BAG3 chaperone complex maintains cardiomyocyte function during proteotoxic stress. JCI Insight 2017; 2:94623. [PMID: 28724793 DOI: 10.1172/jci.insight.94623] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/13/2017] [Indexed: 12/13/2022] Open
Abstract
Molecular chaperones regulate quality control in the human proteome, pathways that have been implicated in many diseases, including heart failure. Mutations in the BAG3 gene, which encodes a co-chaperone protein, have been associated with heart failure due to both inherited and sporadic dilated cardiomyopathy. Familial BAG3 mutations are autosomal dominant and frequently cause truncation of the coding sequence, suggesting a heterozygous loss-of-function mechanism. However, heterozygous knockout of the murine BAG3 gene did not cause a detectable phenotype. To model BAG3 cardiomyopathy in a human system, we generated an isogenic series of human induced pluripotent stem cells (iPSCs) with loss-of-function mutations in BAG3. Heterozygous BAG3 mutations reduced protein expression, disrupted myofibril structure, and compromised contractile function in iPSC-derived cardiomyocytes (iPS-CMs). BAG3-deficient iPS-CMs were particularly sensitive to further myofibril disruption and contractile dysfunction upon exposure to proteasome inhibitors known to cause cardiotoxicity. We performed affinity tagging of the endogenous BAG3 protein and mass spectrometry proteomics to further define the cardioprotective chaperone complex that BAG3 coordinates in the human heart. Our results establish a model for evaluating protein quality control pathways in human cardiomyocytes and their potential as therapeutic targets and susceptibility factors for cardiac drug toxicity.
Collapse
Affiliation(s)
- Luke M Judge
- Department of Pediatrics, UCSF, San Francisco, California, USA.,Gladstone Institutes, San Francisco, California, USA
| | - Juan A Perez-Bermejo
- Gladstone Institutes, San Francisco, California, USA.,Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, California, USA
| | - Annie Truong
- Gladstone Institutes, San Francisco, California, USA
| | - Alexandre Js Ribeiro
- Gladstone Institutes, San Francisco, California, USA.,Stanford Cardiovascular Institute and Mechanical Engineering Department, and
| | - Jennie C Yoo
- Gladstone Institutes, San Francisco, California, USA
| | | | | | | | - Robyn M Kaake
- Gladstone Institutes, San Francisco, California, USA
| | - Po-Lin So
- Gladstone Institutes, San Francisco, California, USA
| | - Deepak Srivastava
- Department of Pediatrics, UCSF, San Francisco, California, USA.,Gladstone Institutes, San Francisco, California, USA
| | - Beth L Pruitt
- Stanford Cardiovascular Institute and Mechanical Engineering Department, and.,Bioengineering and Molecular and Cellular Physiology Departments, Stanford University, Stanford, California, USA
| | - Nevan J Krogan
- Gladstone Institutes, San Francisco, California, USA.,Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, California, USA
| | - Bruce R Conklin
- Gladstone Institutes, San Francisco, California, USA.,Department of Medicine, UCSF, San Francisco, California, USA
| |
Collapse
|
34
|
McClung JM, McCord TJ, Ryan TE, Schmidt CA, Green TD, Southerland KW, Reinardy JL, Mueller SB, Venkatraman TN, Lascola CD, Keum S, Marchuk DA, Spangenburg EE, Dokun A, Annex BH, Kontos CD. BAG3 (Bcl-2-Associated Athanogene-3) Coding Variant in Mice Determines Susceptibility to Ischemic Limb Muscle Myopathy by Directing Autophagy. Circulation 2017; 136:281-296. [PMID: 28442482 PMCID: PMC5537727 DOI: 10.1161/circulationaha.116.024873] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 04/14/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Critical limb ischemia is a manifestation of peripheral artery disease that carries significant mortality and morbidity risk in humans, although its genetic determinants remain largely unknown. We previously discovered 2 overlapping quantitative trait loci in mice, Lsq-1 and Civq-1, that affected limb muscle survival and stroke volume after femoral artery or middle cerebral artery ligation, respectively. Here, we report that a Bag3 variant (Ile81Met) segregates with tissue protection from hind-limb ischemia. METHODS We treated mice with either adeno-associated viruses encoding a control (green fluorescent protein) or 2 BAG3 (Bcl-2-associated athanogene-3) variants, namely Met81 or Ile81, and subjected the mice to hind-limb ischemia. RESULTS We found that the BAG3 Ile81Met variant in the C57BL/6 (BL6) mouse background segregates with protection from tissue necrosis in a shorter congenic fragment of Lsq-1 (C.B6-Lsq1-3). BALB/c mice treated with adeno-associated virus encoding the BL6 BAG3 variant (Ile81; n=25) displayed reduced limb-tissue necrosis and increased limb tissue perfusion compared with Met81- (n=25) or green fluorescent protein- (n=29) expressing animals. BAG3Ile81, but not BAG3Met81, improved ischemic muscle myopathy and muscle precursor cell differentiation and improved muscle regeneration in a separate, toxin-induced model of injury. Systemic injection of adeno-associated virus-BAG3Ile81 (n=9), but not BAG3Met81 (n=10) or green fluorescent protein (n=5), improved ischemic limb blood flow and limb muscle histology and restored muscle function (force production). Compared with BAG3Met81, BAG3Ile81 displayed improved binding to the small heat shock protein (HspB8) in ischemic skeletal muscle cells and enhanced ischemic muscle autophagic flux. CONCLUSIONS Taken together, our data demonstrate that genetic variation in BAG3 plays an important role in the prevention of ischemic tissue necrosis. These results highlight a pathway that preserves tissue survival and muscle function in the setting of ischemia.
Collapse
Affiliation(s)
- Joseph M McClung
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville.
| | - Timothy J McCord
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Terence E Ryan
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Cameron A Schmidt
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Tom D Green
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Kevin W Southerland
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Jessica L Reinardy
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Sarah B Mueller
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Talaignair N Venkatraman
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Christopher D Lascola
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Sehoon Keum
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Douglas A Marchuk
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Espen E Spangenburg
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Ayotunde Dokun
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Brian H Annex
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Christopher D Kontos
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| |
Collapse
|
35
|
Non-Targeted Metabolomics Analysis of the Effects of Tyrosine Kinase Inhibitors Sunitinib and Erlotinib on Heart, Muscle, Liver and Serum Metabolism In Vivo. Metabolites 2017. [PMID: 28640223 PMCID: PMC5618316 DOI: 10.3390/metabo7030031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: More than 90 tyrosine kinases have been implicated in the pathogenesis of malignant transformation and tumor angiogenesis. Tyrosine kinase inhibitors (TKIs) have emerged as effective therapies in treating cancer by exploiting this kinase dependency. The TKI erlotinib targets the epidermal growth factor receptor (EGFR), whereas sunitinib targets primarily vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR).TKIs that impact the function of non-malignant cells and have on- and off-target toxicities, including cardiotoxicities. Cardiotoxicity is very rare in patients treated with erlotinib, but considerably more common after sunitinib treatment. We hypothesized that the deleterious effects of TKIs on the heart were related to their impact on cardiac metabolism. Methods: Female FVB/N mice (10/group) were treated with therapeutic doses of sunitinib (40 mg/kg), erlotinib (50 mg/kg), or vehicle daily for two weeks. Echocardiographic assessment of the heart in vivo was performed at baseline and on Day 14. Heart, skeletal muscle, liver and serum were flash frozen and prepped for non-targeted GC-MS metabolomics analysis. Results: Compared to vehicle-treated controls, sunitinib-treated mice had significant decreases in systolic function, whereas erlotinib-treated mice did not. Non-targeted metabolomics analysis of heart identified significant decreases in docosahexaenoic acid (DHA), arachidonic acid (AA)/ eicosapentaenoic acid (EPA), O-phosphocolamine, and 6-hydroxynicotinic acid after sunitinib treatment. DHA was significantly decreased in skeletal muscle (quadriceps femoris), while elevated cholesterol was identified in liver and elevated ethanolamine identified in serum. In contrast, erlotinib affected only one metabolite (spermidine significantly increased). Conclusions: Mice treated with sunitinib exhibited systolic dysfunction within two weeks, with significantly lower heart and skeletal muscle levels of long chain omega-3 fatty acids docosahexaenoic acid (DHA), arachidonic acid (AA)/eicosapentaenoic acid (EPA) and increased serum O-phosphocholine phospholipid. This is the first link between sunitinib-induced cardiotoxicity and depletion of the polyunsaturated fatty acids (PUFAs) and inflammatory mediators DHA and AA/EPA in the heart. These compounds have important roles in maintaining mitochondrial function, and their loss may contribute to cardiac dysfunction.
Collapse
|
36
|
BIS overexpression does not affect the sensitivity of HEK 293T cells against apoptosis. Mol Cell Toxicol 2017. [DOI: 10.1007/s13273-017-0010-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
37
|
Garlid AO, Polson JS, Garlid KD, Hermjakob H, Ping P. Equipping Physiologists with an Informatics Tool Chest: Toward an Integerated Mitochondrial Phenome. Handb Exp Pharmacol 2017; 240:377-401. [PMID: 27995389 DOI: 10.1007/164_2016_93] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Understanding the complex involvement of mitochondrial biology in disease development often requires the acquisition, analysis, and integration of large-scale molecular and phenotypic data. An increasing number of bioinformatics tools are currently employed to aid in mitochondrial investigations, most notably in predicting or corroborating the spatial and temporal dynamics of mitochondrial molecules, in retrieving structural data of mitochondrial components, and in aggregating as well as transforming mitochondrial centric biomedical knowledge. With the increasing prevalence of complex Big Data from omics experiments and clinical cohorts, informatics tools have become indispensable in our quest to understand mitochondrial physiology and pathology. Here we present an overview of the various informatics resources that are helping researchers explore this vital organelle and gain insights into its form, function, and dynamics.
Collapse
Affiliation(s)
- Anders Olav Garlid
- The NIH BD2K Center of Excellence in Biomedical Computing at UCLA, Department of Physiology, University of California, Los Angeles, CA, 90095, USA.
| | - Jennifer S Polson
- The NIH BD2K Center of Excellence in Biomedical Computing at UCLA, Department of Physiology, University of California, Los Angeles, CA, 90095, USA.
| | - Keith D Garlid
- The NIH BD2K Center of Excellence in Biomedical Computing at UCLA, Department of Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Henning Hermjakob
- The NIH BD2K Center of Excellence in Biomedical Computing at UCLA, Department of Physiology, University of California, Los Angeles, CA, 90095, USA
- Molecular Systems Cluster, European Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | - Peipei Ping
- The NIH BD2K Center of Excellence in Biomedical Computing at UCLA, Departments of Physiology, Medicine, and Bioinformatics, University of California, Los Angeles, CA, 90095, USA
| |
Collapse
|
38
|
Jensen BC, Willis MS. The Head and the Heart. J Am Coll Cardiol 2016; 68:2408-2411. [DOI: 10.1016/j.jacc.2016.09.934] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 09/19/2016] [Indexed: 01/03/2023]
|