1
|
Huang W, Zhou R, Jiang C, Wang J, Zhou Y, Xu X, Wang T, Li A, Zhang Y. Mitochondrial dysfunction is associated with hypertrophic cardiomyopathy in Pompe disease-specific induced pluripotent stem cell-derived cardiomyocytes. Cell Prolif 2024; 57:e13573. [PMID: 37916452 PMCID: PMC10984102 DOI: 10.1111/cpr.13573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023] Open
Abstract
Pompe disease (PD) is a rare autosomal recessive disorder that presents with progressive hypertrophic cardiomyopathy. However, the detailed mechanism remains clarified. Herein, PD patient-specific induced pluripotent stem cells were differentiated into cardiomyocytes (PD-iCMs) that exhibited cardiomyopathic features of PD, including decreased acid alpha-glucosidase activity, lysosomal glycogen accumulation and hypertrophy. The defective mitochondria were involved in the cardiac pathology as shown by the significantly decreased number of mitochondria and impaired respiratory function and ATP production in PD-iCMs, which was partially due to elevated levels of intracellular reactive oxygen species produced from depolarized mitochondria. Further analysis showed that impaired fusion and autophagy of mitochondria and declined expression of mitochondrial complexes underlies the mechanism of dysfunctional mitochondria. This was alleviated by supplementation with recombinant human acid alpha-glucosidase that improved the mitochondrial function and concomitantly mitigated the cardiac pathology. Therefore, this study suggests that defective mitochondria underlie the pathogenesis of cardiomyopathy in patients with PD.
Collapse
Affiliation(s)
- Wenjun Huang
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and DiseasesShaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Rui Zhou
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and DiseasesShaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Congshan Jiang
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and DiseasesShaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Jie Wang
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and DiseasesShaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yafei Zhou
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and DiseasesShaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Xiaoyan Xu
- Department of CardiologyXi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Tao Wang
- Department of CardiologyXi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Anmao Li
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and DiseasesShaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yanmin Zhang
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and DiseasesShaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong UniversityXi'anChina
- Department of CardiologyXi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
2
|
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
|
3
|
Tian R, Yuan L, Huang Y, Zhang R, Lyu H, Xiao S, Guo D, Ali DW, Michalak M, Chen XZ, Zhou C, Tang J. Perturbed autophagy intervenes systemic lupus erythematosus by active ingredients of traditional Chinese medicine. Front Pharmacol 2023; 13:1053602. [PMID: 36733375 PMCID: PMC9887156 DOI: 10.3389/fphar.2022.1053602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/05/2022] [Indexed: 01/19/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a common multisystem, multiorgan heterozygous autoimmune disease. The main pathological features of the disease are autoantibody production and immune complex deposition. Autophagy is an important mechanism to maintain cell homeostasis. Autophagy functional abnormalities lead to the accumulation of apoptosis and induce the autoantibodies that result in immune disorders. Therefore, improving autophagy may alleviate the development of SLE. For SLE, glucocorticoids or immunosuppressive agents are commonly used in clinical treatment, but long-term use of these drugs causes serious side effects in humans. Immunosuppressive agents are expensive. Traditional Chinese medicines (TCMs) are widely used for immune diseases due to their low toxicity and few side effects. Many recent studies found that TCM and its active ingredients affected the pathological development of SLE by regulating autophagy. This article explains how autophagy interferes with immune system homeostasis and participates in the occurrence and development of SLE. It also summarizes several studies on TCM-regulated autophagy intervention in SLE to generate new ideas for basic research, the development of novel medications, and the clinical treatment of SLE.
Collapse
Affiliation(s)
- Rui Tian
- National “111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- College of Biological Science and Technology, Hubei MinZu University, Enshi, China
| | - Lin Yuan
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Enshi, China
| | - Yuan Huang
- National “111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Rui Zhang
- National “111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Hao Lyu
- National “111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Shuai Xiao
- National “111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Dong Guo
- National “111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Declan William Ali
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Cefan Zhou
- National “111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Jingfeng Tang
- National “111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
- Lead Contact, Wuhan, China
| |
Collapse
|
4
|
Huang W, Zhang Y, Zhou R. Induced pluripotent stem cell for modeling Pompe disease. Front Cardiovasc Med 2022; 9:1061384. [PMID: 36620633 PMCID: PMC9815144 DOI: 10.3389/fcvm.2022.1061384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
Pompe disease (PD) is a rare, autosomal recessive, inherited, and progressive metabolic disorder caused by α-glucosidase defect in lysosomes, resulting in abnormal glycogen accumulation. Patients with PD characteristically have multisystem pathological disorders, particularly hypertrophic cardiomyopathy, muscle weakness, and hepatomegaly. Although the pathogenesis and clinical outcomes of PD are well-established, disease-modeling ability, mechanism elucidation, and drug development targeting PD have been substantially limited by the unavailable PD-relevant cell models. This obstacle has been overcome with the help of induced pluripotent stem cell (iPSC) reprogramming technology, thus providing a powerful tool for cell replacement therapy, disease modeling, drug screening, and drug toxicity assessment. This review focused on the exciting achievement of PD disease modeling and mechanism exploration using iPSC.
Collapse
Affiliation(s)
- Wenjun Huang
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yanmin Zhang
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, China,Department of Cardiology, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rui Zhou
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, China,*Correspondence: Rui Zhou ✉
| |
Collapse
|
5
|
Cell Autophagy in NASH and NASH-Related Hepatocellular Carcinoma. Int J Mol Sci 2022; 23:ijms23147734. [PMID: 35887082 PMCID: PMC9322157 DOI: 10.3390/ijms23147734] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 12/21/2022] Open
Abstract
Autophagy, a cellular self-digestion process, involves the degradation of targeted cell components such as damaged organelles, unfolded proteins, and intracellular pathogens by lysosomes. It is a major quality control system of the cell and plays an important role in cell differentiation, survival, development, and homeostasis. Alterations in the cell autophagic machinery have been implicated in several disease conditions, including neurodegeneration, autoimmunity, cancer, infection, inflammatory diseases, and aging. In non-alcoholic fatty liver disease, including its inflammatory form, non-alcoholic steatohepatitis (NASH), a decrease in cell autophagic activity, has been implicated in the initial development and progression of steatosis to NASH and hepatocellular carcinoma (HCC). We present an overview of autophagy as it occurs in mammalian cells with an insight into the emerging understanding of the role of autophagy in NASH and NASH-related HCC.
Collapse
|
6
|
Aguilar-González A, González-Correa JE, Barriocanal-Casado E, Ramos-Hernández I, Lerma-Juárez MA, Greco S, Rodríguez-Sevilla JJ, Molina-Estévez FJ, Montalvo-Romeral V, Ronzitti G, Sánchez-Martín RM, Martín F, Muñoz P. Isogenic GAA-KO Murine Muscle Cell Lines Mimicking Severe Pompe Mutations as Preclinical Models for the Screening of Potential Gene Therapy Strategies. Int J Mol Sci 2022; 23:6298. [PMID: 35682977 PMCID: PMC9181599 DOI: 10.3390/ijms23116298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
Pompe disease (PD) is a rare disorder caused by mutations in the acid alpha-glucosidase (GAA) gene. Most gene therapies (GT) partially rely on the cross-correction of unmodified cells through the uptake of the GAA enzyme secreted by corrected cells. In the present study, we generated isogenic murine GAA-KO cell lines resembling severe mutations from Pompe patients. All of the generated GAA-KO cells lacked GAA activity and presented an increased autophagy and increased glycogen content by means of myotube differentiation as well as the downregulation of mannose 6-phosphate receptors (CI-MPRs), validating them as models for PD. Additionally, different chimeric murine GAA proteins (IFG, IFLG and 2G) were designed with the aim to improve their therapeutic activity. Phenotypic rescue analyses using lentiviral vectors point to IFG chimera as the best candidate in restoring GAA activity, normalising the autophagic marker p62 and surface levels of CI-MPRs. Interestingly, in vivo administration of liver-directed AAVs expressing the chimeras further confirmed the good behaviour of IFG, achieving cross-correction in heart tissue. In summary, we generated different isogenic murine muscle cell lines mimicking the severe PD phenotype, as well as validating their applicability as preclinical models in order to reduce animal experimentation.
Collapse
Affiliation(s)
- Araceli Aguilar-González
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry Applied to Biomedicine and the Environment”, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Juan Elías González-Correa
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Eliana Barriocanal-Casado
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Iris Ramos-Hernández
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Miguel A. Lerma-Juárez
- Instituto de Investigación del Hospital Universitario La Paz, IdiPAZ, 28029 Madrid, Spain;
| | - Sara Greco
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Juan José Rodríguez-Sevilla
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Francisco Javier Molina-Estévez
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Fundación para la Investigación Biosanitaria de Andalucía Oriental-Alejandro Otero (FIBAO), 18012 Granada, Spain
| | - Valle Montalvo-Romeral
- Généthon, Integrare Research Unit UMR_S951, INSERM, Université Paris-Saclay, Univ Evry, 91002 Evry, France; (V.M.-R.); (G.R.)
| | - Giuseppe Ronzitti
- Généthon, Integrare Research Unit UMR_S951, INSERM, Université Paris-Saclay, Univ Evry, 91002 Evry, France; (V.M.-R.); (G.R.)
| | - Rosario María Sánchez-Martín
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry Applied to Biomedicine and the Environment”, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Francisco Martín
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Departamento de Bioquímica y Biología Molecular 3 e Inmunología, Facultad de Medicina, Universidad de Granada, Avda. de la Investigación 11, 18071 Granada, Spain
| | - Pilar Muñoz
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Departmento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Campus Fuentenueva, 18071 Granada, Spain
| |
Collapse
|
7
|
Wang J, Zhou CJ, Khodabukus A, Tran S, Han SO, Carlson AL, Madden L, Kishnani PS, Koeberl DD, Bursac N. Three-dimensional tissue-engineered human skeletal muscle model of Pompe disease. Commun Biol 2021; 4:524. [PMID: 33953320 PMCID: PMC8100136 DOI: 10.1038/s42003-021-02059-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 03/31/2021] [Indexed: 01/24/2023] Open
Abstract
In Pompe disease, the deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA) causes skeletal and cardiac muscle weakness, respiratory failure, and premature death. While enzyme replacement therapy using recombinant human GAA (rhGAA) can significantly improve patient outcomes, detailed disease mechanisms and incomplete therapeutic effects require further studies. Here we report a three-dimensional primary human skeletal muscle ("myobundle") model of infantile-onset Pompe disease (IOPD) that recapitulates hallmark pathological features including reduced GAA enzyme activity, elevated glycogen content and lysosome abundance, and increased sensitivity of muscle contractile function to metabolic stress. In vitro treatment of IOPD myobundles with rhGAA or adeno-associated virus (AAV)-mediated hGAA expression yields increased GAA activity and robust glycogen clearance, but no improvements in stress-induced functional deficits. We also apply RNA sequencing analysis to the quadriceps of untreated and AAV-treated GAA-/- mice and wild-type controls to establish a Pompe disease-specific transcriptional signature and reveal novel disease pathways. The mouse-derived signature is enriched in the transcriptomic profile of IOPD vs. healthy myobundles and partially reversed by in vitro rhGAA treatment, further confirming the utility of the human myobundle model for studies of Pompe disease and therapy.
Collapse
Affiliation(s)
- Jason Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Chris J Zhou
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Sabrina Tran
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Sang-Oh Han
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Aaron L Carlson
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Lauran Madden
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Dwight D Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
8
|
Abstract
Lysosomes are membrane-bound organelles with roles in processes involved in degrading and recycling cellular waste, cellular signalling and energy metabolism. Defects in genes encoding lysosomal proteins cause lysosomal storage disorders, in which enzyme replacement therapy has proved successful. Growing evidence also implicates roles for lysosomal dysfunction in more common diseases including inflammatory and autoimmune disorders, neurodegenerative diseases, cancer and metabolic disorders. With a focus on lysosomal dysfunction in autoimmune disorders and neurodegenerative diseases - including lupus, rheumatoid arthritis, multiple sclerosis, Alzheimer disease and Parkinson disease - this Review critically analyses progress and opportunities for therapeutically targeting lysosomal proteins and processes, particularly with small molecules and peptide drugs.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- CNRS-University of Strasbourg, Biotechnology and Cell Signalling, Illkirch, France
- Laboratory of Excellence Medalis, Team Neuroimmunology and Peptide Therapy, Institut de Science et d'Ingénierie Supramoléculaire (ISIS), Strasbourg, France
| | - Fengjuan Wang
- CNRS-University of Strasbourg, Biotechnology and Cell Signalling, Illkirch, France
- Laboratory of Excellence Medalis, Team Neuroimmunology and Peptide Therapy, Institut de Science et d'Ingénierie Supramoléculaire (ISIS), Strasbourg, France
| | - Sylviane Muller
- CNRS-University of Strasbourg, Biotechnology and Cell Signalling, Illkirch, France.
- Laboratory of Excellence Medalis, Team Neuroimmunology and Peptide Therapy, Institut de Science et d'Ingénierie Supramoléculaire (ISIS), Strasbourg, France.
- University of Strasbourg Institute for Advanced Study, Strasbourg, France.
- Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg University, Strasbourg, France.
| |
Collapse
|
9
|
Palhegyi AM, Seranova E, Dimova S, Hoque S, Sarkar S. Biomedical Implications of Autophagy in Macromolecule Storage Disorders. Front Cell Dev Biol 2019; 7:179. [PMID: 31555645 PMCID: PMC6742707 DOI: 10.3389/fcell.2019.00179] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/19/2019] [Indexed: 12/20/2022] Open
Abstract
An imbalance between the production and clearance of macromolecules such as proteins, lipids and carbohydrates can lead to a category of diseases broadly known as macromolecule storage disorders. These include, but not limited to, neurodegenerative diseases such as Alzheimer’s, Parkinson’s and Huntington’s disease associated with accumulation of aggregation-prone proteins, Lafora and Pompe disease associated with glycogen accumulation, whilst lipid accumulation is characteristic to Niemann-Pick disease and Gaucher disease. One of the underlying factors contributing to the build-up of macromolecules in these storage disorders is the intracellular degradation pathway called autophagy. This process is the primary clearance route for unwanted macromolecules, either via bulk non-selective degradation, or selectively via aggrephagy, glycophagy and lipophagy. Since autophagy plays a vital role in maintaining cellular homeostasis, cell viability and human health, malfunction of this process could be detrimental. Indeed, defective autophagy has been reported in a number of macromolecule storage disorders where autophagy is impaired at distinct stages, such as at the level of autophagosome formation, autophagosome maturation or improper lysosomal degradation of the autophagic cargo. Of biomedical relevance, autophagy is regulated by multiple signaling pathways that are amenable to chemical perturbations by small molecules. Induction of autophagy has been shown to improve cell viability and exert beneficial effects in experimental models of various macromolecule storage disorders where the lysosomal functionality is not overtly compromised. In this review, we will discuss the role of autophagy in certain macromolecule storage disorders and highlight the potential therapeutic benefits of autophagy enhancers in these pathological conditions.
Collapse
Affiliation(s)
- Adina Maria Palhegyi
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom
| | - Elena Seranova
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom
| | - Simona Dimova
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom
| | - Sheabul Hoque
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom
| | - Sovan Sarkar
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
10
|
Jiwlawat N, Lynch EM, Napiwocki BN, Stempien A, Ashton RS, Kamp TJ, Crone WC, Suzuki M. Micropatterned substrates with physiological stiffness promote cell maturation and Pompe disease phenotype in human induced pluripotent stem cell-derived skeletal myocytes. Biotechnol Bioeng 2019; 116:2377-2392. [PMID: 31131875 DOI: 10.1002/bit.27075] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/19/2019] [Accepted: 05/21/2019] [Indexed: 12/23/2022]
Abstract
Recent advances in bioengineering have enabled cell culture systems that more closely mimic the native cellular environment. Here, we demonstrated that human induced pluripotent stem cell (iPSC)-derived myogenic progenitors formed highly-aligned myotubes and contracted when seeded on two-dimensional micropatterned platforms. The differentiated cells showed clear nuclear alignment and formed elongated myotubes dependent on the width of the micropatterned lanes. Topographical cues from micropatterning and physiological substrate stiffness improved the formation of well-aligned and multinucleated myotubes similar to myofibers. These aligned myotubes exhibited spontaneous contractions specifically along the long axis of the pattern. Notably, the micropatterned platforms developed bundle-like myotubes using patient-derived iPSCs with a background of Pompe disease (glycogen storage disease type II) and even enhanced the disease phenotype as shown through the specific pathology of abnormal lysosome accumulations. A highly-aligned formation of matured myotubes holds great potential in further understanding the process of human muscle development, as well as advancing in vitro pharmacological studies for skeletal muscle diseases.
Collapse
Affiliation(s)
- Nunnapas Jiwlawat
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| | - Eileen M Lynch
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| | - Brett N Napiwocki
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin
| | - Alana Stempien
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin
| | - Randolph S Ashton
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin.,The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, Wisconsin
| | - Timothy J Kamp
- Department of Medicine, University of Wisconsin, Madison, Wisconsin.,The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, Wisconsin.,Department of Cell and Regenerative Biology, University of Wisconsin, Madison, Wisconsin
| | - Wendy C Crone
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin.,The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, Wisconsin.,Department of Engineering Physics, University of Wisconsin, Madison, Wisconsin
| | - Masatoshi Suzuki
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin.,The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
11
|
Moreno ML, Mérida S, Bosch-Morell F, Miranda M, Villar VM. Autophagy Dysfunction and Oxidative Stress, Two Related Mechanisms Implicated in Retinitis Pigmentosa. Front Physiol 2018; 9:1008. [PMID: 30093867 PMCID: PMC6070619 DOI: 10.3389/fphys.2018.01008] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 07/09/2018] [Indexed: 12/12/2022] Open
Abstract
Retinitis pigmentosa (RP) is one of the most common clinical subtypes of retinal degeneration (RD), and it is a neurodegenerative disease that could cause complete blindness in humans because it ultimately affects the photoreceptors viability. RP afflicts an estimated 1.5 million patients worldwide. The retina is highly susceptible to oxidative stress which can impair mitochondrial function. Many retina pathologies, such as diabetic retinopathy and secondary cone photoreceptor death in RP, have been related directly or indirectly with mitochondrial dysfunction. The possible role of autophagy in retina and cell differentiation is described and also the implications of autophagy dysregulation in RP. The present review shows the crucial role of autophagy in maintaining the retina homeostasis and possible therapeutic approaches for the treatment of RP.
Collapse
Affiliation(s)
- Mari-Luz Moreno
- Department of Basic Sciences, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Salvador Mérida
- Departamento de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Francisco Bosch-Morell
- Departamento de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain.,Department of Medical Ophtalmology, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana, Valencia, Spain
| | - María Miranda
- Departamento de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Vincent M Villar
- Departamento de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| |
Collapse
|
12
|
Castets P, Frank S, Sinnreich M, Rüegg MA. "Get the Balance Right": Pathological Significance of Autophagy Perturbation in Neuromuscular Disorders. J Neuromuscul Dis 2018; 3:127-155. [PMID: 27854220 PMCID: PMC5271579 DOI: 10.3233/jnd-160153] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent research has revealed that autophagy, a major catabolic process in cells, is dysregulated in several neuromuscular diseases and contributes to the muscle wasting caused by non-muscle disorders (e.g. cancer cachexia) or during aging (i.e. sarcopenia). From there, the idea arose to interfere with autophagy or manipulate its regulatory signalling to help restore muscle homeostasis and attenuate disease progression. The major difficulty for the development of therapeutic strategies is to restore a balanced autophagic flux, due to the dynamic nature of autophagy. Thus, it is essential to better understand the mechanisms and identify the signalling pathways at play in the control of autophagy in skeletal muscle. A comprehensive analysis of the autophagic flux and of the causes of its dysregulation is required to assess the pathogenic role of autophagy in diseased muscle. Furthermore, it is essential that experiments distinguish between primary dysregulation of autophagy (prior to disease onset) and impairments as a consequence of the pathology. Of note, in most muscle disorders, autophagy perturbation is not caused by genetic modification of an autophagy-related protein, but rather through indirect alteration of regulatory signalling or lysosomal function. In this review, we will present the mechanisms involved in autophagy, and those ensuring its tight regulation in skeletal muscle. We will then discuss as to how autophagy dysregulation contributes to the pathogenesis of neuromuscular disorders and possible ways to interfere with this process to limit disease progression.
Collapse
Affiliation(s)
| | - Stephan Frank
- Institute of Pathology, Division of Neuropathology Basel University Hospital, Basel, Switzerland
| | - Michael Sinnreich
- Neuromuscular Research Center, Departments of Neurology and Biomedicine, Pharmazentrum, Basel, Switzerland
| | | |
Collapse
|
13
|
A Skeletal Muscle Model of Infantile-onset Pompe Disease with Patient-specific iPS Cells. Sci Rep 2017; 7:13473. [PMID: 29044175 PMCID: PMC5647434 DOI: 10.1038/s41598-017-14063-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 10/05/2017] [Indexed: 12/22/2022] Open
Abstract
Pompe disease is caused by an inborn defect of lysosomal acid α-glucosidase (GAA) and is characterized by lysosomal glycogen accumulation primarily in the skeletal muscle and heart. Patients with the severe type of the disease, infantile-onset Pompe disease (IOPD), show generalized muscle weakness and heart failure in early infancy. They cannot survive over two years. Enzyme replacement therapy with recombinant human GAA (rhGAA) improves the survival rate, but its effect on skeletal muscle is insufficient compared to other organs. Moreover, the patho-mechanism of skeletal muscle damage in IOPD is still unclear. Here we generated induced pluripotent stem cells (iPSCs) from patients with IOPD and differentiated them into myocytes. Differentiated myocytes showed lysosomal glycogen accumulation, which was dose-dependently rescued by rhGAA. We further demonstrated that mammalian/mechanistic target of rapamycin complex 1 (mTORC1) activity was impaired in IOPD iPSC-derived myocytes. Comprehensive metabolomic and transcriptomic analyses suggested the disturbance of mTORC1-related signaling, including deteriorated energy status and suppressed mitochondrial oxidative function. In summary, we successfully established an in vitro skeletal muscle model of IOPD using patient-specific iPSCs. Disturbed mTORC1 signaling may contribute to the pathogenesis of skeletal muscle damage in IOPD, and may be a potential therapeutic target for Pompe disease.
Collapse
|
14
|
Lim JA, Li L, Shirihai OS, Trudeau KM, Puertollano R, Raben N. Modulation of mTOR signaling as a strategy for the treatment of Pompe disease. EMBO Mol Med 2017; 9:353-370. [PMID: 28130275 PMCID: PMC5331267 DOI: 10.15252/emmm.201606547] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) coordinates biosynthetic and catabolic processes in response to multiple extracellular and intracellular signals including growth factors and nutrients. This serine/threonine kinase has long been known as a critical regulator of muscle mass. The recent finding that the decision regarding its activation/inactivation takes place at the lysosome undeniably brings mTOR into the field of lysosomal storage diseases. In this study, we have examined the involvement of the mTOR pathway in the pathophysiology of a severe muscle wasting condition, Pompe disease, caused by excessive accumulation of lysosomal glycogen. Here, we report the dysregulation of mTOR signaling in the diseased muscle cells, and we focus on potential sites for therapeutic intervention. Reactivation of mTOR in the whole muscle of Pompe mice by TSC knockdown resulted in the reversal of atrophy and a striking removal of autophagic buildup. Of particular interest, we found that the aberrant mTOR signaling can be reversed by arginine. This finding can be translated into the clinic and may become a paradigm for targeted therapy in lysosomal, metabolic, and neuromuscular diseases.
Collapse
Affiliation(s)
- Jeong-A Lim
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.,Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lishu Li
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Orian S Shirihai
- Department of Medicine, Obesity and Nutrition Section, Evans Biomedical Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Kyle M Trudeau
- Department of Medicine, Obesity and Nutrition Section, Evans Biomedical Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Rosa Puertollano
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nina Raben
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
15
|
Kong XY, Feng YZ, Eftestøl E, Kase ET, Haugum H, Eskild W, Rustan AC, Thoresen GH. Increased glucose utilization and decreased fatty acid metabolism in myotubes from Glmp(gt/gt) mice. Arch Physiol Biochem 2016; 122:36-45. [PMID: 26707125 DOI: 10.3109/13813455.2015.1120752] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Glycosylated lysosomal membrane protein (GLMP) has been reported to enhance the expression from a peroxisome proliferator-activated receptor alpha (PPARα) responsive promoter, but also to be an integral lysosomal membrane protein. Using myotubes established from wild-type and Glmp(gt/gt) mice, the importance of GLMP in skeletal muscle was examined. Glmp(gt/gt) myotubes expressed a more glycolytic phenotype than wild-type myotubes. Myotubes from Glmp(gt/gt) mice metabolized glucose faster and had a larger pool of intracellular glycogen, while oleic acid uptake, storage and oxidation were significantly reduced. Gene expression analyses indicated lower expression of three PPAR-isoforms, a co-regulator of PPAR (PGC1α) and several genes important for lipid metabolism in Glmp(gt/gt) myotubes. However, ablation of GLMP did not seem to substantially impair the response to PPAR agonists. In conclusion, myotubes established from Glmp(gt/gt) mice were more glycolytic than myotubes from wild-type animals, in spite of no differences in muscle fiber types in vivo.
Collapse
Affiliation(s)
| | - Yuan Zeng Feng
- b Department of Pharmaceutical Biosciences , School of Pharmacy, University of Oslo , Oslo , Norway , and
| | | | - Eili T Kase
- b Department of Pharmaceutical Biosciences , School of Pharmacy, University of Oslo , Oslo , Norway , and
| | - Hanne Haugum
- b Department of Pharmaceutical Biosciences , School of Pharmacy, University of Oslo , Oslo , Norway , and
| | | | - Arild C Rustan
- b Department of Pharmaceutical Biosciences , School of Pharmacy, University of Oslo , Oslo , Norway , and
| | - G Hege Thoresen
- b Department of Pharmaceutical Biosciences , School of Pharmacy, University of Oslo , Oslo , Norway , and
- c Department of Pharmacology , Institute of Clinical Medicine, University of Oslo and Oslo University Hospital , Oslo , Norway
| |
Collapse
|
16
|
Abstract
Autophagy is an important physiological process in the heart, and alterations in autophagic activity can exacerbate or mitigate injury during various pathological processes. Methods to assess autophagy have changed rapidly because the field of research has expanded. As with any new field, methods and standards for data analysis and interpretation evolve as investigators acquire experience and insight. The purpose of this review is to summarize current methods to measure autophagy, selective mitochondrial autophagy (mitophagy), and autophagic flux. We will examine several published studies where confusion arose in data interpretation, to illustrate the challenges. Finally, we will discuss methods to assess autophagy in vivo and in patients.
Collapse
Affiliation(s)
- Roberta A Gottlieb
- From the Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center Cedars-Sinai Medical Center, Los Angeles, CA.
| | - Allen M Andres
- From the Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jon Sin
- From the Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center Cedars-Sinai Medical Center, Los Angeles, CA
| | - David P J Taylor
- From the Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
17
|
Lim JA, Li L, Kakhlon O, Myerowitz R, Raben N. Defects in calcium homeostasis and mitochondria can be reversed in Pompe disease. Autophagy 2015; 11:385-402. [PMID: 25758767 PMCID: PMC4502791 DOI: 10.1080/15548627.2015.1009779] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 09/08/2014] [Accepted: 11/28/2014] [Indexed: 02/08/2023] Open
Abstract
Mitochondria-induced oxidative stress and flawed autophagy are common features of neurodegenerative and lysosomal storage diseases (LSDs). Although defective autophagy is particularly prominent in Pompe disease, mitochondrial function has escaped examination in this typical LSD. We have found multiple mitochondrial defects in mouse and human models of Pompe disease, a life-threatening cardiac and skeletal muscle myopathy: a profound dysregulation of Ca(2+) homeostasis, mitochondrial Ca(2+) overload, an increase in reactive oxygen species, a decrease in mitochondrial membrane potential, an increase in caspase-independent apoptosis, as well as a decreased oxygen consumption and ATP production of mitochondria. In addition, gene expression studies revealed a striking upregulation of the β 1 subunit of L-type Ca(2+) channel in Pompe muscle cells. This study provides strong evidence that disturbance of Ca(2+) homeostasis and mitochondrial abnormalities in Pompe disease represent early changes in a complex pathogenetic cascade leading from a deficiency of a single lysosomal enzyme to severe and hard-to-treat autophagic myopathy. Remarkably, L-type Ca(2+)channel blockers, commonly used to treat other maladies, reversed these defects, indicating that a similar approach can be beneficial to the plethora of lysosomal and neurodegenerative disorders.
Collapse
Key Words
- AIFM1, apoptosis-inducing factor, mitochondrion-associated, 1
- CCCP, carbonyl cyanide m-chlorophenylhydrazone
- DMEM, Dulbecco's modified Eagle's medium
- EGTA, ethylene glycol-bis(2-aminoethylether)-N, N, N′, N′-tetraacetic acid
- ERT, enzyme replacement therapy
- GAA, glucosidase
- GFP, green fluorescent protein
- LAMP1, lysosomal-associated membrane protein 1
- LSD, lysosomal storage disease
- MAP1LC3A/B (LC3), microtubule-associated protein 1 light chain 3 α/β
- MOPS, 3-morpholinopropane-1-sulfonic acid
- MitoG, MitoTracker Green
- OMM, outer mitochondrial membrane
- Pompe disease
- RFP, red fluorescent protein
- ROS, reactive oxygen species
- TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling
- Ub, ubiquitinated
- VDCC, voltage-dependent Ca2+ channel
- autophagy
- calcium
- lysosome
- mitochondria
- mitophagy
- α, acid
Collapse
Affiliation(s)
- Jeong-A Lim
- Laboratory of Muscle Stem Cells and Gene Regulation; National Institute of Arthritis and Musculoskeletal and Skin Diseases; National Institutes of Health; Bethesda; MD USA
| | - Lishu Li
- Laboratory of Muscle Stem Cells and Gene Regulation; National Institute of Arthritis and Musculoskeletal and Skin Diseases; National Institutes of Health; Bethesda; MD USA
| | - Or Kakhlon
- Department of Neurology; Hadassah-Hebrew University Medical Center; Ein Kerem; Jerusalem, Israel
| | - Rachel Myerowitz
- Laboratory of Muscle Stem Cells and Gene Regulation; National Institute of Arthritis and Musculoskeletal and Skin Diseases; National Institutes of Health; Bethesda; MD USA
- St. Mary's College of Maryland; St. Mary's City, MD USA
| | - Nina Raben
- Laboratory of Muscle Stem Cells and Gene Regulation; National Institute of Arthritis and Musculoskeletal and Skin Diseases; National Institutes of Health; Bethesda; MD USA
| |
Collapse
|
18
|
Linton PJ, Gurney M, Sengstock D, Mentzer RM, Gottlieb RA. This old heart: Cardiac aging and autophagy. J Mol Cell Cardiol 2014; 83:44-54. [PMID: 25543002 DOI: 10.1016/j.yjmcc.2014.12.017] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/10/2014] [Accepted: 12/16/2014] [Indexed: 01/02/2023]
Abstract
Autophagy, a cellular housekeeping process, is essential to maintain tissue homeostasis, particularly in long-lived cells such as cardiomyocytes. Autophagic activity declines with age and may explain many features of age-related cardiac dysfunction. In this review we summarize the current state of knowledge regarding age-related changes in autophagy in the heart. Recent findings from studies in human hearts are presented, including evidence that the autophagic response is intact in the aged human heart. Impaired autophagic clearance of protein aggregates or deteriorating mitochondria will have multiple consequences including increased arrhythmia risk, decreased contractile function, reduced tolerance to ischemic stress, and increased inflammation; thus autophagy represents a potentially important therapeutic target to mitigate the cardiac consequences of aging. This article is part of a Special Issue entitled CV Aging.
Collapse
Affiliation(s)
- Phyllis-Jean Linton
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA, USA
| | - Michael Gurney
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA, USA
| | - David Sengstock
- Division of Geriatric Medicine, Oakwood Hospital, Dearborn, MI, USA; Department of Internal Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| | - Robert M Mentzer
- Cardiovascular Research Institute, Departments of Surgery and Physiology, Wayne State University School of Medicine, Detroit, MI, USA; Heart Institute and Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Roberta A Gottlieb
- Heart Institute and Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Raval KK, Tao R, White BE, De Lange WJ, Koonce CH, Yu J, Kishnani PS, Thomson JA, Mosher DF, Ralphe JC, Kamp TJ. Pompe disease results in a Golgi-based glycosylation deficit in human induced pluripotent stem cell-derived cardiomyocytes. J Biol Chem 2014; 290:3121-36. [PMID: 25488666 DOI: 10.1074/jbc.m114.628628] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Infantile-onset Pompe disease is an autosomal recessive disorder caused by the complete loss of lysosomal glycogen-hydrolyzing enzyme acid α-glucosidase (GAA) activity, which results in lysosomal glycogen accumulation and prominent cardiac and skeletal muscle pathology. The mechanism by which loss of GAA activity causes cardiomyopathy is poorly understood. We reprogrammed fibroblasts from patients with infantile-onset Pompe disease to generate induced pluripotent stem (iPS) cells that were differentiated to cardiomyocytes (iPSC-CM). Pompe iPSC-CMs had undetectable GAA activity and pathognomonic glycogen-filled lysosomes. Nonetheless, Pompe and control iPSC-CMs exhibited comparable contractile properties in engineered cardiac tissue. Impaired autophagy has been implicated in Pompe skeletal muscle; however, control and Pompe iPSC-CMs had comparable clearance rates of LC3-II-detected autophagosomes. Unexpectedly, the lysosome-associated membrane proteins, LAMP1 and LAMP2, from Pompe iPSC-CMs demonstrated higher electrophoretic mobility compared with control iPSC-CMs. Brefeldin A induced disruption of the Golgi in control iPSC-CMs reproduced the higher mobility forms of the LAMPs, suggesting that Pompe iPSC-CMs produce LAMPs lacking appropriate glycosylation. Isoelectric focusing studies revealed that LAMP2 has a more alkaline pI in Pompe compared with control iPSC-CMs due largely to hyposialylation. MALDI-TOF-MS analysis of N-linked glycans demonstrated reduced diversity of multiantennary structures and the major presence of a trimannose complex glycan precursor in Pompe iPSC-CMs. These data suggest that Pompe cardiomyopathy has a glycan processing abnormality and thus shares features with hypertrophic cardiomyopathies observed in the congenital disorders of glycosylation.
Collapse
Affiliation(s)
- Kunil K Raval
- From the Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53705, the WiCell Institute, Madison, Wisconsin 53719
| | - Ran Tao
- From the Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53705
| | - Brent E White
- From the Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53705
| | - Willem J De Lange
- the Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792
| | - Chad H Koonce
- From the Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53705
| | - Junying Yu
- Cellular Dynamics International, Madison, Wisconsin 53711
| | - Priya S Kishnani
- the Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina 27710
| | - James A Thomson
- the Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53706, the Genome Center of Wisconsin, University of Wisconsin, Madison, Wisconsin 53706, the Morgridge Institute for Research, Madison, Wisconsin 53715
| | - Deane F Mosher
- From the Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53705, the Department of Biomolecular Chemistry, University of Wisconsin, Madison, Wisconsin 53706, and
| | - John C Ralphe
- the Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792
| | - Timothy J Kamp
- From the Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53705, the Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53706, the WiCell Institute, Madison, Wisconsin 53719,
| |
Collapse
|
20
|
Moyzis AG, Sadoshima J, Gustafsson ÅB. Mending a broken heart: the role of mitophagy in cardioprotection. Am J Physiol Heart Circ Physiol 2014; 308:H183-92. [PMID: 25437922 DOI: 10.1152/ajpheart.00708.2014] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The heart is highly energy dependent with most of its energy provided by mitochondrial oxidative phosphorylation. Mitochondria also play a role in many other essential cellular processes including metabolite synthesis and calcium storage. Therefore, maintaining a functional population of mitochondria is critical for cardiac function. Efficient degradation and replacement of dysfunctional mitochondria ensures cell survival, particularly in terminally differentiated cells such as cardiac myocytes. Mitochondria are eliminated via mitochondrial autophagy or mitophagy. In the heart, mitophagy is an essential housekeeping process and required for cardiac homeostasis. Reduced autophagy and accumulation of impaired mitochondria have been linked to progression of heart failure and aging. In this review, we discuss the pathways that regulate mitophagy in cells and highlight the cardioprotective role of mitophagy in response to stress and aging. We also discuss the therapeutic potential of targeting mitophagy and directions for future investigation.
Collapse
Affiliation(s)
- Alexandra G Moyzis
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California; and
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, New Jersey
| | - Åsa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California; and
| |
Collapse
|
21
|
Shemesh A, Wang Y, Yang Y, Yang GS, Johnson DE, Backer JM, Pessin JE, Zong H. Suppression of mTORC1 activation in acid-α-glucosidase-deficient cells and mice is ameliorated by leucine supplementation. Am J Physiol Regul Integr Comp Physiol 2014; 307:R1251-9. [PMID: 25231351 DOI: 10.1152/ajpregu.00212.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pompe disease is due to a deficiency in acid-α-glucosidase (GAA) and results in debilitating skeletal muscle wasting, characterized by the accumulation of glycogen and autophagic vesicles. Given the role of lysosomes as a platform for mTORC1 activation, we examined mTORC1 activity in models of Pompe disease. GAA-knockdown C2C12 myoblasts and GAA-deficient human skin fibroblasts of infantile Pompe patients were found to have decreased mTORC1 activation. Treatment with the cell-permeable leucine analog L-leucyl-L-leucine methyl ester restored mTORC1 activation. In vivo, Pompe mice also displayed reduced basal and leucine-stimulated mTORC1 activation in skeletal muscle, whereas treatment with a combination of insulin and leucine normalized mTORC1 activation. Chronic leucine feeding restored basal and leucine-stimulated mTORC1 activation, while partially protecting Pompe mice from developing kyphosis and the decline in muscle mass. Leucine-treated Pompe mice showed increased spontaneous activity and running capacity, with reduced muscle protein breakdown and glycogen accumulation. Together, these data demonstrate that GAA deficiency results in reduced mTORC1 activation that is partly responsible for the skeletal muscle wasting phenotype. Moreover, mTORC1 stimulation by dietary leucine supplementation prevented some of the detrimental skeletal muscle dysfunction that occurs in the Pompe disease mouse model.
Collapse
Affiliation(s)
- Adi Shemesh
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Yichen Wang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Yingjuan Yang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Gong-She Yang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Danielle E Johnson
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada; and
| | - Jonathan M Backer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Jeffrey E Pessin
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Haihong Zong
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
22
|
Khanna R, Powe AC, Lun Y, Soska R, Feng J, Dhulipala R, Frascella M, Garcia A, Pellegrino LJ, Xu S, Brignol N, Toth MJ, Do HV, Lockhart DJ, Wustman BA, Valenzano KJ. The pharmacological chaperone AT2220 increases the specific activity and lysosomal delivery of mutant acid alpha-glucosidase, and promotes glycogen reduction in a transgenic mouse model of Pompe disease. PLoS One 2014; 9:e102092. [PMID: 25036864 PMCID: PMC4103853 DOI: 10.1371/journal.pone.0102092] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 06/14/2014] [Indexed: 11/18/2022] Open
Abstract
Pompe disease is an inherited lysosomal storage disorder that results from a deficiency in acid α-glucosidase (GAA) activity due to mutations in the GAA gene. Pompe disease is characterized by accumulation of lysosomal glycogen primarily in heart and skeletal muscles, which leads to progressive muscle weakness. We have shown previously that the small molecule pharmacological chaperone AT2220 (1-deoxynojirimycin hydrochloride, duvoglustat hydrochloride) binds and stabilizes wild-type as well as multiple mutant forms of GAA, and can lead to higher cellular levels of GAA. In this study, we examined the effect of AT2220 on mutant GAA, in vitro and in vivo, with a primary focus on the endoplasmic reticulum (ER)-retained P545L mutant form of human GAA (P545L GAA). AT2220 increased the specific activity of P545L GAA toward both natural (glycogen) and artificial substrates in vitro. Incubation with AT2220 also increased the ER export, lysosomal delivery, proteolytic processing, and stability of P545L GAA. In a new transgenic mouse model of Pompe disease that expresses human P545L on a Gaa knockout background (Tg/KO) and is characterized by reduced GAA activity and elevated glycogen levels in disease-relevant tissues, daily oral administration of AT2220 for 4 weeks resulted in significant and dose-dependent increases in mature lysosomal GAA isoforms and GAA activity in heart and skeletal muscles. Importantly, oral administration of AT2220 also resulted in significant glycogen reduction in disease-relevant tissues. Compared to daily administration, less-frequent AT2220 administration, including repeated cycles of 4 or 5 days with AT2220 followed by 3 or 2 days without drug, respectively, resulted in even greater glycogen reductions. Collectively, these data indicate that AT2220 increases the specific activity, trafficking, and lysosomal stability of P545L GAA, leads to increased levels of mature GAA in lysosomes, and promotes glycogen reduction in situ. As such, AT2220 may warrant further evaluation as a treatment for Pompe disease.
Collapse
Affiliation(s)
- Richie Khanna
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | - Allan C. Powe
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | - Yi Lun
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | - Rebecca Soska
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | - Jessie Feng
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | - Rohini Dhulipala
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | - Michelle Frascella
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | - Anadina Garcia
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | - Lee J. Pellegrino
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | - Su Xu
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | - Nastry Brignol
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | - Matthew J. Toth
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | - Hung V. Do
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | - David J. Lockhart
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | - Brandon A. Wustman
- Amicus Therapeutics Inc., Cranbury, New Jersey, United States of America
| | | |
Collapse
|
23
|
Role of autophagy in glycogen breakdown and its relevance to chloroquine myopathy. PLoS Biol 2013; 11:e1001708. [PMID: 24265594 PMCID: PMC3825659 DOI: 10.1371/journal.pbio.1001708] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 10/04/2013] [Indexed: 01/19/2023] Open
Abstract
Several myopathies are associated with defects in autophagic and lysosomal degradation of glycogen, but it remains unclear how glycogen is targeted to the lysosome and what significance this process has for muscle cells. We have established a Drosophila melanogaster model to study glycogen autophagy in skeletal muscles, using chloroquine (CQ) to simulate a vacuolar myopathy that is completely dependent on the core autophagy genes. We show that autophagy is required for the most efficient degradation of glycogen in response to starvation. Furthermore, we show that CQ-induced myopathy can be improved by reduction of either autophagy or glycogen synthesis, the latter possibly due to a direct role of Glycogen Synthase in regulating autophagy through its interaction with Atg8.
Collapse
|
24
|
Spampanato C, Feeney E, Li L, Cardone M, Lim JA, Annunziata F, Zare H, Polishchuk R, Puertollano R, Parenti G, Ballabio A, Raben N. Transcription factor EB (TFEB) is a new therapeutic target for Pompe disease. EMBO Mol Med 2013; 5:691-706. [PMID: 23606558 PMCID: PMC3662313 DOI: 10.1002/emmm.201202176] [Citation(s) in RCA: 244] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 02/25/2013] [Accepted: 02/27/2013] [Indexed: 01/01/2023] Open
Abstract
A recently proposed therapeutic approach for lysosomal storage disorders (LSDs) relies upon the ability of transcription factor EB (TFEB) to stimulate autophagy and induce lysosomal exocytosis leading to cellular clearance. This approach is particularly attractive in glycogen storage disease type II [a severe metabolic myopathy, Pompe disease (PD)] as the currently available therapy, replacement of the missing enzyme acid alpha-glucosidase, fails to reverse skeletal muscle pathology. PD, a paradigm for LSDs, is characterized by both lysosomal abnormality and dysfunctional autophagy. Here, we show that TFEB is a viable therapeutic target in PD: overexpression of TFEB in a new muscle cell culture system and in mouse models of the disease reduced glycogen load and lysosomal size, improved autophagosome processing, and alleviated excessive accumulation of autophagic vacuoles. Unexpectedly, the exocytosed vesicles were labelled with lysosomal and autophagosomal membrane markers, suggesting that TFEB induces exocytosis of autophagolysosomes. Furthermore, the effects of TFEB were almost abrogated in the setting of genetically suppressed autophagy, supporting the role of autophagy in TFEB-mediated cellular clearance.
Collapse
|
25
|
|
26
|
Li HM, Feeney E, Li L, Zare H, Puertollano R, Raben N. WITHDRAWN: Clearance of lysosomal glycogen accumulation by Transcription factor EB (TFEB) in muscle cells from lysosomal alpha-glucosidase deficient mice. Biochem Biophys Res Commun 2013:S0006-291X(13)00272-6. [PMID: 23416076 PMCID: PMC3687018 DOI: 10.1016/j.bbrc.2013.02.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 02/04/2013] [Indexed: 11/28/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Hoi Ming Li
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Li WW, Li J, Bao JK. Microautophagy: lesser-known self-eating. Cell Mol Life Sci 2012; 69:1125-36. [PMID: 22080117 PMCID: PMC11114512 DOI: 10.1007/s00018-011-0865-5] [Citation(s) in RCA: 515] [Impact Index Per Article: 42.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 09/29/2011] [Accepted: 10/14/2011] [Indexed: 12/14/2022]
Abstract
Microautophagy, the non-selective lysosomal degradative process, involves direct engulfment of cytoplasmic cargo at a boundary membrane by autophagic tubes, which mediate both invagination and vesicle scission into the lumen. With its constitutive characteristics, microautophagy of soluble substrates can be induced by nitrogen starvation or rapamycin via regulatory signaling complex pathways. The maintenance of organellar size, membrane homeostasis, and cell survival under nitrogen restriction are the main functions of microautophagy. In addition, microautophagy is coordinated with and complements macroautophagy, chaperone-mediated autophagy, and other self-eating pathways. Three forms of selective microautophagy, including micropexophagy, piecemeal microautophagy of the nucleus, and micromitophagy, share common ground with microautophagy to some degree. As the accumulation of experimental data, the precise mechanisms that govern microautophagy are becoming more appreciated. Here, we review the microautophagic molecular machinery, its physiological functions, and relevance to human diseases, especially in diseases involving multivesicular bodies and multivesicular lysosomes.
Collapse
Affiliation(s)
- Wen-wen Li
- School of Life Sciences, Sichuan University, Chengdu, China
| | | | | |
Collapse
|
28
|
Chopra I, Li HF, Wang H, Webster KA. Phosphorylation of the insulin receptor by AMP-activated protein kinase (AMPK) promotes ligand-independent activation of the insulin signalling pathway in rodent muscle. Diabetologia 2012; 55:783-94. [PMID: 22207502 PMCID: PMC4648248 DOI: 10.1007/s00125-011-2407-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Accepted: 11/10/2011] [Indexed: 01/22/2023]
Abstract
AIMS/HYPOTHESIS Muscle may experience hypoglycaemia during ischaemia or insulin infusion. During severe hypoglycaemia energy production is blocked, and an increase of AMP:ATP activates the energy sensor and putative insulin-sensitiser AMP-activated protein kinase (AMPK). AMPK promotes energy conservation and survival by shutting down anabolism and activating catabolic pathways. We investigated the molecular mechanism of a unique glucose stress defence pathway involving AMPK-dependent, insulin-independent activation of the insulin signalling pathway. METHODS Cardiac or skeletal myocytes were subjected to glucose and insulin-free incubation for increasing intervals up to 20 h. AMPK, and components of the insulin signalling pathway and their targets were quantified by western blot using phosphor-specific antibodies. Phosphomimetics were used to determine the function of IRS-1 Ser789 phosphorylation and in vitro [³²P]ATP kinase assays were used to measure the phosphorylation of the purified insulin receptor by AMPK. RESULTS Glucose deprivation increased Akt-Thr308 and Akt-Ser473 phosphorylation by almost tenfold. Phosphorylation of glycogen synthase kinase 3 beta increased in parallel, but phosphorylation of ribosomal 70S subunit-S6 protein kinase and mammalian target of rapamycin decreased. AMPK inhibitors blocked and aminoimidazole carboxamide ribonucleotide (AICAR) mimicked the effects of glucose starvation. Glucose deprivation increased the phosphorylation of IRS-1 on serine-789, but phosphomimetics revealed that this conferred negative regulation. Glucose deprivation enhanced tyrosine phosphorylation of IRS-1 and the insulin receptor, effects that were blocked by AMPK inhibition and mimicked by AICAR. In vitro kinase assays using purified proteins confirmed that the insulin receptor is a direct target of AMPK. CONCLUSIONS/INTERPRETATION AMPK phosphorylates and activates the insulin receptor, providing a direct link between AMPK and the insulin signalling pathway; this pathway promotes energy conservation and survival of muscle exposed to severe glucose deprivation.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/antagonists & inhibitors
- AMP-Activated Protein Kinases/metabolism
- Animals
- Animals, Newborn
- Cells, Cultured
- Hep G2 Cells
- Humans
- Hypoglycemia/metabolism
- Hypoglycemic Agents/pharmacology
- Insulin Receptor Substrate Proteins/genetics
- Insulin Receptor Substrate Proteins/metabolism
- Ligands
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Mutant Proteins/metabolism
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Phosphorylation/drug effects
- Protein Kinase Inhibitors/pharmacology
- Protein Processing, Post-Translational/drug effects
- Rats
- Receptor, Insulin/isolation & purification
- Receptor, Insulin/metabolism
- Recombinant Proteins/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- I. Chopra
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, RMSB 6038, Miami, FL 33136, USA
| | - H. F. Li
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, RMSB 6038, Miami, FL 33136, USA
| | - H. Wang
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, RMSB 6038, Miami, FL 33136, USA
| | - K. A. Webster
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, RMSB 6038, Miami, FL 33136, USA
| |
Collapse
|
29
|
Huang HP, Chen PH, Hwu WL, Chuang CY, Chien YH, Stone L, Chien CL, Li LT, Chiang SC, Chen HF, Ho HN, Chen CH, Kuo HC. Human Pompe disease-induced pluripotent stem cells for pathogenesis modeling, drug testing and disease marker identification. Hum Mol Genet 2011; 20:4851-64. [PMID: 21926084 DOI: 10.1093/hmg/ddr424] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pompe disease is caused by autosomal recessive mutations in the acid alpha-glucosidase (GAA) gene, which encodes GAA. Although enzyme replacement therapy has recently improved patient survival greatly, the results in skeletal muscles and for advanced disease are still not satisfactory. Here, we report the derivation of Pompe disease-induced pluripotent stem cells (PomD-iPSCs) from two patients with different GAA mutations and their potential for pathogenesis modeling, drug testing and disease marker identification. PomD-iPSCs maintained pluripotent features and had low GAA activity and high glycogen content. Cardiomyocyte-like cells (CMLCs) differentiated from PomD-iPSCs recapitulated the hallmark Pompe disease pathophysiological phenotypes, including high levels of glycogen and multiple ultrastructural aberrances. Drug rescue assessment showed that exposure of PomD-iPSC-derived CMLCs to recombinant human GAA reversed the major pathologic phenotypes. Furthermore, l-carnitine treatment reduced defective cellular respiration in the diseased cells. By comparative transcriptome analysis, we identified glycogen metabolism, lysosome and mitochondria-related marker genes whose expression robustly correlated with the therapeutic effect of drug treatment in PomD-iPSC-derived CMLCs. Collectively, these results demonstrate that PomD-iPSCs are a promising in vitro disease model for the development of novel therapeutic strategies for Pompe disease.
Collapse
Affiliation(s)
- Hsiang-Po Huang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Raben N, Schreiner C, Baum R, Takikita S, Xu S, Xie T, Myerowitz R, Komatsu M, Van der Meulen JH, Nagaraju K, Ralston E, Plotz PH. Suppression of autophagy permits successful enzyme replacement therapy in a lysosomal storage disorder--murine Pompe disease. Autophagy 2011; 6:1078-89. [PMID: 20861693 DOI: 10.4161/auto.6.8.13378] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Autophagy, an intracellular system for delivering portions of cytoplasm and damaged organelles to lysosomes for degradation/recycling, plays a role in many physiological processes and is disturbed in many diseases. We recently provided evidence for the role of autophagy in Pompe disease, a lysosomal storage disorder in which acid alphaglucosidase, the enzyme involved in the breakdown of glycogen, is deficient or absent. Clinically the disease manifests as a cardiac and skeletal muscle myopathy. The current enzyme replacement therapy (ERT) clears lysosomal glycogen effectively from the heart but less so from skeletal muscle. In our Pompe model, the poor muscle response to therapy is associated with the presence of pools of autophagic debris. To clear the fibers of the autophagic debris, we have generated a Pompe model in which an autophagy gene, Atg7, is inactivated in muscle. Suppression of autophagy alone reduced the glycogen level by 50–60%. Following ERT, muscle glycogen was reduced to normal levels, an outcome not observed in Pompe mice with genetically intact autophagy. The suppression of autophagy, which has proven successful in the Pompe model, is a novel therapeutic approach that may be useful in other diseases with disturbed autophagy.
Collapse
Affiliation(s)
- Nina Raben
- Arthritis and Rheumatism Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Douillard-Guilloux G, Mouly V, Caillaud C, Richard E. Immortalization of murine muscle cells from lysosomal alpha-glucosidase deficient mice: a new tool to study pathophysiology and assess therapeutic strategies for Pompe disease. Biochem Biophys Res Commun 2009; 388:333-8. [PMID: 19665008 DOI: 10.1016/j.bbrc.2009.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Accepted: 08/01/2009] [Indexed: 10/20/2022]
Abstract
Glycogen storage disease type II (GSDII) is an autosomal recessive disorder caused by defects in the acid alpha-glucosidase (GAA) gene leading to lysosomal glycogen accumulation, mainly in cardiac and muscle tissues. In order to facilitate biological investigation on this disease and to avoid time-consuming direct cell isolation and culture, we have established murine myogenic GSDII cell lines. Lentiviral/retroviral expression of SV40 T antigen, Bmi-1 or cyclin-dependent kinase 4 (CDK4) genes was used to induce the immortalization of primary satellite cells from GSDII mice. The resulting immortalized myoblasts exhibit phenotypic characteristics of their parental cells, including profound GAA deficiency, glycogen accumulation and the ability to fully differentiate into myotubes when placed in proper culture conditions. These cell lines will constitute a powerful tool for both basic and applied studies focused on a better understanding of the pathophysiological mechanisms involved in GSDII and for assessing putative therapeutic strategies.
Collapse
Affiliation(s)
- Gaëlle Douillard-Guilloux
- Institut Cochin, Université Paris Descartes. CNRS (UMR 8104), Paris, France; INSERM, U567, Paris, France
| | | | | | | |
Collapse
|