1
|
Shukla M, Narayan M. Proteostasis and Its Role in Disease Development. Cell Biochem Biophys 2025; 83:1725-1741. [PMID: 39422790 DOI: 10.1007/s12013-024-01581-6] [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] [Accepted: 09/22/2024] [Indexed: 10/19/2024]
Abstract
Proteostasis (protein homeostasis) refers to the general biological process that maintains the proper balance between the synthesis of proteins, their folding, trafficking, and degradation. It ensures proteins are functional, locally distributed, and appropriately folded inside cells. Genetic information enclosed in mRNA is translated into proteins. To ensure newly synthesized proteins take on the exact three-dimensional conformation, molecular chaperones assist in proper folding. Misfolded proteins can be refolded or targeted for elimination to stop aggregation. Cells utilize different degradation pathways, for instance, the ubiquitin-proteasome system, the autophagy-lysosome pathway, and the unfolded protein response, to degrade unwanted or damaged proteins. Quality control systems of the cell monitor the folding of proteins. These checkpoint mechanisms are aimed at degrading or refolding misfolded or damaged proteins. Under stress response pathways, such as heat shock response and unfolded protein response, which are triggered under conditions that perturb proteostasis, the capacity for folding is increased, and degradation pathways are activated to help cells handle stressful conditions. The deregulation of proteostasis is implicated in a variety of illnesses, comprising cancer, metabolic diseases, cardiovascular diseases, and neurological disorders. Therapeutic strategies with a deeper insight into the mechanism of proteostasis are crucial for the treatment of illnesses linked with proteostasis and to support cellular health. Thus, proteostasis is required not only for the maintenance of cellular homeostasis and function but also for proper protein function and prevention of injurious protein aggregation. In this review, we have covered the concept of proteostasis, its mechanism, and how disruptions to it can result in a number of disorders.
Collapse
Affiliation(s)
- Manisha Shukla
- Department of Biotechnology, Pandit S.N. Shukla University, Shahdol, Madhya Pradesh, India
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, University of Texas, El Paso, TX, USA.
| |
Collapse
|
2
|
Rohun J, Dudzik D, Raczak-Gutknecht J, Wabich E, Młodziński K, Markuszewski MJ, Daniłowicz-Szymanowicz L. Metabolomics in Atrial Fibrillation: Unlocking Novel Biomarkers and Pathways for Diagnosis, Prognosis, and Personalized Treatment. J Clin Med 2024; 14:34. [PMID: 39797116 PMCID: PMC11722095 DOI: 10.3390/jcm14010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Atrial fibrillation (AF) is the most frequent arrhythmia in the adult population associated with a high rate of severe consequences leading to significant morbidity and mortality worldwide. Therefore, its prompt recognition is of high clinical importance. AF detection often remains challenging due to unspecific symptoms and a lack of reliable biomarkers for its prediction. Herein, novel bioanalytical methodologies, such as metabolomics, offer new opportunities for a better understanding of the underlying pathological mechanisms of cardiovascular diseases, including AF. The metabolome, considered a complete set of small molecules present in the organism, directly reflects the current phenotype of the studied system and is highly sensitive to any changes, including arrhythmia's onset. A growing body of evidence suggests that metabolite profiling has prognostic value in AF prediction, highlighting its potential role not only in early diagnosis but also in guiding therapeutic interventions. By identifying specific metabolites as a disease biomarker or recognising particular metabolomic pathways involved in the AF pathomechanisms, metabolomics could be of great clinical value for further clinical decision-making, risk stratification, and an individual personalised approach. The presented narrative review aims to summarise the current state of knowledge on metabolomics in AF with a special emphasis on its implications for clinical practice and personalised medicine.
Collapse
Affiliation(s)
- Justyna Rohun
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Danuta Dudzik
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Joanna Raczak-Gutknecht
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Elżbieta Wabich
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Krzysztof Młodziński
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Michał J. Markuszewski
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Ludmiła Daniłowicz-Szymanowicz
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| |
Collapse
|
3
|
Burg S, Levi O, Elyagon S, Shapiro S, Murninkas M, Etzion S, Gradwohl G, Makarovsky D, Lichtenstein A, Gordon Y, Attali B, Etzion Y. The SK4 channel allosteric blocker, BA6b9, reduces atrial fibrillation substrate in rats with reduced ejection fraction. PNAS NEXUS 2024; 3:pgae192. [PMID: 38783894 PMCID: PMC11114471 DOI: 10.1093/pnasnexus/pgae192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
Atrial fibrillation (AF), the most common cardiac arrhythmia, is strongly associated with several comorbidities including heart failure (HF). AF in general, and specifically in the context of HF, is progressive in nature and associated with poor clinical outcomes. Current therapies for AF are limited in number and efficacy and do not target the underlying causes of atrial remodeling such as inflammation or fibrosis. We previously identified the calcium-activated SK4 K+ channels, which are preferentially expressed in the atria relative to the ventricles in both rat and human hearts, as attractive druggable target for AF treatment. Here, we examined the ability of BA6b9, a novel allosteric inhibitor of SK4 channels that targets the specific calmodulin-PIP2 binding domain, to alter AF susceptibility and atrial remodeling in a systolic HF rat postmyocardial infarction (post-MI) model. Daily BA6b9 injection (20 mg/kg/day) for 3 weeks starting 1-week post-MI prolonged the atrial effective refractory period, reduced AF induction and duration, and dramatically prevented atrial structural remodeling. In the post-MI left atrium (LA), pronounced upregulation of the SK4 K+ channel was observed, with corresponding increases in collagen deposition, α-SMA levels, and NLRP3 inflammasome expression. Strikingly, BA6b9 treatment reversed these changes while also significantly reducing the lateralization of the atrial connexin Cx43 in the LA of post-MI rats. Our findings indicate that the blockade of SK4 K+ channels using BA6b9 not only favors rhythm control but also remarkably reduces atrial structural remodeling, a property that is highly desirable for novel AF therapies, particularly in patients with comorbid HF.
Collapse
Affiliation(s)
- Shira Burg
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Or Levi
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Sigal Elyagon
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Shir Shapiro
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Michael Murninkas
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Sharon Etzion
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Gideon Gradwohl
- Medical Engineering Unit, The Jerusalem College of Technology, Jerusalem 9116001, Israel
| | - Daria Makarovsky
- Inter-Departmental Core Facility, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Alexandra Lichtenstein
- Inter-Departmental Core Facility, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yaara Gordon
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Bernard Attali
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yoram Etzion
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| |
Collapse
|
4
|
Yang Z, Cao Y, Kong L, Xi J, Liu S, Zhang J, Cheng W. Small molecules as modulators of the proteostasis machinery: Implication in cardiovascular diseases. Eur J Med Chem 2024; 264:116030. [PMID: 38071793 DOI: 10.1016/j.ejmech.2023.116030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/25/2023] [Accepted: 12/03/2023] [Indexed: 12/30/2023]
Abstract
With the escalating prevalence of cardiovascular diseases, the substantial socioeconomic burden on healthcare systems is intensifying. Accumulating empirical evidence underscores the pivotal role of the proteostasis network in regulating cardiac homeostasis and function. Disruptions in proteostasis may contribute to the loss of protein function or the acquisition of toxic functions, which are intricately linked to the development of cardiovascular ailments such as atrial fibrillation, heart failure, atherosclerosis, and cardiac aging. It is widely acknowledged that the proteostasis network encompasses molecular chaperones, autophagy, and the ubiquitin proteasome system (UPS). Consequently, the proteostasis network emerges as an appealing target for therapeutic interventions in cardiovascular diseases. Numerous small molecules, acting as modulators of the proteostasis machinery, have exhibited therapeutic efficacy in managing cardiovascular diseases. This review centers on elucidating the role of the proteostasis network in various cardiovascular diseases and explores the potential of small molecules as therapeutic agents.
Collapse
Affiliation(s)
- Zhiheng Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yu Cao
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, China
| | - Limin Kong
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Jianjun Xi
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, China
| | - Shourong Liu
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, China.
| | - Jiankang Zhang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
| | - Weiyan Cheng
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
5
|
Rutledge CA. Molecular mechanisms underlying sarcopenia in heart failure. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:7. [PMID: 38455513 PMCID: PMC10919908 DOI: 10.20517/jca.2023.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
The loss of skeletal muscle, also known as sarcopenia, is an aging-associated muscle disorder that is disproportionately present in heart failure (HF) patients. HF patients with sarcopenia have poor outcomes compared to the overall HF patient population. The prevalence of sarcopenia in HF is only expected to grow as the global population ages, and novel treatment strategies are needed to improve outcomes in this cohort. Multiple mechanistic pathways have emerged that may explain the increased prevalence of sarcopenia in the HF population, and a better understanding of these pathways may lead to the development of therapies to prevent muscle loss. This review article aims to explore the molecular mechanisms linking sarcopenia and HF, and to discuss treatment strategies aimed at addressing such molecular signals.
Collapse
Affiliation(s)
- Cody A. Rutledge
- Acute Medicine Section, Division of Medicine, Louis Stokes Cleveland Veteran Affairs Medical Center, Cleveland, OH 44106, USA
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
6
|
Benjamin EJ, Al‐Khatib SM, Desvigne‐Nickens P, Alonso A, Djoussé L, Forman DE, Gillis AM, Hendriks JML, Hills MT, Kirchhof P, Link MS, Marcus GM, Mehra R, Murray KT, Parkash R, Piña IL, Redline S, Rienstra M, Sanders P, Somers VK, Van Wagoner DR, Wang PJ, Cooper LS, Go AS. Research Priorities in the Secondary Prevention of Atrial Fibrillation: A National Heart, Lung, and Blood Institute Virtual Workshop Report. J Am Heart Assoc 2021; 10:e021566. [PMID: 34351783 PMCID: PMC8475065 DOI: 10.1161/jaha.121.021566] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/28/2021] [Indexed: 12/24/2022]
Abstract
There has been sustained focus on the secondary prevention of coronary heart disease and heart failure; yet, apart from stroke prevention, the evidence base for the secondary prevention of atrial fibrillation (AF) recurrence, AF progression, and AF-related complications is modest. Although there are multiple observational studies, there are few large, robust, randomized trials providing definitive effective approaches for the secondary prevention of AF. Given the increasing incidence and prevalence of AF nationally and internationally, the AF field needs transformative research and a commitment to evidenced-based secondary prevention strategies. We report on a National Heart, Lung, and Blood Institute virtual workshop directed at identifying knowledge gaps and research opportunities in the secondary prevention of AF. Once AF has been detected, lifestyle changes and novel models of care delivery may contribute to the prevention of AF recurrence, AF progression, and AF-related complications. Although benefits seen in small subgroups, cohort studies, and selected randomized trials are impressive, the widespread effectiveness of AF secondary prevention strategies remains unknown, calling for development of scalable interventions suitable for diverse populations and for identification of subpopulations who may particularly benefit from intensive management. We identified critical research questions for 6 topics relevant to the secondary prevention of AF: (1) weight loss; (2) alcohol intake, smoking cessation, and diet; (3) cardiac rehabilitation; (4) approaches to sleep disorders; (5) integrated, team-based care; and (6) nonanticoagulant pharmacotherapy. Our goal is to stimulate innovative research that will accelerate the generation of the evidence to effectively pursue the secondary prevention of AF.
Collapse
Affiliation(s)
- Emelia J. Benjamin
- Cardiovascular MedicineDepartment of MedicineBoston University School of MedicineBostonMA
- Department of EpidemiologyBoston University School of Public HealthBostonMA
| | - Sana M. Al‐Khatib
- Division of Cardiology and Duke Clinical Research InstituteDuke University Medical CenterDurhamNC
| | - Patrice Desvigne‐Nickens
- Division of Cardiovascular SciencesNational Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Alvaro Alonso
- Department of EpidemiologyRollins School of Public HealthEmory UniversityAtlantaGA
| | - Luc Djoussé
- Division of AgingDepartment of MedicineBrigham and Women’s Hospital and Harvard Medical SchoolBostonMA
| | - Daniel E. Forman
- Divisions of Geriatrics and CardiologyUniversity of Pittsburgh Medical CenterAging InstituteUniversity of PittsburghVA Pittsburgh Healthcare SystemPittsburghPA
| | - Anne M. Gillis
- Libin Cardiovascular Institute of AlbertaUniversity of CalgaryAlbertaCanada
| | - Jeroen M. L. Hendriks
- Centre for Heart Rhythm DisordersUniversity of Adelaide, and Royal Adelaide HospitalAdelaideAustralia
- Caring Futures InstituteCollege of Nursing and Health SciencesFlinders UniversityAdelaideAustralia
| | | | - Paulus Kirchhof
- Department of CardiologyUniversity Heart and Vascular Center UKE HamburgHamburgGermany
- Institute of Cardiovascular ScienceUniversity of BirminghamUnited Kingdom
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/LübeckBerlinGermany
- AFNETMünsterGermany
| | - Mark S. Link
- Division of CardiologyDepartment of MedicineUT Southwestern Medical CenterDallasTX
| | - Gregory M. Marcus
- Division of CardiologyUniversity of California, San FranciscoSan FranciscoCA
| | - Reena Mehra
- Sleep Disorders CenterNeurologic InstituteRespiratory InstituteHeart and Vascular Institute, and Molecular Cardiology Department of the Lerner Research InstituteCleveland ClinicClevelandOH
| | | | - Ratika Parkash
- Division of CardiologyQEII Health Sciences Center/Dalhousie UniversityHalifaxNova ScotiaCanada
| | - Ileana L. Piña
- Wayne State UniversityDetroitMI
- Central Michigan UniversityMt PleasantMI
- FDAOPEQCenter for Devices and Radiological HealthSilver SpringMD
| | - Susan Redline
- Department of MedicineBrigham and Women’s HospitalBostonMA
| | - Michiel Rienstra
- Department of CardiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Prashanthan Sanders
- Centre for Heart Rhythm DisordersUniversity of Adelaide, and Royal Adelaide HospitalAdelaideAustralia
| | | | | | - Paul J. Wang
- Stanford University School of MedicinePalo AltoCA
| | - Lawton S. Cooper
- Division of Cardiovascular SciencesNational Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Alan S. Go
- Division of ResearchKaiser Permanente Northern CaliforniaOaklandCA
- Department of Health System ScienceKaiser Permanente Bernard J. Tyson School of MedicinePasadenaCA
- Departments of Epidemiology, Biostatistics and MedicineUniversity of California, San FranciscoSan FranciscoCA
- Departments of MedicineHealth Research and PolicyStanford UniversityStanfordCA
| |
Collapse
|
7
|
Xintarakou A, Tzeis S, Psarras S, Asvestas D, Vardas P. Atrial fibrosis as a dominant factor for the development of atrial fibrillation: facts and gaps. Europace 2021; 22:342-351. [PMID: 31998939 DOI: 10.1093/europace/euaa009] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/03/2020] [Indexed: 01/08/2023] Open
Abstract
Atrial fibrillation (AF), the most commonly diagnosed arrhythmia, affects a notable percentage of the population and constitutes a major risk factor for thromboembolic events and other heart-related conditions. Fibrosis plays an important role in the onset and perpetuation of AF through structural and electrical remodelling processes. Multiple molecular pathways are involved in atrial substrate modification and the subsequent maintenance of AF. In this review, we aim to recapitulate underlying molecular pathways leading to atrial fibrosis and to indicate existing gaps in the complex interplay of atrial fibrosis and AF.
Collapse
Affiliation(s)
| | - Stylianos Tzeis
- Cardiology Department, Mitera General Hospital, Hygeia Group, Athens, Greece
| | - Stelios Psarras
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Greece
| | - Dimitrios Asvestas
- Cardiology Department, Mitera General Hospital, Hygeia Group, Athens, Greece
| | - Panos Vardas
- Heart Sector, Hygeia Hospitals Group, 5, Erithrou Stavrou, Marousi, Athens 15123, Greece
| |
Collapse
|
8
|
Diteepeng T, Del Monte F, Luciani M. The long and winding road to target protein misfolding in cardiovascular diseases. Eur J Clin Invest 2021; 51:e13504. [PMID: 33527342 DOI: 10.1111/eci.13504] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/18/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND In the last decades, cardiovascular diseases (CVD) have remained the first leading cause of mortality and morbidity in the world. Although several therapeutic approaches have been introduced in the past, the development of novel treatments remains an important research goal, which is hampered by the lack of understanding of key mechanisms and targets. Emerging evidences in recent years indicate the involvement of misfolded proteins aggregation and the derailment of protein quality control in the pathogenesis of cardiovascular diseases. Several potential interventions targeting protein quality control have been translated from the bench to the bedside to effectively employ the misfolded proteins as promising therapeutic targets for cardiac diseases, but with trivial results. DESIGN In this review, we describe the recent progresses in preclinical and clinical studies of protein misfolding and compromised protein quality control by selecting and reporting studies focusing on cardiovascular diseases including cardiomyopathies, cardiac amyloidosis, atherosclerosis, atrial fibrillation and thrombosis. RESULTS In preclinical models, modulators of several molecular targets (eg heat shock proteins, unfolded protein response, ubiquitin protein system, autophagy and histone deacetylases) have been tested in various conditions with promising results although lacking an adequate transition towards clinical setting. CONCLUSIONS At present, no therapeutic strategies have been reported to attenuate proteotoxicity in patients with CVD due to a lack of specific biomarkers for pinpointing upstream events in protein folding defects at a subclinical stage of the diseases requiring an intensive collaboration between basic scientists and clinicians.
Collapse
Affiliation(s)
- Thamonwan Diteepeng
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Federica Del Monte
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC, USA.,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna Alma Mater, Bologna, Italy
| | - Marco Luciani
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.,Department of Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| |
Collapse
|
9
|
Role of Oxidative DNA Damage and Repair in Atrial Fibrillation and Ischemic Heart Disease. Int J Mol Sci 2021; 22:ijms22083838. [PMID: 33917194 PMCID: PMC8068079 DOI: 10.3390/ijms22083838] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023] Open
Abstract
Atrial fibrillation (AF) and ischemic heart disease (IHD) represent the two most common clinical cardiac diseases, characterized by angina, arrhythmia, myocardial damage, and cardiac dysfunction, significantly contributing to cardiovascular morbidity and mortality and posing a heavy socio-economic burden on society worldwide. Current treatments of these two diseases are mainly symptomatic and lack efficacy. There is thus an urgent need to develop novel therapies based on the underlying pathophysiological mechanisms. Emerging evidence indicates that oxidative DNA damage might be a major underlying mechanism that promotes a variety of cardiac diseases, including AF and IHD. Antioxidants, nicotinamide adenine dinucleotide (NAD+) boosters, and enzymes involved in oxidative DNA repair processes have been shown to attenuate oxidative damage to DNA, making them potential therapeutic targets for AF and IHD. In this review, we first summarize the main molecular mechanisms responsible for oxidative DNA damage and repair both in nuclei and mitochondria, then describe the effects of oxidative DNA damage on the development of AF and IHD, and finally discuss potential targets for oxidative DNA repair-based therapeutic approaches for these two cardiac diseases.
Collapse
|
10
|
Role of HDACs in cardiac electropathology: Therapeutic implications for atrial fibrillation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118459. [DOI: 10.1016/j.bbamcr.2019.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 03/07/2019] [Accepted: 03/13/2019] [Indexed: 12/21/2022]
|
11
|
Li J, Zhang D, Brundel BJJM, Wiersma M. Imbalance of ER and Mitochondria Interactions: Prelude to Cardiac Ageing and Disease? Cells 2019; 8:cells8121617. [PMID: 31842269 PMCID: PMC6952992 DOI: 10.3390/cells8121617] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022] Open
Abstract
Cardiac disease is still the leading cause of morbidity and mortality worldwide, despite some exciting and innovative improvements in clinical management. In particular, atrial fibrillation (AF) and heart failure show a steep increase in incidence and healthcare costs due to the ageing population. Although research revealed novel insights in pathways driving cardiac disease, the exact underlying mechanisms have not been uncovered so far. Emerging evidence indicates that derailed proteostasis (i.e., the homeostasis of protein expression, function and clearance) is a central component driving cardiac disease. Within proteostasis derailment, key roles for endoplasmic reticulum (ER) and mitochondrial stress have been uncovered. Here, we describe the concept of ER and mitochondrial stress and the role of interactions between the ER and mitochondria, discuss how imbalance in the interactions fuels cardiac ageing and cardiac disease (including AF), and finally assess the potential of drugs directed at conserving the interaction as an innovative therapeutic target to improve cardiac function.
Collapse
Affiliation(s)
- Jin Li
- Correspondence: (J.L.); (M.W.)
| | | | | | | |
Collapse
|
12
|
Dorsch LM, Schuldt M, Knežević D, Wiersma M, Kuster DWD, van der Velden J, Brundel BJJM. Untying the knot: protein quality control in inherited cardiomyopathies. Pflugers Arch 2018; 471:795-806. [PMID: 30109411 PMCID: PMC6475634 DOI: 10.1007/s00424-018-2194-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/06/2018] [Indexed: 12/27/2022]
Abstract
Mutations in genes encoding sarcomeric proteins are the most important causes of inherited cardiomyopathies, which are a major cause of mortality and morbidity worldwide. Although genetic screening procedures for early disease detection have been improved significantly, treatment to prevent or delay mutation-induced cardiac disease onset is lacking. Recent findings indicate that loss of protein quality control (PQC) is a central factor in the disease pathology leading to derailment of cellular protein homeostasis. Loss of PQC includes impairment of heat shock proteins, the ubiquitin-proteasome system, and autophagy. This may result in accumulation of misfolded and aggregation-prone mutant proteins, loss of sarcomeric and cytoskeletal proteins, and, ultimately, loss of cardiac function. PQC derailment can be a direct effect of the mutation-induced activation, a compensatory mechanism due to mutation-induced cellular dysfunction or a consequence of the simultaneous occurrence of the mutation and a secondary hit. In this review, we discuss recent mechanistic findings on the role of proteostasis derailment in inherited cardiomyopathies, with special focus on sarcomeric gene mutations and possible therapeutic applications.
Collapse
Affiliation(s)
- Larissa M Dorsch
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| | - Maike Schuldt
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| | - Dora Knežević
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Marit Wiersma
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Diederik W D Kuster
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Bianca J J M Brundel
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Selection and evaluation of reference genes for expression analysis using quantitative real-time PCR in the Asian Ladybird Harmonia axyridis (Coleoptera: Coccinellidae). PLoS One 2018; 13:e0192521. [PMID: 29889877 PMCID: PMC5995347 DOI: 10.1371/journal.pone.0192521] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 12/28/2017] [Indexed: 01/31/2023] Open
Abstract
Harmonia axyridis (Coleoptera: Coccinellidae) is a polyphagous insect that is an important biological agent used to control agricultural and forestry pests. The role of functional genes in H. axyridis based on quantitative real-time PCR (qRT-PCR) is increasingly well understood to investigate biology, physiology, feeding behavior and the role of important genes in physiological processes. Quantitative real-time PCR (qRT-PCR) is a powerful and reliable technique to quantify gene expression. Using qRT-PCR, expression levels of target genes are determined based on the levels of internal reference genes; therefore, reference genes need to be stably expressed under specific experimental conditions. However, there have been no studies on the stability of reference genes used in H. axyridis. In this study, we systematically investigated expression profiles of nine candidate reference genes from H. axyridis, including β-actin (ACTIN); elongation factor 1 α (EF1A); ribosomal proteins L10, L18, L28, S13, and S15 (RPL10, RPL18, RPL28, RPS13 and RPS15); glyceraldehyde-3-phosphate dehydrogenase (GAPDH); and superoxide dismutase (SOD). Four analytical methods (geNorm, BestKeeper, NormFinder, and the ΔCt method) were used to evaluate the suitability of these genes as internal reference genes for three biotic factors (developmental stage, tissue, and sex) and two abiotic treatments (temperature and photoperiod). RefFinder, a comprehensive evaluation platform integrating the four analytical methods, was used to rank the overall stability of these reference genes. Among the nine candidate genes, different reference genes were identified as having the most stable expression across biotic and abiotic factors. Genes encoding ribosomal proteins typically had the most stable expression, though EF1A was the most stable across developmental stages and photoperiods. To validate the suitability of these reference genes, heat shock protein 90 (HSP90) was chosen as a target. Significant up-regulation in HSP90 expression level in response to both low and high temperature was observed when using the most suitable reference genes but not when using an arbitrarily selected reference gene. The reference genes identified in this study will provide the basis for future functional genomics research in H. axyridis and will also facilitate the establishment of a standardized qRT-PCR program for other related insects.
Collapse
|
14
|
Wiersma M, Beuren TMA, de Vrij EL, Reitsema VA, Bruintjes JJ, Bouma HR, Brundel BJJM, Henning RH. Torpor-arousal cycles in Syrian hamster heart are associated with transient activation of the protein quality control system. Comp Biochem Physiol B Biochem Mol Biol 2018; 223:23-28. [PMID: 29894736 DOI: 10.1016/j.cbpb.2018.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/28/2018] [Accepted: 06/04/2018] [Indexed: 12/14/2022]
Abstract
Hibernation consists of torpor, with marked suppression of metabolism and physiological functions, alternated with arousal periods featuring their full restoration. The heart is particularly challenged, exemplified by its rate reduction from 400 to 5-10 beats per minute during torpor in Syrian hamsters. In addition, during arousals, the heart needs to accommodate the very rapid return to normal function, which lead to our hypothesis that cardiac function during hibernation is supported by maintenance of protein homeostasis through adaptations in the protein quality control (PQC) system. Hereto, we examined autophagy, the endoplasmic reticulum (ER) unfolded protein (UPRER) response and the heat shock response (HSR) in Syrian hamster hearts during torpor and arousal. Transition from torpor to arousal (1.5 h) was associated with stimulation of the PQC system during early arousal, demonstrated by induction of autophagosomes, as shown by an increase in LC3B-II protein abundance, likely related to the activation of the UPRER during late torpor in response to proteotoxic stress. The HSR was not activated during torpor or arousal. Our results demonstrate activation of the cardiac PQC system - particularly autophagosomal degradation - in early arousal in response to cardiac stress, to clear excess aberrant or damaged proteins, being gradually formed during the torpor bout and/or the rapid increase in heart rate during the transition from torpor to arousal. This mechanism may enable the large gain in cardiac function during the transition from torpor to arousal, which may hold promise to further understand 'hibernation' of cardiomyocytes in human heart disease.
Collapse
Affiliation(s)
- Marit Wiersma
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands; Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, The Netherlands.
| | - Thais M A Beuren
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands; Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Edwin L de Vrij
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, The Netherlands; Department of Surgery, Martini Hospital, Groningen, The Netherlands
| | - Vera A Reitsema
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Jantje J Bruintjes
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Hjalmar R Bouma
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, The Netherlands; Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands; Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Robert H Henning
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, The Netherlands
| |
Collapse
|
15
|
Abstract
The incidence and prevalence of cardiac diseases, which are the main cause of death worldwide, are likely to increase because of population ageing. Prevailing theories about the mechanisms of ageing feature the gradual derailment of cellular protein homeostasis (proteostasis) and loss of protein quality control as central factors. In the heart, loss of protein patency, owing to flaws in genetically-determined design or because of environmentally-induced 'wear and tear', can overwhelm protein quality control, thereby triggering derailment of proteostasis and contributing to cardiac ageing. Failure of protein quality control involves impairment of chaperones, ubiquitin-proteosomal systems, autophagy, and loss of sarcomeric and cytoskeletal proteins, all of which relate to induction of cardiomyocyte senescence. Targeting protein quality control to maintain cardiac proteostasis offers a novel therapeutic strategy to promote cardiac health and combat cardiac disease. Currently marketed drugs are available to explore this concept in the clinical setting.
Collapse
Affiliation(s)
- Robert H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, De Boelelaan 1117, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
16
|
Wang HJ, Shi ZK, Shen QD, Xu CD, Wang B, Meng ZJ, Wang SG, Tang B, Wang S. Molecular Cloning and Induced Expression of Six Small Heat Shock Proteins Mediating Cold-Hardiness in Harmonia axyridis (Coleoptera: Coccinellidae). Front Physiol 2017; 8:60. [PMID: 28232804 PMCID: PMC5299025 DOI: 10.3389/fphys.2017.00060] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 01/23/2017] [Indexed: 12/18/2022] Open
Abstract
The main function of small heat shock proteins (sHSPs) as molecular chaperones is to protect proteins from denaturation under adverse conditions. Molecular and physiological data were used to examine the sHSPs underlying cold-hardiness in Harmonia axyridis. Complementary DNA sequences were obtained for six H. axyridis sHSPs based on its transcriptome, and the expression of the genes coding for these sHSPs was evaluated by quantitative real-time PCR (qRT-PCR) in several developmental stages, under short-term cooling or heating conditions, and in black and yellow females of experimental and overwintering populations under low-temperature storage. In addition, we measured water content and the super cooling and freezing points (SCP and FP, respectively) of H. axyridis individuals from experimental and overwintering populations. The average water content was not significantly different between adults of both populations, but the SCP and FP of the overwintering population were significantly lower than that of the experimental population. Overall, the six sHSPs genes showed different expression patterns among developmental stages. In the short-term cooling treatment, Hsp16.25 and Hsp21.00 expressions first increased and then decreased, while Hsp10.87 and Hsp21.56 expressions increased during the entire process. Under short-term heating, the expressions of Hsp21.00, Hsp21.62, Hsp10.87, and Hsp16.25 showed an increasing trend, whereas Hsp36.77 first decreased and then increased. Under low-temperature storage conditions, the expression of Hsp36.77 decreased, while the expressions of Hsp21.00 and Hsp21.62 were higher than that of the control group in the experimental population. The expression of Hsp36.77 first increased and then decreased, whereas Hsp21.56 expression was always higher than that of the control group in the overwintering population. Thus, differences in sHSPs gene expression were correlated with the H. axyridis forms, suggesting that the mechanism of cold resistance might differ among them. Although, Hsp36.77, Hsp16.25, Hsp21.00, and Hsp21.62 regulated cold- hardiness, the only significant differences between overwintering and experimental populations were found for Hsp16.25 and Hsp21.00.
Collapse
Affiliation(s)
- Hui-Juan Wang
- College of Life and Environmental Sciences, Hangzhou Normal UniversityHangzhou, China
| | - Zuo-Kun Shi
- College of Life and Environmental Sciences, Hangzhou Normal UniversityHangzhou, China
| | - Qi-Da Shen
- College of Life and Environmental Sciences, Hangzhou Normal UniversityHangzhou, China
| | - Cai-Di Xu
- College of Life and Environmental Sciences, Hangzhou Normal UniversityHangzhou, China
| | - Bing Wang
- Institute of Plant and Environment Protection, Beijing Academy of Agriculture and Forestry SciencesBeijing, China
| | - Zhao-Jun Meng
- College of Forestry, Northeast Forestry UniversityHarbin, China
| | - Shi-Gui Wang
- College of Life and Environmental Sciences, Hangzhou Normal UniversityHangzhou, China
| | - Bin Tang
- College of Life and Environmental Sciences, Hangzhou Normal UniversityHangzhou, China
| | - Su Wang
- Institute of Plant and Environment Protection, Beijing Academy of Agriculture and Forestry SciencesBeijing, China
| |
Collapse
|
17
|
Zhang D, Hu X, Henning RH, Brundel BJJM. Keeping up the balance: role of HDACs in cardiac proteostasis and therapeutic implications for atrial fibrillation. Cardiovasc Res 2015; 109:519-26. [PMID: 26645980 DOI: 10.1093/cvr/cvv265] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 10/29/2015] [Indexed: 12/16/2022] Open
Abstract
Cardiomyocytes are long-lived post-mitotic cells with limited regenerative capacity. Proper cardiomyocyte function depends critically on the maintenance of a healthy homeostasis of protein expression, folding, assembly, trafficking, function, and degradation, together commonly referred to as proteostasis. Impairment of proteostasis has a prominent role in the pathophysiology of ageing-related neurodegenerative diseases including Huntington's, Parkinson's, and Alzheimer's disease. Emerging evidence reveals also a role for impaired proteostasis in the pathophysiology of common human cardiac diseases such as cardiac hypertrophy, dilated and ischaemic cardiomyopathies, and atrial fibrillation (AF). Histone deacetylases (HDACs) have recently been recognized as key modulators which control cardiac proteostasis by deacetylating various proteins. By deacetylating chromatin proteins, including histones, HDACs modulate epigenetic regulation of pathological gene expression. Also, HDACs exert a broad range of functions outside the nucleus by deacetylating structural and contractile proteins. The cytosolic actions of HDACs result in changed protein function through post-translational modifications and/or modulation of their degradation. This review describes the mechanisms underlying the derailment of proteostasis in AF and subsequently focuses on the role of HDACs herein. In addition, the therapeutic potential of HDAC inhibition to maintain a healthy proteostasis resulting in a delay in AF onset and progression is discussed.
Collapse
Affiliation(s)
- Deli Zhang
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, PO Box 30 001, 9700RB Groningen, The Netherlands
| | - Xu Hu
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, PO Box 30 001, 9700RB Groningen, The Netherlands
| | - Robert H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, PO Box 30 001, 9700RB Groningen, The Netherlands
| | - Bianca J J M Brundel
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, PO Box 30 001, 9700RB Groningen, The Netherlands Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Lanters EAH, van Marion DMS, Kik C, Steen H, Bogers AJJC, Allessie MA, Brundel BJJM, de Groot NMS. HALT & REVERSE: Hsf1 activators lower cardiomyocyt damage; towards a novel approach to REVERSE atrial fibrillation. J Transl Med 2015; 13:347. [PMID: 26541406 PMCID: PMC4635598 DOI: 10.1186/s12967-015-0714-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 10/23/2015] [Indexed: 11/22/2022] Open
Abstract
Background Atrial fibrillation is a progressive arrhythmia, the exact mechanism underlying the progressive nature of recurrent AF episodes is still unknown. Recently, it was found that key players of the protein quality control system of the cardiomyocyte, i.e. Heat Shock Proteins, protect against atrial fibrillation progression by attenuating atrial electrical and structural remodeling (electropathology). HALT & REVERSE aims to investigate the correlation between electropathology, as defined by endo- or epicardial mapping, Heat Shock Protein levels and development or recurrence of atrial fibrillation following pulmonary vein isolation, or electrical cardioversion or cardiothoracic surgery. Study design This study is a prospective observational study. Three separate study groups are defined: (1) cardiothoracic surgery, (2) pulmonary vein isolation and (3) electrical cardioversion. An intra-operative high-resolution epicardial (group 1) or endocardial (group 2) mapping procedure of the atria is performed to study atrial electropathology. Blood samples for Heat Shock Protein determination are obtained at baseline and during the follow-up period at 3 months (group 2), 6 months (groups 1 and 2) and 1 year (group 1 and 2). Tissue samples of the right and left atrial appendages in patients in group 1 are analysed for Heat Shock Protein levels and for tissue characteristics. Early post procedural atrial fibrillation is detected by continuous rhythm monitoring, whereas late post procedural atrial fibrillation is documented by either electrocardiogram or 24-h Holter registration. Conclusion HALT & REVERSE aims to identify the correlation between Heat Shock Protein levels and degree of electropathology. The study outcome will contribute to novel diagnostic tools for the early recognition of clinical atrial fibrillation. Trial Registrations: Rotterdam Medical Ethical Committee MEC-2014-393, Dutch Trial Registration NTR4658
Collapse
Affiliation(s)
- Eva A H Lanters
- Department of Cardiology, Ba 579, Erasmus Medical Center, 's Gravendijkwal 230, 3015, CE, Rotterdam, The Netherlands.
| | - Denise M S van Marion
- Department of Clinical Pharmacy and Pharmacology, EB71, University Institute for Drug Exploration (GUIDE), University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30 001, 9700, RB, Groningen, The Netherlands.
| | - Charles Kik
- Department of Cardiothoracic Surgery, Bd 557, Erasmus Medical Center, 's Gravendijkwal 230, 3015, CE, Rotterdam, The Netherlands.
| | - Herman Steen
- Nyken Therapeutics B.V., L.J. Zielstraweg 2, 9713, GX, Groningen, The Netherlands.
| | - Ad J J C Bogers
- Department of Cardiothoracic Surgery, Bd 557, Erasmus Medical Center, 's Gravendijkwal 230, 3015, CE, Rotterdam, The Netherlands.
| | - Maurits A Allessie
- Department of Cardiology, Ba 579, Erasmus Medical Center, 's Gravendijkwal 230, 3015, CE, Rotterdam, The Netherlands.
| | - Bianca J J M Brundel
- Department of Clinical Pharmacy and Pharmacology, EB71, University Institute for Drug Exploration (GUIDE), University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30 001, 9700, RB, Groningen, The Netherlands. .,Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Van der Boechorststraat 7, 1081, BT, Amsterdam, The Netherlands.
| | - Natasja M S de Groot
- Department of Cardiology, Ba 579, Erasmus Medical Center, 's Gravendijkwal 230, 3015, CE, Rotterdam, The Netherlands.
| |
Collapse
|
19
|
van Marion DMS, Lanters EAH, Wiersma M, Allessie MA, Brundel BBJJM, de Groot NMS. Diagnosis and Therapy of Atrial Fibrillation: The Past, The Present and The Future. J Atr Fibrillation 2015; 8:1216. [PMID: 27957185 DOI: 10.4022/jafib.1216] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 07/05/2015] [Accepted: 01/10/2015] [Indexed: 02/03/2023]
Abstract
Atrial fibrillation (AF) is the most common age-related cardiac arrhythmia. It is a progressive disease, which makes treatment difficult. The progression of AF is caused by the accumulation of damage in cardiomyocytes which makes the atria more vulnerable for AF. Especially structural remodeling and electrical remodeling, together called electropathology are sustainable in the atria and impair functional recovery to sinus rhythm after cardioversion. The exact electropathological mechanisms underlying persistence of AF are at present unknown. High resolution wavemapping studies in patients with different types of AF showed that longitudinal dissociation in conduction and epicardial breakthrough were the key elements of the substrate of longstanding persistent AF. A double layer of electrically dissociated waves propagating transmurally can explain persistence of AF (Double Layer Hypothesis) but the molecular mechanism is unknown. Derailment of proteasis -defined as the homeostasis in protein synthesis, folding, assembly, trafficking, guided by chaperones, and clearance by protein degradation systems - may play an important role in remodeling of the cardiomyocyte. As current therapies are not effective in attenuating AF progression, step-by-step analysis of this process, in order to identify potential targets for drug therapy, is essential. In addition, novel mapping approaches enabling assessment of the degree of electropathology in the individual patient are mandatory to develop patient-tailored therapies. The aims of this review are to 1) summarize current knowledge of the electrical and molecular mechanisms underlying AF 2) discuss the shortcomings of present diagnostic instruments and therapeutic options and 3) to present potential novel diagnostic tools and therapeutic targets.
Collapse
Affiliation(s)
- Denise M S van Marion
- Department of Clinical Pharmacy and Pharmacology, University Institute for Drug Exploration (GUIDE), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Eva A H Lanters
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marit Wiersma
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Maurits A Allessie
- Department of Clinical Pharmacy and Pharmacology, University Institute for Drug Exploration (GUIDE), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bianca B J J M Brundel
- Department of Clinical Pharmacy and Pharmacology, University Institute for Drug Exploration (GUIDE), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Clinical Pharmacy and Pharmacology, University Institute for Drug Exploration (GUIDE), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Natasja M S de Groot
- Department of Clinical Pharmacy and Pharmacology, University Institute for Drug Exploration (GUIDE), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
20
|
Progress toward the prevention and treatment of atrial fibrillation: A summary of the Heart Rhythm Society Research Forum on the Treatment and Prevention of Atrial Fibrillation, Washington, DC, December 9-10, 2013. Heart Rhythm 2014; 12:e5-e29. [PMID: 25460864 DOI: 10.1016/j.hrthm.2014.11.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Indexed: 02/07/2023]
|
21
|
Gemel J, Simon AR, Patel D, Xu Q, Matiukas A, Veenstra RD, Beyer EC. Degradation of a connexin40 mutant linked to atrial fibrillation is accelerated. J Mol Cell Cardiol 2014; 74:330-9. [PMID: 24973497 PMCID: PMC4135452 DOI: 10.1016/j.yjmcc.2014.06.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 06/12/2014] [Accepted: 06/18/2014] [Indexed: 12/31/2022]
Abstract
Several Cx40 mutants have been identified in patients with atrial fibrillation (AF). We have been working to identify physiological or cell biological abnormalities of several of these human mutants that might explain how they contribute to disease pathogenesis. Wild type (wt) Cx40 or four different mutants (P88S, G38D, V85I, and L229M) were expressed by the transfection of communication-deficient HeLa cells or HL-1 cardiomyocytes. Biophysical channel properties and the sub-cellular localization and protein levels of Cx40 were characterized. Wild type Cx40 and all mutants except P88S formed gap junction plaques and induced significant gap junctional conductances. The functional mutants showed only modest alterations of single channel conductances or gating by trans-junctional voltage as compared to wtCx40. However, immunoblotting indicated that the steady state levels of G38D, V85I, and L229M were reduced relative to wtCx40; most strikingly, G38D was only 20-31% of wild type levels. After the inhibition of protein synthesis with cycloheximide, G38D (and to a lesser extent the other mutants) disappeared much faster than wtCx40. Treatment with the proteasomal inhibitor, epoxomicin, greatly increased levels of G38D and restored the abundance of gap junctions and the extent of intercellular dye transfer. Thus, G38D, V85I, and L229M are functional mutants of Cx40 with small alterations of physiological properties, but accelerated degradation by the proteasome. These findings suggest a novel mechanism (protein instability) for the pathogenesis of AF due to a connexin mutation and a novel approach to therapy (protease inhibition).
Collapse
Affiliation(s)
- Joanna Gemel
- Department of Pediatrics, University of Chicago, Chicago, IL 60637, USA
| | - Adria R Simon
- Department of Pediatrics, University of Chicago, Chicago, IL 60637, USA
| | - Dakshesh Patel
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Qin Xu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Arvydas Matiukas
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Richard D Veenstra
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Eric C Beyer
- Department of Pediatrics, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
22
|
Theis JL, Zimmermann MT, Larsen BT, Rybakova IN, Long PA, Evans JM, Middha S, de Andrade M, Moss RL, Wieben ED, Michels VV, Olson TM. TNNI3K mutation in familial syndrome of conduction system disease, atrial tachyarrhythmia and dilated cardiomyopathy. Hum Mol Genet 2014; 23:5793-804. [PMID: 24925317 DOI: 10.1093/hmg/ddu297] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Locus mapping has uncovered diverse etiologies for familial atrial fibrillation (AF), dilated cardiomyopathy (DCM), and mixed cardiac phenotype syndromes, yet the molecular basis for these disorders remains idiopathic in most cases. Whole-exome sequencing (WES) provides a powerful new tool for familial disease gene discovery. Here, synergistic application of these genomic strategies identified the pathogenic mutation in a familial syndrome of atrial tachyarrhythmia, conduction system disease (CSD), and DCM vulnerability. Seven members of a three-generation family exhibited the variably expressed phenotype, three of whom manifested CSD and clinically significant arrhythmia in childhood. Genome-wide linkage analysis mapped two equally plausible loci to chromosomes 1p3 and 13q12. Variants from WES of two affected cousins were filtered for rare, predicted-deleterious, positional variants, revealing an unreported heterozygous missense mutation disrupting the highly conserved kinase domain in TNNI3K. The G526D substitution in troponin I interacting kinase, with the most deleterious SIFT and Polyphen2 scores possible, resulted in abnormal peptide aggregation in vitro and in silico docking models predicted altered yet energetically favorable wild-type mutant dimerization. Ventricular tissue from a mutation carrier displayed histopathological hallmarks of DCM and reduced TNNI3K protein staining with unique amorphous nuclear and sarcoplasmic inclusions. In conclusion, mutation of TNNI3K, encoding a heart-specific kinase previously shown to modulate cardiac conduction and myocardial function in mice, underlies a familial syndrome of electrical and myopathic heart disease. The identified substitution causes a TNNI3K aggregation defect and protein deficiency, implicating a dominant-negative loss of function disease mechanism.
Collapse
Affiliation(s)
| | - Michael T Zimmermann
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research
| | | | - Inna N Rybakova
- Department of Cell and Regenerative Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Jared M Evans
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research
| | - Sumit Middha
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research
| | - Mariza de Andrade
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research
| | - Richard L Moss
- Department of Cell and Regenerative Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | | | - Timothy M Olson
- Cardiovascular Genetics Research Laboratory, Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine,
| |
Collapse
|