1
|
Sugie K, Nishino I. History and Perspective of LAMP-2 Deficiency (Danon Disease). Biomolecules 2024; 14:1272. [PMID: 39456205 PMCID: PMC11506487 DOI: 10.3390/biom14101272] [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: 08/31/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Danon disease, an X-linked dominant vacuolar cardiomyopathy and skeletal myopathy, is caused by a primary deficiency of lysosome-associated membrane protein-2 (LAMP-2). This disease is one of the autophagy-related muscle diseases. Male patients present with the triad of cardiomyopathy, myopathy, and intellectual disability, while female patients present with cardiomyopathy. The disease's leading cause of death is heart failure, and its prognostic factor is cardiomyopathy. Pathologically, the disease is characterized by the appearance of unique autophagic vacuoles with sarcolemmal features (AVSFs). Twenty-six families have been found to have this disease in Japan. It has been over 40 years since the first report of this disease by Danon et al. and over 20 years since the identification of the causative gene, LAMP2, by Nishino et al. Although the pathogenetic mechanism of Danon disease remains unestablished, the first clinical trials using AAV vectors have finally begun in recent years. The development of novel therapies is expected in the future.
Collapse
Affiliation(s)
- Kazuma Sugie
- Department of Neurology, School of Medicine, Nara Medical University, Nara 634-8521, Japan;
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo 187-8551, Japan
| |
Collapse
|
2
|
Pinheiro CV, Ribeiro RT, Roginski AC, Brondani M, Zemniaçak ÂB, Hoffmann CIH, Amaral AU, Wajner M, Baldo G, Leipnitz G. Disturbances in mitochondrial bioenergetics and control quality and unbalanced redox homeostasis in the liver of a mouse model of mucopolysaccharidosis type II. Mol Cell Biochem 2024:10.1007/s11010-024-04952-y. [PMID: 38498105 DOI: 10.1007/s11010-024-04952-y] [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: 07/18/2023] [Accepted: 01/26/2024] [Indexed: 03/20/2024]
Abstract
Mucopolysaccharidosis type II (MPS II; Hunter syndrome) is a lysosomal storage disease caused by mutations in the gene encoding the enzyme iduronate 2-sulfatase (IDS) and biochemically characterized by the accumulation of glycosaminoglycans (GAGs) in different tissues. It is a multisystemic disorder that presents liver abnormalities, the pathophysiology of which is not yet established. In the present study, we evaluated bioenergetics, redox homeostasis, and mitochondrial dynamics in the liver of 6-month-old MPS II mice (IDS-). Our findings show a decrease in the activity of α-ketoglutarate dehydrogenase and an increase in the activities of succinate dehydrogenase and malate dehydrogenase. The activity of mitochondrial complex I was also increased whereas the other complex activities were not affected. In contrast, mitochondrial respiration, membrane potential, ATP production, and calcium retention capacity were not altered. Furthermore, malondialdehyde levels and 2',7'-dichlorofluorescein oxidation were increased in the liver of MPS II mice, indicating lipid peroxidation and increased ROS levels, respectively. Sulfhydryl and reduced glutathione levels, as well as glutathione S-transferase, glutathione peroxidase (GPx), superoxide dismutase, and catalase activities were also increased. Finally, the levels of proteins involved in mitochondrial mass and dynamics were decreased in knockout mice liver. Taken together, these data suggest that alterations in energy metabolism, redox homeostasis, and mitochondrial dynamics can be involved in the pathophysiology of liver abnormalities observed in MPS II.
Collapse
Affiliation(s)
- Camila Vieira Pinheiro
- Postgraduation Program in Biological Sciences: Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Rafael Teixeira Ribeiro
- Postgraduation Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Ana Cristina Roginski
- Postgraduation Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Morgana Brondani
- Postgraduation Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Ângela Beatris Zemniaçak
- Postgraduation Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Chrístofer Ian Hernandez Hoffmann
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Alexandre Umpierrez Amaral
- Postgraduation Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
- Postgraduation Program in Integral Health Care, Universidade Regional Integrada do Alto Uruguai e das Missões, Erechim, Rio Grande do Sul, 99709-910, Brazil
| | - Moacir Wajner
- Postgraduation Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, 90035-903, Brazil
| | - Guilherme Baldo
- Postgraduation Program in Biological Sciences: Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, 90035-903, Brazil
| | - Guilhian Leipnitz
- Postgraduation Program in Biological Sciences: Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil.
- Postgraduation Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| |
Collapse
|
3
|
Ago Y, Rintz E, Musini KS, Ma Z, Tomatsu S. Molecular Mechanisms in Pathophysiology of Mucopolysaccharidosis and Prospects for Innovative Therapy. Int J Mol Sci 2024; 25:1113. [PMID: 38256186 PMCID: PMC10816168 DOI: 10.3390/ijms25021113] [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: 12/11/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Mucopolysaccharidoses (MPSs) are a group of inborn errors of the metabolism caused by a deficiency in the lysosomal enzymes required to break down molecules called glycosaminoglycans (GAGs). These GAGs accumulate over time in various tissues and disrupt multiple biological systems, including catabolism of other substances, autophagy, and mitochondrial function. These pathological changes ultimately increase oxidative stress and activate innate immunity and inflammation. We have described the pathophysiology of MPS and activated inflammation in this paper, starting with accumulating the primary storage materials, GAGs. At the initial stage of GAG accumulation, affected tissues/cells are reversibly affected but progress irreversibly to: (1) disruption of substrate degradation with pathogenic changes in lysosomal function, (2) cellular dysfunction, secondary/tertiary accumulation (toxins such as GM2 or GM3 ganglioside, etc.), and inflammatory process, and (3) progressive tissue/organ damage and cell death (e.g., skeletal dysplasia, CNS impairment, etc.). For current and future treatment, several potential treatments for MPS that can penetrate the blood-brain barrier and bone have been proposed and/or are in clinical trials, including targeting peptides and molecular Trojan horses such as monoclonal antibodies attached to enzymes via receptor-mediated transport. Gene therapy trials with AAV, ex vivo LV, and Sleeping Beauty transposon system for MPS are proposed and/or underway as innovative therapeutic options. In addition, possible immunomodulatory reagents that can suppress MPS symptoms have been summarized in this review.
Collapse
Affiliation(s)
- Yasuhiko Ago
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA; (Y.A.); (K.S.M.); (Z.M.)
| | - Estera Rintz
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland;
| | - Krishna Sai Musini
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA; (Y.A.); (K.S.M.); (Z.M.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Zhengyu Ma
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA; (Y.A.); (K.S.M.); (Z.M.)
| | - Shunji Tomatsu
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA; (Y.A.); (K.S.M.); (Z.M.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu 501-1112, Japan
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA 19144, USA
| |
Collapse
|
4
|
Uribe-Carretero E, Rey V, Fuentes JM, Tamargo-Gómez I. Lysosomal Dysfunction: Connecting the Dots in the Landscape of Human Diseases. BIOLOGY 2024; 13:34. [PMID: 38248465 PMCID: PMC10813815 DOI: 10.3390/biology13010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024]
Abstract
Lysosomes are the main organelles responsible for the degradation of macromolecules in eukaryotic cells. Beyond their fundamental role in degradation, lysosomes are involved in different physiological processes such as autophagy, nutrient sensing, and intracellular signaling. In some circumstances, lysosomal abnormalities underlie several human pathologies with different etiologies known as known as lysosomal storage disorders (LSDs). These disorders can result from deficiencies in primary lysosomal enzymes, dysfunction of lysosomal enzyme activators, alterations in modifiers that impact lysosomal function, or changes in membrane-associated proteins, among other factors. The clinical phenotype observed in affected patients hinges on the type and location of the accumulating substrate, influenced by genetic mutations and residual enzyme activity. In this context, the scientific community is dedicated to exploring potential therapeutic approaches, striving not only to extend lifespan but also to enhance the overall quality of life for individuals afflicted with LSDs. This review provides insights into lysosomal dysfunction from a molecular perspective, particularly in the context of human diseases, and highlights recent advancements and breakthroughs in this field.
Collapse
Affiliation(s)
- Elisabet Uribe-Carretero
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Caceres, Spain; (E.U.-C.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativa, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Caceres, Spain
| | - Verónica Rey
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Jose Manuel Fuentes
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Caceres, Spain; (E.U.-C.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativa, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Caceres, Spain
| | - Isaac Tamargo-Gómez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
5
|
Ryan T, Roberts JD. Emerging Targeted Therapies for Inherited Cardiomyopathies and Arrhythmias. Card Electrophysiol Clin 2023; 15:261-271. [PMID: 37558297 DOI: 10.1016/j.ccep.2023.04.006] [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] [Indexed: 08/11/2023]
Abstract
Inherited cardiomyopathy and arrhythmia syndromes are associated with significant morbidity and mortality, particularly in young people. Medical management of these conditions has primarily been limited to agents previously developed for more common forms of heart disease and not tailored to their distinct pathophysiology. As our understanding of their underlying genetics and disease mechanisms has improved, an era of targeted therapies for these rare conditions has begun to emerge. In recent years, several novel agents have been developed and tested in preclinical models and, in some cases, have advanced to both the clinical trial and clinical approval stages with exciting results. These new treatments are derived from multiple classes of therapeutics, including small molecules, antisense oligonucleotides, small interfering RNAs, adeno-associated virus-mediated gene therapies, and in vivo gene editing. Collectively, they carry the promise of revolutionizing management of affected patients and their families.
Collapse
Affiliation(s)
- Tammy Ryan
- McMaster University, Hamilton, Ontario, Canada; Department of Medicine, Division of Cardiology, DBCVSRI, Hamilton General Hospital, Room C3-121, 237 Barton Street East, Hamilton, Ontario L8L2X2, Canada
| | - Jason D Roberts
- McMaster University, Hamilton, Ontario, Canada; DBCVSRI, Room C3-111, 237 Barton Street East, Hamilton, Ontario L8L2X2, Canada; Population Health Research Institute and Hamilton Health Sciences, Hamilton, Ontario, Canada.
| |
Collapse
|
6
|
Burban A, Pucyło S, Sikora A, Opolski G, Grabowski M, Kołodzińska A. Hypertrophic Cardiomyopathy versus Storage Diseases with Myocardial Involvement. Int J Mol Sci 2023; 24:13239. [PMID: 37686045 PMCID: PMC10488064 DOI: 10.3390/ijms241713239] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/20/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
One of the main causes of heart failure is cardiomyopathies. Among them, the most common is hypertrophic cardiomyopathy (HCM), characterized by thickening of the left ventricular muscle. This article focuses on HCM and other cardiomyopathies with myocardial hypertrophy, including Fabry disease, Pompe disease, and Danon disease. The genetics and pathogenesis of these diseases are described, as well as current and experimental treatment options, such as pharmacological intervention and the potential of gene therapies. Although genetic approaches are promising and have the potential to become the best treatments for these diseases, further research is needed to evaluate their efficacy and safety. This article describes current knowledge and advances in the treatment of the aforementioned cardiomyopathies.
Collapse
Affiliation(s)
- Anna Burban
- First Department of Cardiology, Medical University of Warsaw, ul. Banacha 1A, 02-097 Warszawa, Poland; (A.B.); (S.P.); (A.S.); (G.O.); (M.G.)
- Doctoral School, Medical University of Warsaw, 81 Żwirki i Wigury Street, 02-091 Warsaw, Poland
| | - Szymon Pucyło
- First Department of Cardiology, Medical University of Warsaw, ul. Banacha 1A, 02-097 Warszawa, Poland; (A.B.); (S.P.); (A.S.); (G.O.); (M.G.)
| | - Aleksandra Sikora
- First Department of Cardiology, Medical University of Warsaw, ul. Banacha 1A, 02-097 Warszawa, Poland; (A.B.); (S.P.); (A.S.); (G.O.); (M.G.)
| | - Grzegorz Opolski
- First Department of Cardiology, Medical University of Warsaw, ul. Banacha 1A, 02-097 Warszawa, Poland; (A.B.); (S.P.); (A.S.); (G.O.); (M.G.)
| | - Marcin Grabowski
- First Department of Cardiology, Medical University of Warsaw, ul. Banacha 1A, 02-097 Warszawa, Poland; (A.B.); (S.P.); (A.S.); (G.O.); (M.G.)
| | - Agnieszka Kołodzińska
- First Department of Cardiology, Medical University of Warsaw, ul. Banacha 1A, 02-097 Warszawa, Poland; (A.B.); (S.P.); (A.S.); (G.O.); (M.G.)
| |
Collapse
|
7
|
Liang W, Sagar S, Ravindran R, Najor RH, Quiles JM, Chi L, Diao RY, Woodall BP, Leon LJ, Zumaya E, Duran J, Cauvi DM, De Maio A, Adler ED, Gustafsson ÅB. Mitochondria are secreted in extracellular vesicles when lysosomal function is impaired. Nat Commun 2023; 14:5031. [PMID: 37596294 PMCID: PMC10439183 DOI: 10.1038/s41467-023-40680-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 08/04/2023] [Indexed: 08/20/2023] Open
Abstract
Mitochondrial quality control is critical for cardiac homeostasis as these organelles are responsible for generating most of the energy needed to sustain contraction. Dysfunctional mitochondria are normally degraded via intracellular degradation pathways that converge on the lysosome. Here, we identified an alternative mechanism to eliminate mitochondria when lysosomal function is compromised. We show that lysosomal inhibition leads to increased secretion of mitochondria in large extracellular vesicles (EVs). The EVs are produced in multivesicular bodies, and their release is independent of autophagy. Deletion of the small GTPase Rab7 in cells or adult mouse heart leads to increased secretion of EVs containing ubiquitinated cargos, including intact mitochondria. The secreted EVs are captured by macrophages without activating inflammation. Hearts from aged mice or Danon disease patients have increased levels of secreted EVs containing mitochondria indicating activation of vesicular release during cardiac pathophysiology. Overall, these findings establish that mitochondria are eliminated in large EVs through the endosomal pathway when lysosomal degradation is inhibited.
Collapse
Affiliation(s)
- Wenjing Liang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Shakti Sagar
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Rishith Ravindran
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Rita H Najor
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Justin M Quiles
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Liguo Chi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Rachel Y Diao
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Benjamin P Woodall
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Leonardo J Leon
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Erika Zumaya
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jason Duran
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - David M Cauvi
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
| | - Antonio De Maio
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
| | - Eric D Adler
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Åsa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Kleefeld F, Hentschel A, von Moers A, Hahn K, Horvath R, Goebel HH, Preusse C, Schallner J, Schuelke M, Roos A, Stenzel W. Beyond vacuolar pathology: Multiomic profiling of Danon disease reveals dysfunctional mitochondrial homeostasis. Neuropathol Appl Neurobiol 2023; 49:e12920. [PMID: 37328427 DOI: 10.1111/nan.12920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/06/2023] [Accepted: 06/11/2023] [Indexed: 06/18/2023]
Affiliation(s)
- Felix Kleefeld
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Neurology, Berlin, Germany
| | - Andreas Hentschel
- Leibniz-Institut für Analytische Wissenschaften-ISAS e.V., Dortmund, Germany
| | - Arpad von Moers
- Klinik für Kinder- und Jugendmedizin, DRK Kliniken Berlin Westend, Berlin, Germany
| | - Katrin Hahn
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Neurology, Berlin, Germany
| | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Hans-Hilmar Goebel
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Neuropathology, Berlin, Germany
| | - Corinna Preusse
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Neuropathology, Berlin, Germany
| | - Jens Schallner
- Department of Neuropaediatrics, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Markus Schuelke
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), NeuroCure Clinical Research Center, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Neuropediatrics, Berlin, Germany
| | - Andreas Roos
- Pediatric Neurology, University Children's Hospital, Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Werner Stenzel
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Neuropathology, Berlin, Germany
| |
Collapse
|
9
|
Liang W, Diao RY, Quiles JM, Najor RH, Chi L, Woodall BP, Leon LJ, Duran J, Cauvi DM, De Maio A, Adler ED, Gustafsson ÃSB. The Small GTPase Rab7 Regulates Release of Mitochondria in Extracellular Vesicles in Response to Lysosomal Dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.11.528148. [PMID: 36824711 PMCID: PMC9949095 DOI: 10.1101/2023.02.11.528148] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Mitochondrial quality control is critical for cardiac homeostasis as these organelles are responsible for generating most of the energy needed to sustain contraction. Dysfunctional mitochondria are normally degraded via intracellular degradation pathways that converge on the lysosome. Here, we identified an alternative mechanism to eliminate mitochondria when lysosomal function is compromised. We show that lysosomal inhibition leads to increased secretion of mitochondria in large extracellular vesicles (EVs). The EVs are produced in multivesicular bodies, and their release is independent of autophagy. Deletion of the small GTPase Rab7 in cells or adult mouse heart leads to increased secretion of EVs containing ubiquitinated cargos, including intact mitochondria. The secreted EVs are captured by macrophages without activating inflammation. Hearts from aged mice or Danon disease patients have increased levels of secreted EVs containing mitochondria indicating activation of vesicular release during cardiac pathophysiology. Overall, these findings establish that mitochondria are eliminated in large EVs through the endosomal pathway when lysosomal degradation is inhibited.
Collapse
|
10
|
Zhai Y, Miao J, Peng Y, Wang Y, Dong J, Zhao X. Clinical features of Danon disease and insights gained from LAMP-2 deficiency models. Trends Cardiovasc Med 2023; 33:81-89. [PMID: 34737089 DOI: 10.1016/j.tcm.2021.10.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 10/06/2021] [Accepted: 10/28/2021] [Indexed: 02/07/2023]
Abstract
Danon disease (DD) is an X-linked multisystem disorder with clinical features characterized by the triad of hypertrophic cardiomyopathy, skeletal muscle weakness, and mental retardation. Cardiac involvement can be fatal in the absence of an effective treatment option such as heart transplantation. Molecular studies have proved that LAMP-2 protein deficiency, mainly LAMP-2B isoform, resulting from LAMP2 gene mutation, is the culprit for DD. Autophagy impairment due to LAMP-2 deficiency mediated the accumulation of abnormal autophagic vacuoles in cells. While it is not ideal for mimicking DD phenotypes in humans, the emergence of LAMP-2-deficient animal models and induced pluripotent stem cells from DD patients provided powerful tools for exploring DD mechanism. In both in vitro and in vivo studies, much evidence has demonstrated that mitochondria dysfunction and fragmentation can result in DD pathology. Fundamental research contributes to the therapeutic transformation. By targeting the molecular core, several potential therapies have demonstrated promising results in partial phenotypes improvement. Among them, gene therapies anticipate inaugurate a class of symptom control and prevention drugs as their in vivo effects are promising, and one clinical trial is currently underway.
Collapse
Affiliation(s)
- Yafei Zhai
- Centre for Cardiovascular Diseases, Henan Key Laboratory of Hereditary Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, P.R. China
| | - Jinxin Miao
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, P.R. China;Department of Science and Technology, Henan University of Chinese Medicine, Zhengzhou, Henan, P.R. China; Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Ying Peng
- Centre for Cardiovascular Diseases, Henan Key Laboratory of Hereditary Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, P.R. China
| | - Yaohe Wang
- Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, P. R. China; Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Jianzeng Dong
- Centre for Cardiovascular Diseases, Henan Key Laboratory of Hereditary Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, P.R. China; Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Xiaoyan Zhao
- Centre for Cardiovascular Diseases, Henan Key Laboratory of Hereditary Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, P.R. China.
| |
Collapse
|
11
|
Barndt RJ, Liu Q, Tang Y, Haugh MP, Cui J, Chan SY, Wu H. Metabolic Maturation Exaggerates Abnormal Calcium Handling in a Lamp2 Knockout Human Pluripotent Stem Cell-Derived Cardiomyocyte Model of Danon Disease. Biomolecules 2022; 13:69. [PMID: 36671453 PMCID: PMC9855424 DOI: 10.3390/biom13010069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
Danon disease (DD) is caused by mutations of the gene encoding lysosomal-associated membrane protein type 2 (LAMP2), which lead to impaired autophagy, glycogen accumulation, and cardiac hypertrophy. However, it is not well understood why a large portion of DD patients develop arrhythmia and sudden cardiac death. In the current study, we generated LAMP2 knockout (KO) human iPSC-derived cardiomyocytes (CM), which mimic the LAMP2 dysfunction in DD heart. Morphologic analysis demonstrated the sarcomere disarrangement in LAMP2 KO CMs. In functional studies, LAMP2 KO CMs showed near-normal calcium handling at base level. However, treatment of pro-maturation medium (MM) exaggerated the disease phenotype in the KO cells as they exhibited impaired calcium recycling and increased irregular beating events, which recapitulates the pro-arrhythmia phenotypes of DD patients. Further mechanistic study confirmed that MM treatment significantly enhanced the autophagic stress in the LAMP2 KO CMs, which was accompanied by an increase of both cellular and mitochondrial reactive oxygen species (ROS) levels. Excess ROS accumulation in LAMP2 KO CMs resulted in the over-activation of calcium/calmodulin dependent protein kinase IIδ (CaMKIIδ) and arrhythmogenesis, which was partially rescued by the treatment of ROS scavenger. In summary, our study has revealed ROS induced CaMKIIδ overactivation as a key mechanism that promotes cardiac arrhythmia in DD patients.
Collapse
Affiliation(s)
- Robert J. Barndt
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Qing Liu
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
- Center for Human Genetics, Clemson University, Greenwood, SC 29646, USA
| | - Ying Tang
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Michael P. Haugh
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15213, USA
| | - Jeffery Cui
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Stephen Y. Chan
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Haodi Wu
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15213, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
12
|
Vučković S, Dinani R, Nollet EE, Kuster DWD, Buikema JW, Houtkooper RH, Nabben M, van der Velden J, Goversen B. Characterization of cardiac metabolism in iPSC-derived cardiomyocytes: lessons from maturation and disease modeling. STEM CELL RESEARCH & THERAPY 2022; 13:332. [PMID: 35870954 PMCID: PMC9308297 DOI: 10.1186/s13287-022-03021-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/25/2022] [Indexed: 12/02/2022]
Abstract
Background Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) have emerged as a powerful tool for disease modeling, though their immature nature currently limits translation into clinical practice. Maturation strategies increasingly pay attention to cardiac metabolism because of its pivotal role in cardiomyocyte development and function. Moreover, aberrances in cardiac metabolism are central to the pathogenesis of cardiac disease. Thus, proper modeling of human cardiac disease warrants careful characterization of the metabolic properties of iPSC-CMs. Methods Here, we examined the effect of maturation protocols on healthy iPSC-CMs applied in 23 studies and compared fold changes in functional metabolic characteristics to assess the level of maturation. In addition, pathological metabolic remodeling was assessed in 13 iPSC-CM studies that focus on hypertrophic cardiomyopathy (HCM), which is characterized by abnormalities in metabolism. Results Matured iPSC-CMs were characterized by mitochondrial maturation, increased oxidative capacity and enhanced fatty acid use for energy production. HCM iPSC-CMs presented varying degrees of metabolic remodeling ranging from compensatory to energy depletion stages, likely due to the different types of mutations and clinical phenotypes modeled. HCM further displayed early onset hypertrophy, independent of the type of mutation or disease stage. Conclusions Maturation strategies improve the metabolic characteristics of iPSC-CMs, but not to the level of the adult heart. Therefore, a combination of maturation strategies might prove to be more effective. Due to early onset hypertrophy, HCM iPSC-CMs may be less suitable to detect early disease modifiers in HCM and might prove more useful to examine the effects of gene editing and new drugs in advanced disease stages. With this review, we provide an overview of the assays used for characterization of cardiac metabolism in iPSC-CMs and advise on which metabolic assays to include in future maturation and disease modeling studies.
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03021-9.
Collapse
|
13
|
Barnes AM, Holmstoen TB, Bonham AJ, Rowland TJ. Differentiating Human Pluripotent Stem Cells to Cardiomyocytes Using Purified Extracellular Matrix Proteins. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120720. [PMID: 36550926 PMCID: PMC9774171 DOI: 10.3390/bioengineering9120720] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/11/2022] [Accepted: 11/21/2022] [Indexed: 11/23/2022]
Abstract
Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) can be differentiated into cardiomyocytes (hESC-CMs and iPSC-CMs, respectively), which hold great promise for cardiac regenerative medicine and disease modeling efforts. However, the most widely employed differentiation protocols require undefined substrates that are derived from xenogeneic (animal) products, contaminating resultant hESC- and iPSC-CM cultures with xenogeneic proteins and limiting their clinical applicability. Additionally, typical hESC- and iPSC-CM protocols produce CMs that are significantly contaminated by non-CMs and that are immature, requiring lengthy maturation procedures. In this review, we will summarize recent studies that have investigated the ability of purified extracellular matrix (ECM) proteins to support hESC- and iPSC-CM differentiation, with a focus on commercially available ECM proteins and coatings to make such protocols widely available to researchers. The most promising of the substrates reviewed here include laminin-521 with laminin-221 together or Synthemax (a synthetic vitronectin-based peptide coating), which both resulted in highly pure CM cultures. Future efforts are needed to determine whether combinations of specific purified ECM proteins or derived peptides could further improve CM maturation and culture times, and significantly improve hESC- and iPSC-CM differentiation protocols.
Collapse
Affiliation(s)
- Ashlynn M. Barnes
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Tessa B. Holmstoen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Andrew J. Bonham
- Department of Chemistry & Biochemistry, Metropolitan State University of Denver, Denver, CO 80217, USA
| | - Teisha J. Rowland
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- Correspondence:
| |
Collapse
|
14
|
Yadin D, Petrover Z, Shainberg A, Alcalai R, Waldman M, Seidman J, Seidman CE, Abraham NG, Hochhauser E, Arad M. Autophagy Guided Interventions to Modify the Cardiac Phenotype of Danon Disease. Biochem Pharmacol 2022; 204:115229. [PMID: 36027926 DOI: 10.1016/j.bcp.2022.115229] [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: 06/09/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 11/26/2022]
Abstract
Danon disease is a lethal X-linked genetic syndrome resulting from radical mutations in the LAMP2 gene. LAMP2 protein deficiency results in defective lysosomal function, autophagy arrest and a multisystem disorder primarily involving the heart, skeletal muscle and the central nervous system. Cardiomyopathy is the main cause of morbidity and mortality. To investigate the mechanisms of and develop therapies for cardiac Danon disease we engineered a mouse model carrying an exon 6 deletion human mutation in LAMP2, which recapitulates the human cardiac disease phenotype. Mice develop cardiac hypertrophy followed by left ventricular dilatation and systolic dysfunction, in association with progressive fibrosis, oxidative stress, accumulation of autophagosomes and activation of proteasome. Stimulation of autophagy in Danon mice (by exercise training, caloric restriction, and rapamycin) aggravate the disease phenotype by promoting dilated cardiomyopathy. Inhibiting autophagy (by high fat diet or hydroxychloroquine) is better tolerated by Danon mice compared to wild type but is not curative. Inhibiting proteasome by Velcade was found to be highly toxic to Danon mice, suggesting that proteasome is activated to compensate for defective autophagy. In conclusion, activation of autophagy should be avoided in Danon patients. Since Danon's is a lifelong disease, we suggest that lifestyle interventions to decrease cardiac stress may be useful to slow progression of Danon's cardiomyopathy. While Danon mice better tolerate high fat diet and sedentary lifestyle, the benefit regarding cardiomyopathy in humans needs to be balanced against other health consequences of such interventions.
Collapse
Affiliation(s)
- Dor Yadin
- Felsenstein Research Center and the Department of Cardiothoracic, Rabin Medical Center, Sackler School of Medicine, Tel-Aviv University, Petach Tikva, Israel; Leviev Heart Center, Sheba Medical Center, Sackler School of Medicine, Tel-Aviv University, Israel
| | - Zachary Petrover
- Felsenstein Research Center and the Department of Cardiothoracic, Rabin Medical Center, Sackler School of Medicine, Tel-Aviv University, Petach Tikva, Israel; Bar-Ilan University, Ramat Gan, Israel
| | | | - Ronny Alcalai
- Heart Institute, Hadassah Hebrew University Medical Center, Jerusalem, Israel. 5. Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Maayan Waldman
- Felsenstein Research Center and the Department of Cardiothoracic, Rabin Medical Center, Sackler School of Medicine, Tel-Aviv University, Petach Tikva, Israel
| | - Jon Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Christine E Seidman
- Howard Hughes Medical Institute and Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 02115
| | - Nader G Abraham
- Departments of Medicine and Pharmacology, New York Medical College, Valhalla, NY, USA
| | - Edith Hochhauser
- Felsenstein Research Center and the Department of Cardiothoracic, Rabin Medical Center, Sackler School of Medicine, Tel-Aviv University, Petach Tikva, Israel
| | - Michael Arad
- Leviev Heart Center, Sheba Medical Center, Sackler School of Medicine, Tel-Aviv University, Israel.
| |
Collapse
|
15
|
Abstract
Purpose of Review The advent of induced pluripotent stem cells (iPSC) has paved the way for new in vitro models of human cardiomyopathy. Herein, we will review existing models of disease as well as strengths and limitations of the system. Recent Findings Preclinical studies have now demonstrated that iPSCs generated from patients with both acquired or heritable genetic diseases retain properties of the disease in vitro and can be used as a model to study novel therapeutics. iPSCs can be differentiated in vitro into the cardiomyocyte lineage into cells resembling adult ventricular myocytes that retain properties of cardiovascular disease from their respective donor. iPSC pluripotency allows for them to be frozen, stored, and continually used to generate iPSC-derived myocytes for future experiments without need for invasive procedures or repeat myocyte isolations to obtain animal or human cardiac tissues. Summary While not without their limitations, iPSC models offer new ways for studying patient-specific cardiomyopathies. iPSCs offer a high-throughput avenue for drug development, modeling of disease pathophysiology in vitro, and enabling experimental repair strategies without need for invasive procedures to obtain cardiac tissues.
Collapse
|
16
|
Bileck A, Bortel P, Kriz M, Janker L, Kiss E, Gerner C, Del Favero G. Inward Outward Signaling in Ovarian Cancer: Morpho-Phospho-Proteomic Profiling Upon Application of Hypoxia and Shear Stress Characterizes the Adaptive Plasticity of OVCAR-3 and SKOV-3 Cells. Front Oncol 2022; 11:746411. [PMID: 35251951 PMCID: PMC8896345 DOI: 10.3389/fonc.2021.746411] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/27/2021] [Indexed: 12/26/2022] Open
Abstract
With the onset of resistance, ovarian cancer cells display almost unpredictable adaptive potential. This may derive from the tumor genetic ancestry and can be additionally tailored by post translational protein modifications (PTMs). In this study, we took advantage of high-end (phospho)-proteome analysis combined with multiparametric morphometric profiling in high-grade serous (OVCAR-3) and non-serous (SKOV-3) ovarian carcinoma cells. For functional experiments, we applied two different protocols, representing typical conditions of the abdominal cavity and of the growing tumor tissue: on the one side hypoxia (oxygen 1%) which develops within the tumor mass or is experienced during migration/extravasation in non-vascularized areas. On the other hand, fluid shear stress (250 rpm, 2.8 dyn/cm2) which affects tumor surface in the peritoneum or metastases in the bloodstream. After 3 hours incubation, treatment groups were clearly distinguishable by PCA analysis. Whereas basal proteome profiles of OVCAR-3 and SKOV-3 cells appeared almost unchanged, phosphoproteome analysis revealed multiple regulatory events. These affected primarily cellular structure and proliferative potential and consolidated in the proteome signature after 24h treatment. Upon oxygen reduction, metabolism switched toward glycolysis (e.g. upregulation hexokinase-2; HK2) and cell size increased, in concerted regulation of pathways related to Rho-GTPases and/or cytoskeletal elements, resembling a vasculogenic mimicry response. Shear stress regulated proteins governing cell cycle and structure, as well as the lipid metabolism machinery including the delta(14)-sterol reductase, kinesin-like proteins (KIF-22/20A) and the actin-related protein 2/3 complex. Independent microscopy-based validation experiments confirmed cell-type specific morphometric responses. In conclusion, we established a robust workflow enabling the description of the adaptive potential of ovarian cancer cells to physical and chemical stressors typical for the abdominal cavity and supporting the identification of novel molecular mechanisms sustaining tumor plasticity and pharmacologic resistance.
Collapse
Affiliation(s)
- Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Patricia Bortel
- Department of Analytical Chemistry, Faculty of Chemistry University of Vienna, Vienna, Austria
| | - Michelle Kriz
- Department of Analytical Chemistry, Faculty of Chemistry University of Vienna, Vienna, Austria
- Department of Food Chemistry and Toxicology, Faculty of Chemistry University of Vienna, Vienna, Austria
| | - Lukas Janker
- Department of Analytical Chemistry, Faculty of Chemistry University of Vienna, Vienna, Austria
| | - Endre Kiss
- Core Facility Multimodal Imaging, Faculty of Chemistry University of Vienna, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Vienna, Austria
- Core Facility Multimodal Imaging, Faculty of Chemistry University of Vienna, Vienna, Austria
- *Correspondence: Giorgia Del Favero, ; Christopher Gerner,
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry University of Vienna, Vienna, Austria
- Core Facility Multimodal Imaging, Faculty of Chemistry University of Vienna, Vienna, Austria
- *Correspondence: Giorgia Del Favero, ; Christopher Gerner,
| |
Collapse
|
17
|
Zhao M, Lian A, Zhong L, Guo R. The regulatory mechanism between lysosomes and mitochondria in the aetiology of cardiovascular diseases. Acta Physiol (Oxf) 2022; 234:e13757. [PMID: 34978753 DOI: 10.1111/apha.13757] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/16/2021] [Accepted: 01/01/2022] [Indexed: 11/28/2022]
Abstract
Coordinated action among various organelles maintains cellular functions. For instance, mitochondria and lysosomes are the main organelles contributing to cellular metabolism and provide energy for cardiomyocyte contraction. They also provide essential signalling platforms in the cell that regulate many key processes such as autophagy, apoptosis, oxidative stress, inflammation and cell death. Often, abnormalities in mitochondrial or lysosomal structures and functions bring about cardiovascular diseases (CVDs). Although the communication between mitochondria and lysosomes throughout the cardiovascular system is intensely studied, the regulatory mechanisms have not been completely understood. Thus, we summarize the most recent studies related to mitochondria and lysosomes' role in CVDs and their potential connections and communications under cardiac pathophysiological conditions. Further, we discuss limitations and future perspectives regarding diagnosis, therapeutic strategies and drug discovery in CVDs.
Collapse
Affiliation(s)
- Mengxue Zhao
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
| | - Andrew Lian
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona California USA
| | - Li Zhong
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona California USA
| | - Rui Guo
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
- The Key Laboratory of Zoological Systematics and Application College of Life Sciences Hebei University Baoding China
| |
Collapse
|
18
|
Wen X, Yang Y, Klionsky DJ. Moments in autophagy and disease: Past and present. Mol Aspects Med 2021; 82:100966. [PMID: 33931245 PMCID: PMC8548407 DOI: 10.1016/j.mam.2021.100966] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/13/2021] [Accepted: 04/19/2021] [Indexed: 01/18/2023]
Abstract
Over the past several decades, research on autophagy, a highly conserved lysosomal degradation pathway, has been advanced by studies in different model organisms, especially in the field of its molecular mechanism and regulation. The malfunction of autophagy is linked to various diseases, among which cancer and neurodegenerative diseases are the major focus. In this review, we cover some other important diseases, including cardiovascular diseases, infectious and inflammatory diseases, and metabolic disorders, as well as rare diseases, with a hope of providing a more complete understanding of the spectrum of autophagy's role in human health.
Collapse
Affiliation(s)
- Xin Wen
- Life Sciences Institute, Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Ying Yang
- Life Sciences Institute, Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel J Klionsky
- Life Sciences Institute, Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
19
|
Lewis-Israeli YR, Volmert BD, Gabalski MA, Huang AR, Aguirre A. Generating Self-Assembling Human Heart Organoids Derived from Pluripotent Stem Cells. J Vis Exp 2021. [PMID: 34605811 DOI: 10.3791/63097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The ability to study human cardiac development in health and disease is highly limited by the capacity to model the complexity of the human heart in vitro. Developing more efficient organ-like platforms that can model complex in vivo phenotypes, such as organoids and organs-on-a-chip, will enhance the ability to study human heart development and disease. This paper describes a protocol to generate highly complex human heart organoids (hHOs) by self-organization using human pluripotent stem cells and stepwise developmental pathway activation using small molecule inhibitors. Embryoid bodies (EBs) are generated in a 96-well plate with round-bottom, ultra-low attachment wells, facilitating suspension culture of individualized constructs. The EBs undergo differentiation into hHOs by a three-step Wnt signaling modulation strategy, which involves an initial Wnt pathway activation to induce cardiac mesoderm fate, a second step of Wnt inhibition to create definitive cardiac lineages, and a third Wnt activation step to induce proepicardial organ tissues. These steps, carried out in a 96-well format, are highly efficient, reproducible, and produce large amounts of organoids per run. Analysis by immunofluorescence imaging from day 3 to day 11 of differentiation reveals first and second heart field specifications and highly complex tissues inside hHOs at day 15, including myocardial tissue with regions of atrial and ventricular cardiomyocytes, as well as internal chambers lined with endocardial tissue. The organoids also exhibit an intricate vascular network throughout the structure and an external lining of epicardial tissue. From a functional standpoint, hHOs beat robustly and present normal calcium activity as determined by Fluo-4 live imaging. Overall, this protocol constitutes a solid platform for in vitro studies in human organ-like cardiac tissues.
Collapse
Affiliation(s)
- Yonatan R Lewis-Israeli
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University; Department of Biomedical Engineering, College of Engineering, Michigan State University
| | - Brett D Volmert
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University; Department of Biomedical Engineering, College of Engineering, Michigan State University
| | - Mitchell A Gabalski
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University; Department of Biomedical Engineering, College of Engineering, Michigan State University
| | - Amanda R Huang
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University; Department of Biomedical Engineering, College of Engineering, Michigan State University
| | - Aitor Aguirre
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University; Department of Biomedical Engineering, College of Engineering, Michigan State University;
| |
Collapse
|
20
|
Alcalai R, Arad M, Wakimoto H, Yadin D, Gorham J, Wang L, Burns E, Maron BJ, Roberts WC, Konno T, Conner DA, Perez-Atayde AR, Seidman JG, Seidman CE. LAMP2 Cardiomyopathy: Consequences of Impaired Autophagy in the Heart. J Am Heart Assoc 2021; 10:e018829. [PMID: 34459252 PMCID: PMC8649277 DOI: 10.1161/jaha.120.018829] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Human mutations in the X‐linked lysosome‐associated membrane protein‐2 (LAMP2) gene can cause a multisystem Danon disease or a primary cardiomyopathy characterized by massive hypertrophy, conduction system abnormalities, and malignant ventricular arrhythmias. We introduced an in‐frame LAMP2 gene exon 6 deletion mutation (denoted L2Δ6) causing human cardiomyopathy, into mouse LAMP2 gene, to elucidate its consequences on cardiomyocyte biology. This mutation results in in‐frame deletion of 41 amino acids, compatible with presence of some defective LAMP2 protein. Methods and Results Left ventricular tissues from L2Δ6 and wild‐type mice had equivalent amounts of LAMP2 RNA, but a significantly lower level of LAMP2 protein. By 20 weeks of age male mutant mice developed left ventricular hypertrophy which was followed by left ventricular dilatation and reduced systolic function. Cardiac electrophysiology and isolated cardiomyocyte studies demonstrated ventricular arrhythmia, conduction disturbances, abnormal calcium transients and increased sensitivity to catecholamines. Myocardial fibrosis was strikingly increased in 40‐week‐old L2Δ6 mice, recapitulating findings of human LAMP2 cardiomyopathy. Immunofluorescence and transmission electron microscopy identified mislocalization of lysosomes and accumulation of autophagosomes between sarcomeres, causing profound morphological changes disrupting the cellular ultrastructure. Transcription profile and protein expression analyses of L2Δ6 hearts showed significantly increased expression of genes encoding activators and protein components of autophagy, hypertrophy, and apoptosis. Conclusions We suggest that impaired autophagy results in cardiac hypertrophy and profound transcriptional reactions that impacted metabolism, calcium homeostasis, and cell survival. These responses define the molecular pathways that underlie the pathology and aberrant electrophysiology in cardiomyopathy of Danon disease.
Collapse
Affiliation(s)
- Ronny Alcalai
- Heart InstituteHadassah Hebrew University Medical Center Jerusalem Israel.,Department of Genetics Harvard Medical School Boston MA
| | - Michael Arad
- Division of Cardiology Sheba Medical Centre and Tel Aviv University Ramat Gan Israel
| | | | - Dor Yadin
- Division of Cardiology Sheba Medical Centre and Tel Aviv University Ramat Gan Israel
| | - Joshua Gorham
- Department of Genetics Harvard Medical School Boston MA
| | - Libin Wang
- Department of Genetics Harvard Medical School Boston MA
| | - Elia Burns
- Heart InstituteHadassah Hebrew University Medical Center Jerusalem Israel
| | - Barry J Maron
- Minneapolis Heart Institute Foundation Minneapolis MN
| | - William C Roberts
- Baylor Heart & Vascular InstituteBaylor University Medical Center Dallas TX
| | - Tetsuo Konno
- Department of Genetics Harvard Medical School Boston MA
| | | | | | - Jon G Seidman
- Department of Genetics Harvard Medical School Boston MA
| | - Christine E Seidman
- Department of Genetics Harvard Medical School Boston MA.,Howard Hughes Medical Institute and Cardiovascular DivisionBrigham and Women's Hospital Boston MA
| |
Collapse
|
21
|
Angiotensin Receptor Blocker and Neprilysin Inhibitor Suppresses Cardiac Dysfunction by Accelerating Myocardial Angiogenesis in Apolipoprotein E-Knockout Mice Fed a High-Fat Diet. J Renin Angiotensin Aldosterone Syst 2021; 2021:9916789. [PMID: 34394711 PMCID: PMC8357528 DOI: 10.1155/2021/9916789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022] Open
Abstract
Materials and Methods Male apolipoprotein E-knockout mice fed a high-fat diet were divided into control (CTL), valsartan (30 mg/kg) (VAL), sacubitril (30 mg/kg) (SAC), and valsartan plus sacubitril (30 mg/kg each) (VAL/SAC) groups after 4 weeks of prefeeding and were subsequently treated for 12 weeks. Results The VAL/SAC group exhibited significantly higher serum brain natriuretic peptide levels; more subtle changes in left ventricular systolic diameter, fractional shortening, and ejection fraction, and significantly higher expression levels of natriuretic peptide precursor B and markers of angiogenesis, including clusters of differentiation 34, vascular endothelial growth factor A, and monocyte chemotactic protein 1, than the CTL group. Conclusions Valsartan plus sacubitril preserved left ventricular systolic function in apolipoprotein E-knockout mice fed a high-fat diet. This result suggests that myocardial angiogenic factors induced by ARNI might provide cardioprotective effects.
Collapse
|
22
|
Zhang J, Chou OHI, Tse YL, Ng KM, Tse HF. Application of Patient-Specific iPSCs for Modelling and Treatment of X-Linked Cardiomyopathies. Int J Mol Sci 2021; 22:ijms22158132. [PMID: 34360897 PMCID: PMC8347533 DOI: 10.3390/ijms22158132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 12/11/2022] Open
Abstract
Inherited cardiomyopathies are among the major causes of heart failure and associated with significant mortality and morbidity. Currently, over 70 genes have been linked to the etiology of various forms of cardiomyopathy, some of which are X-linked. Due to the lack of appropriate cell and animal models, it has been difficult to model these X-linked cardiomyopathies. With the advancement of induced pluripotent stem cell (iPSC) technology, the ability to generate iPSC lines from patients with X-linked cardiomyopathy has facilitated in vitro modelling and drug testing for the condition. Nonetheless, due to the mosaicism of the X-chromosome inactivation, disease phenotypes of X-linked cardiomyopathy in heterozygous females are also usually more heterogeneous, with a broad spectrum of presentation. Recent advancements in iPSC procedures have enabled the isolation of cells with different lyonisation to generate isogenic disease and control cell lines. In this review, we will summarise the current strategies and examples of using an iPSC-based model to study different types of X-linked cardiomyopathy. The potential application of isogenic iPSC lines derived from a female patient with heterozygous Danon disease and drug screening will be demonstrated by our preliminary data. The limitations of an iPSC-derived cardiomyocyte-based platform will also be addressed.
Collapse
Affiliation(s)
- Jennifer Zhang
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.Z.); (O.H.-I.C.); (Y.-L.T.)
| | - Oscar Hou-In Chou
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.Z.); (O.H.-I.C.); (Y.-L.T.)
| | - Yiu-Lam Tse
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.Z.); (O.H.-I.C.); (Y.-L.T.)
| | - Kwong-Man Ng
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.Z.); (O.H.-I.C.); (Y.-L.T.)
- Correspondence: (K.-M.N.); (H.-F.T.); Tel.: +852-3917-9955 (K.-M.N.); +852-2255-3598 (H.-F.T.)
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.Z.); (O.H.-I.C.); (Y.-L.T.)
- Centre of Translational Stem Cell Biology, Hong Kong Science and Technology Park, Hong Kong, China
- Correspondence: (K.-M.N.); (H.-F.T.); Tel.: +852-3917-9955 (K.-M.N.); +852-2255-3598 (H.-F.T.)
| |
Collapse
|
23
|
Manso AM, Hashem SI, Nelson BC, Gault E, Soto-Hermida A, Villarruel E, Brambatti M, Bogomolovas J, Bushway PJ, Chen C, Battiprolu P, Keravala A, Schwartz JD, Shah G, Gu Y, Dalton ND, Hammond K, Peterson K, Saftig P, Adler ED. Systemic AAV9.LAMP2B injection reverses metabolic and physiologic multiorgan dysfunction in a murine model of Danon disease. Sci Transl Med 2021; 12:12/535/eaax1744. [PMID: 32188720 DOI: 10.1126/scitranslmed.aax1744] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 08/18/2019] [Accepted: 02/20/2020] [Indexed: 12/27/2022]
Abstract
Danon disease (DD) is a rare X-linked autophagic vacuolar myopathy associated with multiorgan dysfunction, including the heart, skeletal muscle, and liver. There are no specific treatments, and most male patients die from advanced heart failure during the second or third decade of life. DD is caused by mutations in the lysosomal-associated membrane protein 2 (LAMP2) gene, a key mediator of autophagy. LAMP2 has three isoforms: LAMP2A, LAMP2B, and LAMP2C. LAMP2B is the predominant isoform expressed in cardiomyocytes. This study evaluates the efficacy of human LAMP2B gene transfer using a recombinant adeno-associated virus 9 carrying human LAMP2B (AAV9.LAMP2B) in a Lamp2 knockout (KO) mouse, a DD model. AAV9.LAMP2B was intravenously injected into 2- and 6-month-old Lamp2 KO male mice to assess efficacy in adolescent and adult phenotypes. Lamp2 KO mice receiving AAV9.LAMP2B demonstrated dose-dependent restoration of human LAMP2B protein in the heart, liver, and skeletal muscle tissue. Impaired autophagic flux, evidenced by increased LC3-II, was abrogated by LAMP2B gene transfer in all tissues in both cohorts. Cardiac function was also improved, and transaminases were reduced in AAV9.LAMP2B-treated KO mice, indicating favorable effects on the heart and liver. Survival was also higher in the older cohort receiving high vector doses. No anti-LAMP2 antibodies were detected in mice that received AAV9.LAMP2B. In summary, LAMP2B gene transfer improves metabolic and physiologic function in a DD murine model, suggesting that a similar therapeutic approach may be effective for treating patients with this highly morbid disease.
Collapse
Affiliation(s)
- Ana Maria Manso
- Division of Cardiology, Department of Medicine, UC San Diego, San Diego, CA 92037, USA
| | - Sherin I Hashem
- Department of Pathology, UC San Diego, San Diego, CA 92037, USA
| | - Bradley C Nelson
- Division of Cardiology, Department of Medicine, UC San Diego, San Diego, CA 92037, USA
| | - Emily Gault
- Division of Cardiology, Department of Medicine, UC San Diego, San Diego, CA 92037, USA
| | - Angel Soto-Hermida
- Division of Cardiology, Department of Medicine, UC San Diego, San Diego, CA 92037, USA
| | - Elizza Villarruel
- Division of Cardiology, Department of Medicine, UC San Diego, San Diego, CA 92037, USA
| | - Michela Brambatti
- Division of Cardiology, Department of Medicine, UC San Diego, San Diego, CA 92037, USA
| | - Julius Bogomolovas
- Division of Cardiology, Department of Medicine, UC San Diego, San Diego, CA 92037, USA
| | - Paul J Bushway
- Division of Cardiology, Department of Medicine, UC San Diego, San Diego, CA 92037, USA
| | - Chao Chen
- Division of Cardiology, Department of Medicine, UC San Diego, San Diego, CA 92037, USA
| | | | | | | | - Gaurav Shah
- Rocket Pharmaceuticals, New York, NY 10118, USA
| | - Yusu Gu
- Division of Cardiology, Department of Medicine, UC San Diego, San Diego, CA 92037, USA
| | - Nancy D Dalton
- Division of Cardiology, Department of Medicine, UC San Diego, San Diego, CA 92037, USA
| | - Kirk Hammond
- Division of Cardiology, Department of Medicine, UC San Diego, San Diego, CA 92037, USA
| | - Kirk Peterson
- Division of Cardiology, Department of Medicine, UC San Diego, San Diego, CA 92037, USA
| | - Paul Saftig
- Biochemical Institute, Christian Albrechts-University, Kiel 24118, Germany
| | - Eric D Adler
- Division of Cardiology, Department of Medicine, UC San Diego, San Diego, CA 92037, USA.
| |
Collapse
|
24
|
Triolo M, Hood DA. Manifestations of Age on Autophagy, Mitophagy and Lysosomes in Skeletal Muscle. Cells 2021; 10:cells10051054. [PMID: 33946883 PMCID: PMC8146406 DOI: 10.3390/cells10051054] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 01/18/2023] Open
Abstract
Sarcopenia is the loss of both muscle mass and function with age. Although the molecular underpinnings of sarcopenia are not fully understood, numerous pathways are implicated, including autophagy, in which defective cargo is selectively identified and degraded at the lysosome. The specific tagging and degradation of mitochondria is termed mitophagy, a process important for the maintenance of an organelle pool that functions efficiently in energy production and with relatively low reactive oxygen species production. Emerging data, yet insufficient, have implicated various steps in this pathway as potential contributors to the aging muscle atrophy phenotype. Included in this is the lysosome, the end-stage organelle possessing a host of proteolytic and degradative enzymes, and a function devoted to the hydrolysis and breakdown of defective molecular complexes and organelles. This review provides a summary of our current understanding of how the autophagy-lysosome system is regulated in aging muscle, highlighting specific areas where knowledge gaps exist. Characterization of the autophagy pathway with a particular focus on the lysosome will undoubtedly pave the way for the development of novel therapeutic strategies to combat age-related muscle loss.
Collapse
Affiliation(s)
- Matthew Triolo
- Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada;
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada
| | - David A. Hood
- Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada;
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada
- Correspondence: ; Tel.: +(416)-736-2100 (ext. 66640)
| |
Collapse
|
25
|
Hong KN, Brambatti M, John S, Bui QM, Rigolli M, Taylor M, Adler ED. Recommendations and guidance on the diagnosis and management of Danon disease. Expert Opin Orphan Drugs 2021. [DOI: 10.1080/21678707.2021.1882994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Kimberly N. Hong
- Department of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Michela Brambatti
- Department of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Sonya John
- Department of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Quan M. Bui
- Department of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Marzia Rigolli
- Department of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Matthew Taylor
- CU-Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Eric D. Adler
- Department of Cardiology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
26
|
Pasqua T, Rocca C, Giglio A, Angelone T. Cardiometabolism as an Interlocking Puzzle between the Healthy and Diseased Heart: New Frontiers in Therapeutic Applications. J Clin Med 2021; 10:721. [PMID: 33673114 PMCID: PMC7918460 DOI: 10.3390/jcm10040721] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiac metabolism represents a crucial and essential connecting bridge between the healthy and diseased heart. The cardiac muscle, which may be considered an omnivore organ with regard to the energy substrate utilization, under physiological conditions mainly draws energy by fatty acids oxidation. Within cardiomyocytes and their mitochondria, through well-concerted enzymatic reactions, substrates converge on the production of ATP, the basic chemical energy that cardiac muscle converts into mechanical energy, i.e., contraction. When a perturbation of homeostasis occurs, such as an ischemic event, the heart is forced to switch its fatty acid-based metabolism to the carbohydrate utilization as a protective mechanism that allows the maintenance of its key role within the whole organism. Consequently, the flexibility of the cardiac metabolic networks deeply influences the ability of the heart to respond, by adapting to pathophysiological changes. The aim of the present review is to summarize the main metabolic changes detectable in the heart under acute and chronic cardiac pathologies, analyzing possible therapeutic targets to be used. On this basis, cardiometabolism can be described as a crucial mechanism in keeping the physiological structure and function of the heart; furthermore, it can be considered a promising goal for future pharmacological agents able to appropriately modulate the rate-limiting steps of heart metabolic pathways.
Collapse
Affiliation(s)
- Teresa Pasqua
- Department of Health Science, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy
| | - Anita Giglio
- Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy;
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy
- National Institute of Cardiovascular Research (I.N.R.C.), 40126 Bologna, Italy
| |
Collapse
|
27
|
The Role of iPSC Modeling Toward Projection of Autophagy Pathway in Disease Pathogenesis: Leader or Follower. Stem Cell Rev Rep 2020; 17:539-561. [PMID: 33245492 DOI: 10.1007/s12015-020-10077-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2020] [Indexed: 12/12/2022]
Abstract
Autophagy is responsible for degradation of non-essential or damaged cellular constituents and damaged organelles. The autophagy pathway maintains efficient cellular metabolism and reduces cellular stress by removing additional and pathogenic components. Dysfunctional autophagy underlies several diseases. Thus, several research groups have worked toward elucidating key steps in this pathway. Autophagy can be studied by animal modeling, chemical modulators, and in vitro disease modeling with induced pluripotent stem cells (iPSC) as a loss-of-function platform. The introduction of iPSC technology, which has the capability to maintain the genetic background, has facilitated in vitro modeling of some diseases. Furthermore, iPSC technology can be used as a platform to study defective cellular and molecular pathways during development and unravel novel steps in signaling pathways of health and disease. Different studies have used iPSC technology to explore the role of autophagy in disease pathogenesis which could not have been addressed by animal modeling or chemical inducers/inhibitors. In this review, we discuss iPSC models of autophagy-associated disorders where the disease is caused due to mutations in autophagy-related genes. We classified this group as "primary autophagy induced defects (PAID)". There are iPSC models of diseases in which the primary cause is not dysfunctional autophagy, but autophagy is impaired secondary to disease phenotypes. We call this group "secondary autophagy induced defects (SAID)" and discuss them. Graphical abstract.
Collapse
|
28
|
Lotan D, Salazar-Mendiguchía J, Mogensen J, Rathore F, Anastasakis A, Kaski J, Garcia-Pavia P, Olivotto I, Charron P, Biagini E, Baban A, Limongelli G, Ashram W, Wasserstrum Y, Galvin J, Zorio E, Iacovoni A, Monserrat L, Spirito P, Iascone M, Arad M. Clinical Profile of Cardiac Involvement in Danon Disease: A Multicenter European Registry. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2020; 13:e003117. [PMID: 33151750 DOI: 10.1161/circgen.120.003117] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The X-linked Danon disease manifests by severe cardiomyopathy, myopathy, and neuropsychiatric problems. We designed this registry to generate a comprehensive picture of clinical presentations and outcome of patients with Danon disease in cardiomyopathy centers throughout Europe. METHODS Clinical and genetic data were collected in 16 cardiology centers from 8 European countries. RESULTS The cohort comprised 30 male and 27 female patients. The age at diagnosis was birth to 42 years in men and 2 to 65 in women. Cardiac involvement was observed in 96%. Extracardiac manifestations were prominent in men but not in women. Left ventricular (LV) hypertrophy was reported in 73% of male and 74% of female patients. LV systolic dysfunction was reported in 40% of men (who had LV ejection fraction, 34±11%) and 59% of women (LV ejection fraction, 28±13%). The risk of arrhythmia and heart failure was comparable among sexes. The age of first heart failure hospitalization was lower in men (18±6 versus 28±17 years; P<0.003). Heart failure was the leading cause of death (10 of 17; 59%), and LV systolic dysfunction predicted an adverse outcome. Eight men and 8 women (28%) underwent heart transplantation or received an LV assist device. Our cohort suggests better prognosis of female compared with male heart transplant recipients. CONCLUSIONS Danon disease presents earlier in men than in women and runs a malignant course in both sexes, due to cardiac complications. Cardiomyopathy features, heart failure and arrhythmia, are similar among the sexes. Clinical diagnosis and management is extremely challenging in women due to phenotypic diversity and the absence of extracardiac manifestations.
Collapse
Affiliation(s)
- Dor Lotan
- Leviev Heart Center, Sheba Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (D.L., Y.W., M.A.)
| | - Joel Salazar-Mendiguchía
- Cardiomyopathy Unit, Hospital Universitari de Bellvitge, Barcelona, Spain (J.S.-M.).,Health in Code, Spain (J.S.-M.)
| | - Jens Mogensen
- Department of Cardiology, Odense University Hospital, Denmark (J.M.)
| | - Faizan Rathore
- Department of Cardiology, University College Dublin School of Medicine, Mater Misericordiae University Hospital, Ireland (F.R., W.A., J.G.)
| | - Aris Anastasakis
- Unit of Inherited Diseases, Onassis Cardiac Surgery Center, Athens, Greece (A.A.)
| | - Juan Kaski
- Great Ormond Street Hospital Center for Inherited Cardiovascular Diseases, University College London Institute of Cardiovascular Science, United Kingdom (J.K.).,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (J.K., P.G.-P., P.C., A.B., G.L.)
| | - Pablo Garcia-Pavia
- Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, CIBERCV, Madrid, Spain (P.G.-P.).,Universidad Francisco de Vitoria, Pozuelo de Alarcón, Spain (P.G.-P.).,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (J.K., P.G.-P., P.C., A.B., G.L.)
| | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O.)
| | - Philippe Charron
- Sorbonne Université, Paris, France (P.C.).,APHP, Centre de référence des maladies cardiaques héréditaires ou rares, Paris, France (P.C.).,INSERM UMR S1166, ICAN, Paris, France (P.C.).,Hôpital Pitié-Salpêtrière, Paris, France (P.C.).,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (J.K., P.G.-P., P.C., A.B., G.L.)
| | - Elena Biagini
- Cardio-Thoracic-Vascular Department, S. Orsola Hospital, University of Bologna, Bologna, Italy (E.B.)
| | - Anwar Baban
- Pediatric Cardiology and Arrhythmia/Syncope Units, Bambino Gesù Children Hospital and Research Institute, Rome (A.B.).,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (J.K., P.G.-P., P.C., A.B., G.L.)
| | - Giuseppe Limongelli
- Monaldi Hospital, AO Colli, Second University of Naples, Italy (G.L.).,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (J.K., P.G.-P., P.C., A.B., G.L.)
| | - Waddah Ashram
- Department of Cardiology, University College Dublin School of Medicine, Mater Misericordiae University Hospital, Ireland (F.R., W.A., J.G.)
| | - Yishay Wasserstrum
- Leviev Heart Center, Sheba Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (D.L., Y.W., M.A.)
| | - Joseph Galvin
- Department of Cardiology, University College Dublin School of Medicine, Mater Misericordiae University Hospital, Ireland (F.R., W.A., J.G.)
| | - Esther Zorio
- Inherited Heart Diseases Unit and CaFaMuSMe Research Group, Hospital Universitari i Politècnic La Fe and IIS La Fe, Valencia and Center for Biomedical Network Research on Cardiovascular Diseases (CIBERCV), Madrid, Spain (E.Z.)
| | - Attilio Iacovoni
- Department of Cardiology, ASST Papa Giovanni XXIII, Bergamo, Italy (A.I.)
| | - Lorenzo Monserrat
- Health in Code, Hospital Marítimo de Oza, As Xubias, A Coruña, Spain (L.M.)
| | - Paolo Spirito
- Hypertrophic Cardiomyopathy Center, Policlinico di Monza, Italy (P.S.)
| | - Maria Iascone
- Molecular Genetics Laboratory, A.O. Papa Giovanni XXIII, Bergamo, Italy (M.I.)
| | - Michael Arad
- Leviev Heart Center, Sheba Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (D.L., Y.W., M.A.)
| | | |
Collapse
|
29
|
Danon Disease-Associated LAMP-2 Deficiency Drives Metabolic Signature Indicative of Mitochondrial Aging and Fibrosis in Cardiac Tissue and hiPSC-Derived Cardiomyocytes. J Clin Med 2020; 9:jcm9082457. [PMID: 32751926 PMCID: PMC7465084 DOI: 10.3390/jcm9082457] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/15/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
Danon disease is a severe X-linked disorder caused by deficiency of the lysosome-associated membrane protein-2 (LAMP-2). Clinical manifestations are phenotypically diverse and consist of hypertrophic and dilated cardiomyopathies, skeletal myopathy, retinopathy, and intellectual dysfunction. Here, we investigated the metabolic landscape of Danon disease by applying a multi-omics approach and combined structural and functional readouts provided by Raman and atomic force microscopy. Using these tools, Danon patient-derived cardiac tissue, primary fibroblasts, and human induced pluripotent stem cells differentiated into cardiomyocytes (hiPSC-CMs) were analyzed. Metabolic profiling indicated LAMP-2 deficiency promoted a switch toward glycolysis accompanied by rerouting of tryptophan metabolism. Cardiomyocytes' energetic balance and NAD+/NADH ratio appeared to be maintained despite mitochondrial aging. In turn, metabolic adaption was accompanied by a senescence-associated signature. Similarly, Danon fibroblasts appeared more stress prone and less biomechanically compliant. Overall, shaping of both morphology and metabolism contributed to the loss of cardiac biomechanical competence that characterizes the clinical progression of Danon disease.
Collapse
|
30
|
Bakula D, Scheibye-Knudsen M. MitophAging: Mitophagy in Aging and Disease. Front Cell Dev Biol 2020; 8:239. [PMID: 32373609 PMCID: PMC7179682 DOI: 10.3389/fcell.2020.00239] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/23/2020] [Indexed: 12/19/2022] Open
Abstract
Maintaining mitochondrial health is emerging as a keystone in aging and associated diseases. The selective degradation of mitochondria by mitophagy is of particular importance in keeping a pristine mitochondrial pool. Indeed, inherited monogenic diseases with defects in mitophagy display complex multisystem pathologies but particularly progressive neurodegeneration. Fortunately, therapies are being developed that target mitophagy allowing new hope for treatments for previously incurable diseases. Herein, we describe mitophagy and associated diseases, coin the term mitophaging and describe new small molecule interventions that target different steps in the mitophagic pathway. Consequently, several age-associated diseases may be treated by targeting mitophagy.
Collapse
Affiliation(s)
- Daniela Bakula
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Morten Scheibye-Knudsen
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
31
|
Campos JC, Baehr LM, Ferreira ND, Bozi LHM, Andres AM, Ribeiro MAC, Gottlieb RA, Bodine SC, Ferreira JCB. β 2 -adrenoceptor activation improves skeletal muscle autophagy in neurogenic myopathy. FASEB J 2020; 34:5628-5641. [PMID: 32112488 DOI: 10.1096/fj.201902305r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 02/03/2020] [Accepted: 02/14/2020] [Indexed: 01/21/2023]
Abstract
β2 -adrenoceptor agonists improve autophagy and re-establish proteostasis in cardiac cells; therefore, suggesting autophagy as a downstream effector of β2 -adrenoceptor signaling pathway. Here, we used the pharmacological and genetic tools to determine the autophagy effect of sustained β2 -adrenoceptor activation in rodents with neurogenic myopathy, which display impaired skeletal muscle autophagic flux. Sustained β2 -adrenoceptor activation using Formoterol (10 μg kg-1 day-1 ), starting at the onset of neurogenic myopathy, prevents disruption of autophagic flux in skeletal muscle 14 days after sciatic nerve constriction. These changes are followed by reduction of the cytotoxic protein levels and increased skeletal muscle cross-sectional area and contractility properties. Of interest, sustained administration of Formoterol at lower concentration (1 μg kg-1 day-1 ) induces similar improvements in skeletal muscle autophagic flux and contractility properties in neurogenic myopathy, without affecting the cross-sectional area. Sustained pharmacological inhibition of autophagy using Chloroquine (50 mg kg-1 day-1 ) abolishes the beneficial effects of β2 -adrenoceptor activation on the skeletal muscle proteostasis and contractility properties in neurogenic myopathy. Further supporting an autophagy mechanism for β2 -adrenoceptor activation, skeletal muscle-specific deletion of ATG7 blunts the beneficial effects of β2 -adrenoceptor on skeletal muscle proteostasis and contractility properties in neurogenic myopathy in mice. These findings suggest autophagy as a critical downstream effector of β2 -adrenoceptor signaling pathway in skeletal muscle.
Collapse
Affiliation(s)
- Juliane C Campos
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Leslie M Baehr
- Department of Internal Medicine, Endocrinology and Metabolism Division, University of Iowa, Iowa City, IA, USA
| | - Nikolas D Ferreira
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Luiz H M Bozi
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Allen M Andres
- The Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Márcio A C Ribeiro
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Roberta A Gottlieb
- The Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sue C Bodine
- Department of Internal Medicine, Endocrinology and Metabolism Division, University of Iowa, Iowa City, IA, USA
| | - Julio C B Ferreira
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Department of Chemical & Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
32
|
Ulmer BM, Eschenhagen T. Human pluripotent stem cell-derived cardiomyocytes for studying energy metabolism. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118471. [PMID: 30954570 PMCID: PMC7042711 DOI: 10.1016/j.bbamcr.2019.04.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/26/2019] [Accepted: 04/01/2019] [Indexed: 12/25/2022]
Abstract
Cardiomyocyte energy metabolism is altered in heart failure, and primary defects of metabolic pathways can cause heart failure. Studying cardiac energetics in rodent models has principal shortcomings, raising the question to which extent human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CM) can provide an alternative. As metabolic maturation of CM occurs mostly after birth during developmental hypertrophy, the immaturity of hiPSC-CM is an important limitation. Here we shortly review the physiological drivers of metabolic maturation and concentrate on methods to mature hiPSC-CM with the goal to benchmark the metabolic state of hiPSC-CM against in vivo data and to see how far known abnormalities in inherited metabolic disorders can be modeled in hiPSC-CM. The current data indicate that hiPSC-CM, despite their immature, approximately mid-fetal state of energy metabolism, faithfully recapitulate some basic metabolic disease mechanisms. Efforts to improve their metabolic maturity are underway and shall improve the validity of this model.
Collapse
Affiliation(s)
- Bärbel M Ulmer
- University Medical Center Hamburg-Eppendorf, Institute of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Centre for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | - Thomas Eschenhagen
- University Medical Center Hamburg-Eppendorf, Institute of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Centre for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
33
|
Farah BL, Yen PM, Koeberl DD. Links between autophagy and disorders of glycogen metabolism - Perspectives on pathogenesis and possible treatments. Mol Genet Metab 2020; 129:3-12. [PMID: 31787497 PMCID: PMC7836271 DOI: 10.1016/j.ymgme.2019.11.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 01/17/2023]
Abstract
The glycogen storage diseases are a group of inherited metabolic disorders that are characterized by specific enzymatic defects involving the synthesis or degradation of glycogen. Each disorder presents with a set of symptoms that are due to the underlying enzyme deficiency and the particular tissues that are affected. Autophagy is a process by which cells degrade and recycle unneeded or damaged intracellular components such as lipids, glycogen, and damaged mitochondria. Recent studies showed that several of the glycogen storage disorders have abnormal autophagy which can disturb normal cellular metabolism and/or mitochondrial function. Here, we provide a clinical overview of the glycogen storage disorders, a brief description of autophagy, and the known links between specific glycogen storage disorders and autophagy.
Collapse
Affiliation(s)
- Benjamin L Farah
- Department of Pathology, Singapore General Hospital, Singapore, Singapore.
| | - Paul M Yen
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Dwight D Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, NC, USA; Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA..
| |
Collapse
|
34
|
Sacchetto C, Sequeira V, Bertero E, Dudek J, Maack C, Calore M. Metabolic Alterations in Inherited Cardiomyopathies. J Clin Med 2019; 8:E2195. [PMID: 31842377 PMCID: PMC6947282 DOI: 10.3390/jcm8122195] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022] Open
Abstract
The normal function of the heart relies on a series of complex metabolic processes orchestrating the proper generation and use of energy. In this context, mitochondria serve a crucial role as a platform for energy transduction by supplying ATP to the varying demand of cardiomyocytes, involving an intricate network of pathways regulating the metabolic flux of substrates. The failure of these processes results in structural and functional deficiencies of the cardiac muscle, including inherited cardiomyopathies. These genetic diseases are characterized by cardiac structural and functional anomalies in the absence of abnormal conditions that can explain the observed myocardial abnormality, and are frequently associated with heart failure. Since their original description, major advances have been achieved in the genetic and phenotype knowledge, highlighting the involvement of metabolic abnormalities in their pathogenesis. This review provides a brief overview of the role of mitochondria in the energy metabolism in the heart and focuses on metabolic abnormalities, mitochondrial dysfunction, and storage diseases associated with inherited cardiomyopathies.
Collapse
Affiliation(s)
- Claudia Sacchetto
- IMAiA—Institute for Molecular Biology and RNA Technology, Faculty of Health, Universiteitssingel 50, 6229ER Maastricht, The Netherlands;
- Medicine and Life Sciences, Faculty of Science and Engineering, Universiteitssingel 50, 6229ER Maastricht, The Netherlands
- Department of Biology, University of Padova, via Ugo Bassi 58B, 35121 Padova, Italy
| | - Vasco Sequeira
- Department of Translational Science, Comprehensive Heart Failure Center, University Clinic Würzburg, Am Schwarzenberg 15, 9708 Würzburg, Germany; (V.S.); (E.B.); (J.D.)
| | - Edoardo Bertero
- Department of Translational Science, Comprehensive Heart Failure Center, University Clinic Würzburg, Am Schwarzenberg 15, 9708 Würzburg, Germany; (V.S.); (E.B.); (J.D.)
| | - Jan Dudek
- Department of Translational Science, Comprehensive Heart Failure Center, University Clinic Würzburg, Am Schwarzenberg 15, 9708 Würzburg, Germany; (V.S.); (E.B.); (J.D.)
| | - Christoph Maack
- Department of Translational Science, Comprehensive Heart Failure Center, University Clinic Würzburg, Am Schwarzenberg 15, 9708 Würzburg, Germany; (V.S.); (E.B.); (J.D.)
| | - Martina Calore
- IMAiA—Institute for Molecular Biology and RNA Technology, Faculty of Health, Universiteitssingel 50, 6229ER Maastricht, The Netherlands;
- Medicine and Life Sciences, Faculty of Science and Engineering, Universiteitssingel 50, 6229ER Maastricht, The Netherlands
| |
Collapse
|
35
|
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: 6.4] [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
|
36
|
Brodehl A, Ebbinghaus H, Deutsch MA, Gummert J, Gärtner A, Ratnavadivel S, Milting H. Human Induced Pluripotent Stem-Cell-Derived Cardiomyocytes as Models for Genetic Cardiomyopathies. Int J Mol Sci 2019; 20:ijms20184381. [PMID: 31489928 PMCID: PMC6770343 DOI: 10.3390/ijms20184381] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022] Open
Abstract
In the last few decades, many pathogenic or likely pathogenic genetic mutations in over hundred different genes have been described for non-ischemic, genetic cardiomyopathies. However, the functional knowledge about most of these mutations is still limited because the generation of adequate animal models is time-consuming and challenging. Therefore, human induced pluripotent stem cells (iPSCs) carrying specific cardiomyopathy-associated mutations are a promising alternative. Since the original discovery that pluripotency can be artificially induced by the expression of different transcription factors, various patient-specific-induced pluripotent stem cell lines have been generated to model non-ischemic, genetic cardiomyopathies in vitro. In this review, we describe the genetic landscape of non-ischemic, genetic cardiomyopathies and give an overview about different human iPSC lines, which have been developed for the disease modeling of inherited cardiomyopathies. We summarize different methods and protocols for the general differentiation of human iPSCs into cardiomyocytes. In addition, we describe methods and technologies to investigate functionally human iPSC-derived cardiomyocytes. Furthermore, we summarize novel genome editing approaches for the genetic manipulation of human iPSCs. This review provides an overview about the genetic landscape of inherited cardiomyopathies with a focus on iPSC technology, which might be of interest for clinicians and basic scientists interested in genetic cardiomyopathies.
Collapse
Affiliation(s)
- Andreas Brodehl
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Hans Ebbinghaus
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Marcus-André Deutsch
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Jan Gummert
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Anna Gärtner
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Sandra Ratnavadivel
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| |
Collapse
|
37
|
Dvornikov AV, Wang M, Yang J, Zhu P, Le T, Lin X, Cao H, Xu X. Phenotyping an adult zebrafish lamp2 cardiomyopathy model identifies mTOR inhibition as a candidate therapy. J Mol Cell Cardiol 2019; 133:199-208. [PMID: 31228518 PMCID: PMC6705397 DOI: 10.1016/j.yjmcc.2019.06.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/31/2019] [Accepted: 06/18/2019] [Indexed: 12/23/2022]
Abstract
Adult zebrafish is an emerging vertebrate model for studying genetic basis of cardiomyopathies; but whether the simple fish heart can model essential features of hypertrophic cardiomyopathy (HCM) remained unknown. Here, we report a comprehensive phenotyping of a lamp2 knockout (KO) mutant. LAMP2 encodes a lysosomal protein and is a causative gene of Danon disease that is characterized by HCM and massive autophagic vacuoles accumulation in the tissues. There is no effective therapy yet to treat this most lethal cardiomyopathy in the young. First, we did find the autophagic vacuoles accumulation in cardiac tissues from lamp2 KO. Next, through employing a set of emerging phenotyping tools, we revealed heart failure phenotypes in the lamp2 KO mutants, including decreased ventricular ejection fraction, reduced physical exercise capacity, blunted β-adrenergic contractile response, and enlarged atrium. We also noted changes of the following indices suggesting cardiac hypertrophic remodeling in lamp2 KO: a rounded heart shape, increased end-systolic ventricular volume and density of ventricular myocardium, elevated actomyosin activation kinetics together with increased maximal isometric tension at the level of cardiac myofibrils. Lastly, we assessed the function of lysosomal-localized mTOR on the lamp2-associated Danon disease. We found that haploinsufficiency of mtor was able to normalize some characteristics of the lamp2 KO, including ejection fraction, β-adrenergic response, and the actomyosin activation kinetics. In summary, we demonstrate the feasibility of modeling the inherited HCM in the adult zebrafish, which can be used to develop potential therapies.
Collapse
Affiliation(s)
- Alexey V Dvornikov
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Mingmin Wang
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jingchun Yang
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ping Zhu
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Tai Le
- Department of Electrical Engineering and Computer Science, University of California Irvine, CA, USA
| | - Xueying Lin
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Hung Cao
- Department of Electrical Engineering and Computer Science, University of California Irvine, CA, USA; Department of Biomedical Engineering, University of California Irvine, CA, USA
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
38
|
Brambatti M, Caspi O, Maolo A, Koshi E, Greenberg B, Taylor MRG, Adler ED. Danon disease: Gender differences in presentation and outcomes. Int J Cardiol 2019; 286:92-98. [PMID: 30857840 DOI: 10.1016/j.ijcard.2019.01.020] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/02/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Danon disease (DD) is a rare X-linked autophagic vacuolar myopathy, characterized by high penetrance and severe cardiomyopathy. Because of its rarity, the natural history (NH) is uncertain. OBJECTIVES We aimed to describe disease variability and outcomes through a systematic review of all published DD cases. METHODS Among 83 manuscripts in MEDLINE and EMBASE on DD cases published until October 2017, we identified 146 patients with positive genetic testing for DD or positive muscle biopsy in a relative of a genetically diagnosed proband. RESULTS 56 females and 90 males were identified. 92.5% of patients had cardiac abnormalities. Females presented with either hypertrophic cardiomyopathy (HCM, 70.3%) or dilated cardiomyopathy (DCM, 29.3%) whereas males presented with HCM 96.2% of the time. The composite outcome of death, heart transplant or ventricular assist devices occurred equally in both sexes (32% of females and 37% of males, p = 0.60) but later in females (median age 38 years) than in males (median age 21 years, p < 0.001). Whereas women present with isolated cardiac disease 73% of the time, in males DD was frequently multisystemic and presented as a triad of cognitive impairment, skeletal myopathy, and HCM in 42% of patients. CONCLUSIONS In this first systematic review of DD, we confirmed the severe morbidity and mortality associated with disease in both sexes. Women presented with both HCM and DCM and generally with isolated cardiac disease, whereas in men DD usually presented as HCM and was frequently multi-systemic. Further prospective NH studies will be required to confirm these findings.
Collapse
Affiliation(s)
- Michela Brambatti
- Division of Cardiology, University of California San Diego, San Diego, CA, USA
| | - Oren Caspi
- Division of Cardiology, University of California San Diego, San Diego, CA, USA
| | - Alessandro Maolo
- Division of Cardiology, University of California San Diego, San Diego, CA, USA
| | - Elliott Koshi
- Division of Cardiology, University of California San Diego, San Diego, CA, USA
| | - Barry Greenberg
- Division of Cardiology, University of California San Diego, San Diego, CA, USA
| | - Matthew R G Taylor
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, CO, USA
| | - Eric D Adler
- Division of Cardiology, University of California San Diego, San Diego, CA, USA.
| |
Collapse
|
39
|
LAMP-2B regulates human cardiomyocyte function by mediating autophagosome-lysosome fusion. Proc Natl Acad Sci U S A 2018; 116:556-565. [PMID: 30584088 DOI: 10.1073/pnas.1808618116] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mutations in lysosomal-associated membrane protein 2 (LAMP-2) gene are associated with Danon disease, which often leads to cardiomyopathy/heart failure through poorly defined mechanisms. Here, we identify the LAMP-2 isoform B (LAMP-2B) as required for autophagosome-lysosome fusion in human cardiomyocytes (CMs). Remarkably, LAMP-2B functions independently of syntaxin 17 (STX17), a protein that is essential for autophagosome-lysosome fusion in non-CMs. Instead, LAMP-2B interacts with autophagy related 14 (ATG14) and vesicle-associated membrane protein 8 (VAMP8) through its C-terminal coiled coil domain (CCD) to promote autophagic fusion. CMs derived from induced pluripotent stem cells (hiPSC-CMs) from Danon patients exhibit decreased colocalization between ATG14 and VAMP8, profound defects in autophagic fusion, as well as mitochondrial and contractile abnormalities. This phenotype was recapitulated by LAMP-2B knockout in non-Danon hiPSC-CMs. Finally, gene correction of LAMP-2 mutation rescues the Danon phenotype. These findings reveal a STX17-independent autophagic fusion mechanism in human CMs, providing an explanation for cardiomyopathy in Danon patients and a foundation for targeting defective LAMP-2B-mediated autophagy to treat this patient population.
Collapse
|
40
|
Li J, Zhang D, Wiersma M, Brundel BJJM. Role of Autophagy in Proteostasis: Friend and Foe in Cardiac Diseases. Cells 2018; 7:cells7120279. [PMID: 30572675 PMCID: PMC6316637 DOI: 10.3390/cells7120279] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/13/2018] [Accepted: 12/18/2018] [Indexed: 12/11/2022] Open
Abstract
Due to ageing of the population, the incidence of cardiovascular diseases will increase in the coming years, constituting a substantial burden on health care systems. In particular, atrial fibrillation (AF) is approaching epidemic proportions. It has been identified that the derailment of proteostasis, which is characterized by the loss of homeostasis in protein biosynthesis, folding, trafficking, and clearance by protein degradation systems such as autophagy, underlies the development of common cardiac diseases. Among various safeguards within the proteostasis system, autophagy is a vital cellular process that modulates clearance of misfolded and proteotoxic proteins from cardiomyocytes. On the other hand, excessive autophagy may result in derailment of proteostasis and therefore cardiac dysfunction. Here, we review the interplay between autophagy and proteostasis in the healthy heart, discuss the imbalance between autophagy and proteostasis during cardiac diseases, including AF, and finally explore new druggable targets which may limit cardiac disease initiation and progression.
Collapse
Affiliation(s)
- Jin Li
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands.
| | - Deli Zhang
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands.
| | - Marit Wiersma
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands.
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
41
|
Triolo M, Hood DA. Mitochondrial breakdown in skeletal muscle and the emerging role of the lysosomes. Arch Biochem Biophys 2018; 661:66-73. [PMID: 30439362 DOI: 10.1016/j.abb.2018.11.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/13/2018] [Accepted: 11/05/2018] [Indexed: 11/15/2022]
Abstract
Skeletal muscle mitochondria are essential in providing the energy required for locomotion. In response to contractile activity, the production of mitochondria is upregulated to meet the energy demands placed upon muscle cells. In a coordinated fashion, exercise also promotes the breakdown of dysfunctional mitochondria via mitophagy. Mitophagy is characterized by the selection of poorly functioning organelles, engulfment in an autophagosome and transport to lysosomes for degradation. In addition to the activation of mitophagy, exercise also elevates lysosome biogenesis. This coordinated increase in mitophagy targeting and lysosomal biogenesis serves to enhance the capacity for autophagosomal degradation, thereby aiding in the maintenance of mitochondrial quality. Lysosome dysfunction, as observed in lysosomal storage disorders (LSDs), negatively impacts mitochondrial function likely through the suppression of mitophagy. Since exercise is capable of activating mitophagy and lysosome biogenesis, researchers have begun to investigate physical activity as an effective therapy for LSDs. This review summarizes the current understanding of how mitophagy and lysosomal biogenesis are regulated in exercising skeletal, with potential therapeutic implications.
Collapse
Affiliation(s)
- Matthew Triolo
- Muscle Health Research Centre, York University, Toronto, Ontario, M3J 1P3, Canada; School of Kinesiology and Health Science, York University, Toronto, Ontario, M3J 1P3, Canada.
| | - David A Hood
- Muscle Health Research Centre, York University, Toronto, Ontario, M3J 1P3, Canada; School of Kinesiology and Health Science, York University, Toronto, Ontario, M3J 1P3, Canada.
| |
Collapse
|
42
|
Tan Y, Gong Y, Dong M, Pei Z, Ren J. Role of autophagy in inherited metabolic and endocrine myopathies. Biochim Biophys Acta Mol Basis Dis 2018; 1865:48-55. [PMID: 30343140 DOI: 10.1016/j.bbadis.2018.10.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/11/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022]
Abstract
The prevalence of cardiometabolic disease has reached an exponential rate of rise over the last decades owing to high fat/high caloric diet intake and satiety life style. Although the presence of dyslipidemia, insulin resistance, hypertension and obesity mainly contributes to the increased incidence of cardiometabolic diseases, population-based, clinical and genetic studies have revealed a rather important role for inherited myopathies and endocrine disorders in the ever-rising metabolic anomalies. Inherited metabolic and endocrine diseases such as glycogen storage and lysosomal disorders have greatly contributed to the overall prevalence of cardiometabolic diseases. Recent evidence has demonstrated an essential role for proteotoxicity due to autophagy failure and/or dysregulation in the onset of inherited metabolic and endocrine disorders. Given the key role for autophagy in the degradation and removal of long-lived or injured proteins and organelles for the maintenance of cellular and organismal homeostasis, this mini-review will discuss the potential contribution of autophagy dysregulation in the pathogenesis of inherited myopathies and endocrine disorders, which greatly contribute to an overall rise in prevalence of cardiometabolic disorders. Molecular, clinical, and epidemiological aspects will be covered as well as the potential link between autophagy and metabolic anomalies thus target therapy may be engaged for these comorbidities.
Collapse
Affiliation(s)
- Ying Tan
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Yan Gong
- The Second Department of Cardiology, The Third Hospital of Nanchang, Nanchang, Jiangxi 330009, China
| | - Maolong Dong
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA; Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhaohui Pei
- The Second Department of Cardiology, The Third Hospital of Nanchang, Nanchang, Jiangxi 330009, China.
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| |
Collapse
|
43
|
Eschenhagen T, Carrier L. Cardiomyopathy phenotypes in human-induced pluripotent stem cell-derived cardiomyocytes-a systematic review. Pflugers Arch 2018; 471:755-768. [PMID: 30324321 PMCID: PMC6475632 DOI: 10.1007/s00424-018-2214-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/19/2018] [Accepted: 10/02/2018] [Indexed: 12/12/2022]
Abstract
Human-induced pluripotent stem cells (hiPSC) can be differentiated to cardiomyocytes at high efficiency and are increasingly used to study cardiac disease in a human context. This review evaluated 38 studies on hypertrophic (HCM) and dilated cardiomyopathy (DCM) of different genetic causes asking to which extent published data allow the definition of an in vitro HCM/DCM hiPSC-CM phenotype. The data are put in context with the prevailing hypotheses on HCM/DCM dysfunction and pathophysiology. Relatively consistent findings in HCM not reported in DCM were larger cell size (156 ± 85%, n = 15), more nuclear localization of nuclear factor of activated T cells (NFAT; 175 ± 65%, n = 3), and higher β-myosin heavy chain gene expression levels (500 ± 547%, n = 8) than respective controls. Conversely, DCM lines showed consistently less force development than controls (47 ± 23%, n = 9), while HCM forces scattered without clear trend. Both HCM and DCM lines often showed sarcomere disorganization, higher NPPA/NPPB expression levels, and arrhythmic beating behaviour. The data have to be taken with the caveat that reporting frequencies of the various parameters (e.g. cell size, NFAT expression) differ widely between HCM and DCM lines, in which data scatter is large and that only 9/38 studies used isogenic controls. Taken together, the current data provide interesting suggestions for disease-specific phenotypes in HCM/DCM hiPSC-CM but indicate that the field is still in its early days. Systematic, quantitative comparisons and robust, high content assays are warranted to advance the field.
Collapse
Affiliation(s)
- Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany.
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany.
| |
Collapse
|
44
|
Multiple recycling routes: Canonical vs. non-canonical mitophagy in the heart. Biochim Biophys Acta Mol Basis Dis 2018; 1865:797-809. [PMID: 30290272 DOI: 10.1016/j.bbadis.2018.09.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/13/2018] [Accepted: 09/27/2018] [Indexed: 12/14/2022]
Abstract
The heart is composed of cardiomyocytes that require large amounts of energy to sustain contraction. Mitochondria are distinctive organelles of bacterial origin that generate most of the energy for the heart via oxidative phosphorylation. To ensure a healthy population of mitochondria that efficiently produce ATP, myocytes quickly eliminate any unhealthy or unwanted mitochondria via a process known as mitochondrial autophagy, or mitophagy. It is especially important to selectively remove damaged or aged mitochondria since they can become excessive producers of reactive oxygen species and release pro-death proteins. Because this is such a crucial cellular process, cells have several mechanisms in place to deal with potentially harmful mitochondria. Here, we review the various pathways identified to date and how they are regulated. We also discuss the importance of these canonical and non-canonical pathways in the heart and their link to cardiovascular health, disease and aging.
Collapse
|
45
|
Parousis A, Carter HN, Tran C, Erlich AT, Mesbah Moosavi ZS, Pauly M, Hood DA. Contractile activity attenuates autophagy suppression and reverses mitochondrial defects in skeletal muscle cells. Autophagy 2018; 14:1886-1897. [PMID: 30078345 PMCID: PMC6152519 DOI: 10.1080/15548627.2018.1491488] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 06/05/2018] [Accepted: 06/18/2018] [Indexed: 12/18/2022] Open
Abstract
Macroautophagy/autophagy is a survival mechanism that facilitates protein turnover in post-mitotic cells in a lysosomal-dependent process. Mitophagy is a selective form of autophagy, which arbitrates the selective recognition and targeting of aberrant mitochondria for degradation. Mitochondrial content in cells is the net balance of mitochondrial catabolism via mitophagy, and organelle biogenesis. Although the latter process has been well described, mitophagy in skeletal muscle is less understood, and it is currently unknown how these two opposing mechanisms converge during contractile activity. Here we show that chronic contractile activity (CCA) in muscle cells induced mitochondrial biogenesis and coordinately enhanced the expression of TFEB (transcription factor EB) and PPARGC1A/PGC-1α, master regulators of lysosome and mitochondrial biogenesis, respectively. CCA also enhanced the expression of PINK1 and the lysosomal protease CTSD (cathepsin D). Autophagy blockade with bafilomycin A1 (BafA) reduced mitochondrial state 3 and 4 respiration, increased ROS production and enhanced the accumulation of MAP1LC3B-II/LC3-II and SQSTM1/p62. CCA ameliorated this mitochondrial dysfunction during defective autophagy, increased PPARGC1A, normalized LC3-II levels and reversed mitochondrially-localized SQSTM1 toward control levels. NAC emulated the LC3-II reductions induced by contractile activity, signifying that a decrease in oxidative stress could represent a mechanism of autophagy normalization brought about by CCA. CCA enhances mitochondrial biogenesis and lysosomal activity, and normalizes autophagy flux during autophagy suppression, partly via ROS-dependent mechanisms. Thus, contractile activity represents a potential therapeutic intervention for diseases in which autophagy is inhibited, such as vacuolar myopathies in skeletal muscle, by establishing a healthy equilibrium of anabolic and catabolic pathways. ABBREVIATIONS AMPK: AMP-activated protein kinase; BafA: bafilomycin A1; BNIP3L: BCL2/adenovirus E1B interacting protein 3-like; CCA: chronic contractile activity; COX4I1: cytochrome c oxidase subunit 4I1; DMEM: Dulbecco's modified Eagle's medium; GFP: green fluorescent protein; LSD: lysosomal storage diseases; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MTORC1: mechanistic target of rapamycin kinase complex 1; NAC: N-acetylcysteine; PPARGC1A: peroxisome proliferative activated receptor, gamma, coactivator 1 alpha; PINK1: PTEN induced putative kinase 1; ROS: reactive oxygen species; SOD2: superoxide dismutase 2, mitochondrial; SQSTM1/p62: sequestosome 1; TFEB: transcription factor EB.
Collapse
Affiliation(s)
- Alexa Parousis
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Heather N. Carter
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Claudia Tran
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Avigail T. Erlich
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Zahra S. Mesbah Moosavi
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Marion Pauly
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - David A. Hood
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| |
Collapse
|
46
|
Anderson RH, Francis KR. Modeling rare diseases with induced pluripotent stem cell technology. Mol Cell Probes 2018; 40:52-59. [PMID: 29307697 PMCID: PMC6033695 DOI: 10.1016/j.mcp.2018.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 12/22/2017] [Accepted: 01/02/2018] [Indexed: 12/18/2022]
Abstract
Rare diseases, in totality, affect a significant proportion of the population and represent an unmet medical need facing the scientific community. However, the treatment of individuals affected by rare diseases is hampered by poorly understood mechanisms preventing the development of viable therapeutics. The discovery and application of cellular reprogramming to create novel induced pluripotent stem cell models of rare diseases has revolutionized the rare disease community. Through developmental and functional analysis of differentiated cell types, these stem cell models carrying patient-specific mutations have become an invaluable tool for rare disease research. In this review article, we discuss the reprogramming of samples from individuals affected with rare diseases to induced pluripotent stem cells, current and future applications for this technology, and how integration of genome editing to rare disease research will help to improve our understanding of disease pathogenesis and lead to patient therapies.
Collapse
Affiliation(s)
- Ruthellen H Anderson
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD, USA; Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| | - Kevin R Francis
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD, USA; Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA.
| |
Collapse
|
47
|
Broyles CN, Robinson P, Daniels MJ. Fluorescent, Bioluminescent, and Optogenetic Approaches to Study Excitable Physiology in the Single Cardiomyocyte. Cells 2018; 7:cells7060051. [PMID: 29857560 PMCID: PMC6028913 DOI: 10.3390/cells7060051] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/22/2018] [Accepted: 05/30/2018] [Indexed: 12/11/2022] Open
Abstract
This review briefly summarizes the single cell application of classical chemical dyes used to visualize cardiomyocyte physiology and their undesirable toxicities which have the potential to confound experimental observations. We will discuss, in detail, the more recent iterative development of fluorescent and bioluminescent protein-based indicators and their emerging application to cardiomyocytes. We will discuss the integration of optical control strategies (optogenetics) to augment the standard imaging approach. This will be done in the context of potential applications, and barriers, of these technologies to disease modelling, drug toxicity, and drug discovery efforts at the single-cell scale.
Collapse
Affiliation(s)
- Connor N Broyles
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK.
- BHF Centre of Research Excellence, University of Oxford, Oxford OX3 9DU, UK.
| | - Paul Robinson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK.
- BHF Centre of Research Excellence, University of Oxford, Oxford OX3 9DU, UK.
| | - Matthew J Daniels
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK.
- BHF Centre of Research Excellence, University of Oxford, Oxford OX3 9DU, UK.
- Department of Cardiology, Oxford University NHS Hospitals Trust, Oxford OX3 9DU, UK.
- BHF Centre of Regenerative Medicine, University of Oxford, Oxford OX3 9DU, UK.
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Mihogaoka 8-1, Ibaraki, 567-0047 Osaka, Japan.
| |
Collapse
|
48
|
Saha S, Panigrahi DP, Patil S, Bhutia SK. Autophagy in health and disease: A comprehensive review. Biomed Pharmacother 2018; 104:485-495. [PMID: 29800913 DOI: 10.1016/j.biopha.2018.05.007] [Citation(s) in RCA: 358] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/12/2018] [Accepted: 05/07/2018] [Indexed: 12/13/2022] Open
Abstract
Autophagy, a conserved catabolic process, plays an immensely significant role in a variety of diseases. However, whether it imparts a protective function in diseases remains debatable. During aging, autophagy gradually subsides, manifested by the reduced formation of autophagic vacuoles and improper fusion of these vacuoles with the lysosomes. Similarly, in neurodegenerative disorders, accumulation of tau and synuclein proteins has been attributed to the decline in the autophagic removal of proteins. Equivalently, lysosomal disorders show an impairment of the autophagic process leading to the accumulation of lipid molecules within lysosomes. On the other hand, activation of the autophagic pathway has also proved beneficial in evading various foreign pathogens, thereby contributing to the innate immunity. In the context of cancer, autophagy has shown to play a puzzling role where it serves as a tumor suppressor during initial stages but later protects the tumor cells from the immune system defense mechanisms. Similarly, muscular and heart disorders have been shown to be positively and negatively regulated by autophagy, respectively. In the present review, we, therefore, present a comprehensive review on the role of autophagy in various diseases and their corresponding outcomes.
Collapse
Affiliation(s)
- Sarbari Saha
- Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Debasna P Panigrahi
- Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Shankargouda Patil
- Department of Maxillofacial Surgery and Diagnostic Sciences, Division of Oral Pathology, College of Dentistry, Jazan University, Saudi Arabia
| | - Sujit K Bhutia
- Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India.
| |
Collapse
|
49
|
Zhang C, Quan R, Wang J. Development and application of CRISPR/Cas9 technologies in genomic editing. Hum Mol Genet 2018; 27:R79-R88. [DOI: 10.1093/hmg/ddy120] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 04/03/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- Cui Zhang
- Institute of Cell and Development Biology, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Renfu Quan
- Institute of Orthopedics, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, China
| | - Jinfu Wang
- Institute of Cell and Development Biology, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
50
|
Yoshida S, Nakanishi C, Okada H, Mori M, Yokawa J, Yoshimuta T, Ohta K, Konno T, Fujino N, Kawashiri MA, Yachie A, Yamagishi M, Hayashi K. Characteristics of induced pluripotent stem cells from clinically divergent female monozygotic twins with Danon disease. J Mol Cell Cardiol 2018; 114:234-242. [DOI: 10.1016/j.yjmcc.2017.11.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/04/2017] [Accepted: 11/22/2017] [Indexed: 12/20/2022]
|