1
|
Manoli I, Sysol JR, Head PE, Epping MW, Gavrilova O, Crocker MK, Sloan JL, Koutsoukos SA, Wang C, Ktena YP, Mendelson S, Pass AR, Zerfas PM, Hoffmann V, Vernon HJ, Fletcher LA, Reynolds JC, Tsokos MG, Stratakis CA, Voss SD, Chen KY, Brown RJ, Hamosh A, Berry GT, Chen XS, Yanovski JA, Venditti CP. Lipodystrophy in methylmalonic acidemia associated with elevated FGF21 and abnormal methylmalonylation. JCI Insight 2024; 9:e174097. [PMID: 38271099 DOI: 10.1172/jci.insight.174097] [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: 07/24/2023] [Accepted: 01/09/2024] [Indexed: 01/27/2024] Open
Abstract
A distinct adipose tissue distribution pattern was observed in patients with methylmalonyl-CoA mutase deficiency, an inborn error of branched-chain amino acid (BCAA) metabolism, characterized by centripetal obesity with proximal upper and lower extremity fat deposition and paucity of visceral fat, that resembles familial multiple lipomatosis syndrome. To explore brown and white fat physiology in methylmalonic acidemia (MMA), body composition, adipokines, and inflammatory markers were assessed in 46 patients with MMA and 99 matched controls. Fibroblast growth factor 21 levels were associated with acyl-CoA accretion, aberrant methylmalonylation in adipose tissue, and an attenuated inflammatory cytokine profile. In parallel, brown and white fat were examined in a liver-specific transgenic MMA mouse model (Mmut-/- TgINS-Alb-Mmut). The MMA mice exhibited abnormal nonshivering thermogenesis with whitened brown fat and had an ineffective transcriptional response to cold stress. Treatment of the MMA mice with bezafibrates led to clinical improvement with beiging of subcutaneous fat depots, which resembled the distribution seen in the patients. These studies defined what we believe to be a novel lipodystrophy phenotype in patients with defects in the terminal steps of BCAA oxidation and demonstrated that beiging of subcutaneous adipose tissue in MMA could readily be induced with small molecules.
Collapse
Affiliation(s)
- Irini Manoli
- Metabolic Medicine Branch, National Human Genome Research Institute
| | - Justin R Sysol
- Metabolic Medicine Branch, National Human Genome Research Institute
| | | | | | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases
| | - Melissa K Crocker
- Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development; and
| | - Jennifer L Sloan
- Metabolic Medicine Branch, National Human Genome Research Institute
| | | | - Cindy Wang
- Metabolic Medicine Branch, National Human Genome Research Institute
| | - Yiouli P Ktena
- Metabolic Medicine Branch, National Human Genome Research Institute
| | - Sophia Mendelson
- Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development; and
| | - Alexandra R Pass
- Metabolic Medicine Branch, National Human Genome Research Institute
| | - Patricia M Zerfas
- Office of Research Services, Division of Veterinary Resources, NIH, Bethesda, Maryland, USA
| | - Victoria Hoffmann
- Office of Research Services, Division of Veterinary Resources, NIH, Bethesda, Maryland, USA
| | - Hilary J Vernon
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Laura A Fletcher
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases
| | | | - Maria G Tsokos
- Ultrastructural Pathology Section, Center for Cancer Research; and
| | - Constantine A Stratakis
- Section on Endocrinology & Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Stephan D Voss
- Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kong Y Chen
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases
| | - Rebecca J Brown
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases
| | - Ada Hamosh
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Gerard T Berry
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaoyuan Shawn Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, NIH, Bethesda, Maryland, USA
| | - Jack A Yanovski
- Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development; and
| | | |
Collapse
|
2
|
Yi J, Zhou Q, Huang J, Niu S, Ji G, Zheng T. Lipid metabolism disorder promotes the development of intervertebral disc degeneration. Biomed Pharmacother 2023; 166:115401. [PMID: 37651799 DOI: 10.1016/j.biopha.2023.115401] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/22/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023] Open
Abstract
Lipid metabolism is a complex process that maintains the normal physiological function of the human body. The disorder of lipid metabolism has been implicated in various human diseases, such as cardiovascular diseases and bone diseases. Intervertebral disc degeneration (IDD), an age-related degenerative disease in the musculoskeletal system, is characterized by high morbidity, high treatment cost, and chronic recurrence. Lipid metabolism disorder may promote the pathogenesis of IDD, and the potential mechanisms are complex. Leptin, resistin, nicotinamide phosphoribosyltransferase (NAMPT), fatty acids, and cholesterol may promote the pathogenesis of IDD, while lipocalin, adiponectin, and progranulin (PGRN) exhibit protective activity against IDD development. Lipid metabolism disorder contributes to extracellular matrix (ECM) degradation, cell apoptosis, and cartilage calcification in the intervertebral discs (IVDs) by activating inflammatory responses, endoplasmic reticulum (ER) stress, and oxidative stress and inhibiting autophagy. Several lines of agents have been developed to target lipid metabolism disorder. Inhibition of lipid metabolism disorder may be an effective strategy for the therapeutic management of IDD. However, an in-depth understanding of the molecular mechanism of lipid metabolism disorder in promoting IDD development is still needed.
Collapse
Affiliation(s)
- Jun Yi
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Qingluo Zhou
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Jishang Huang
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Shuo Niu
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Guanglin Ji
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Tiansheng Zheng
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
3
|
Zhong J, Gou Y, Zhao P, Dong X, Guo M, Li A, Hao A, Luu HH, He TC, Reid RR, Fan J. Glycogen storage disease type I: Genetic etiology, clinical manifestations, and conventional and gene therapies. PEDIATRIC DISCOVERY 2023; 1:e3. [PMID: 38370424 PMCID: PMC10874634 DOI: 10.1002/pdi3.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 05/11/2023] [Indexed: 02/20/2024]
Abstract
Glycogen storage disease type I (GSDI) is an inherited metabolic disorder characterized by a deficiency of enzymes or proteins involved in glycogenolysis and gluconeogenesis, resulting in excessive intracellular glycogen accumulation. While GSDI is classified into four different subtypes based on molecular genetic variants, GSDIa accounts for approximately 80%. GSDIa and GSDIb are autosomal recessive disorders caused by deficiencies in glucose-6-phosphatase (G6Pase-α) and glucose-6-phosphate-transporter (G6PT), respectively. For the past 50 years, the care of patients with GSDI has been improved following elaborate dietary managements. GSDI patients currently receive dietary therapies that enable patients to improve hypoglycemia and alleviate early symptomatic signs of the disease. However, dietary therapies have many limitations with a risk of calcium, vitamin D, and iron deficiency and cannot prevent long-term complications, such as progressive liver and renal failure. With the deepening understanding of the pathogenesis of GSDI and the development of gene therapy technology, there is great progress in the treatment of GSDI. Here, we review the underlying molecular genetics and the current clinical management strategies of GSDI patients with an emphasis on promising experimental gene therapies.
Collapse
Affiliation(s)
- Jiamin Zhong
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - Yannian Gou
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - Piao Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangyu Dong
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Meichun Guo
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Aohua Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Ailing Hao
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
- Laboratory of Craniofacial Biology and Development, Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
- Laboratory of Craniofacial Biology and Development, Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - Jiaming Fan
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
| |
Collapse
|
4
|
Masschelin PM, Saha P, Ochsner SA, Cox AR, Kim KH, Felix JB, Sharp R, Li X, Tan L, Park JH, Wang L, Putluri V, Lorenzi PL, Nuotio-Antar AM, Sun Z, Kaipparettu BA, Putluri N, Moore DD, Summers SA, McKenna NJ, Hartig SM. Vitamin B2 enables regulation of fasting glucose availability. eLife 2023; 12:e84077. [PMID: 37417957 PMCID: PMC10328530 DOI: 10.7554/elife.84077] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 06/24/2023] [Indexed: 07/08/2023] Open
Abstract
Flavin adenine dinucleotide (FAD) interacts with flavoproteins to mediate oxidation-reduction reactions required for cellular energy demands. Not surprisingly, mutations that alter FAD binding to flavoproteins cause rare inborn errors of metabolism (IEMs) that disrupt liver function and render fasting intolerance, hepatic steatosis, and lipodystrophy. In our study, depleting FAD pools in mice with a vitamin B2-deficient diet (B2D) caused phenotypes associated with organic acidemias and other IEMs, including reduced body weight, hypoglycemia, and fatty liver disease. Integrated discovery approaches revealed B2D tempered fasting activation of target genes for the nuclear receptor PPARα, including those required for gluconeogenesis. We also found PPARα knockdown in the liver recapitulated B2D effects on glucose excursion and fatty liver disease in mice. Finally, treatment with the PPARα agonist fenofibrate activated the integrated stress response and refilled amino acid substrates to rescue fasting glucose availability and overcome B2D phenotypes. These findings identify metabolic responses to FAD availability and nominate strategies for the management of organic acidemias and other rare IEMs.
Collapse
Affiliation(s)
- Peter M Masschelin
- Department of Diabetes, Endocrinology, and Metabolism, Baylor College of MedicineHoustonUnited States
- Department of Medicine, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Pradip Saha
- Department of Diabetes, Endocrinology, and Metabolism, Baylor College of MedicineHoustonUnited States
- Department of Medicine, Baylor College of MedicineHoustonUnited States
| | - Scott A Ochsner
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Aaron R Cox
- Department of Diabetes, Endocrinology, and Metabolism, Baylor College of MedicineHoustonUnited States
- Department of Medicine, Baylor College of MedicineHoustonUnited States
| | - Kang Ho Kim
- Department of Anesthesiology, University of Texas Health Sciences CenterHoustonUnited States
| | - Jessica B Felix
- Department of Diabetes, Endocrinology, and Metabolism, Baylor College of MedicineHoustonUnited States
- Department of Medicine, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Robert Sharp
- Department of Diabetes, Endocrinology, and Metabolism, Baylor College of MedicineHoustonUnited States
- Department of Medicine, Baylor College of MedicineHoustonUnited States
| | - Xin Li
- Department of Diabetes, Endocrinology, and Metabolism, Baylor College of MedicineHoustonUnited States
- Department of Medicine, Baylor College of MedicineHoustonUnited States
| | - Lin Tan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | - Jun Hyoung Park
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Liping Wang
- Department of Nutrition and Integrative Physiology, University of UtahSalt Lake CityUnited States
| | - Vasanta Putluri
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | | | - Zheng Sun
- Department of Diabetes, Endocrinology, and Metabolism, Baylor College of MedicineHoustonUnited States
- Department of Medicine, Baylor College of MedicineHoustonUnited States
| | | | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - David D Moore
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Department of Nutritional Sciences and Toxicology, University of California, BerkeleyBerkeleyUnited States
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of UtahSalt Lake CityUnited States
| | - Neil J McKenna
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Sean M Hartig
- Department of Diabetes, Endocrinology, and Metabolism, Baylor College of MedicineHoustonUnited States
- Department of Medicine, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
5
|
Chandler RJ. From Puppies to adults: In vivo editing of hepatocytes in a canine model of glycogen storage disease type Ia. Mol Ther Methods Clin Dev 2023; 29:347-349. [PMID: 37206367 PMCID: PMC10188620 DOI: 10.1016/j.omtm.2023.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Affiliation(s)
- Randy J. Chandler
- Metabolic Medicine Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Corresponding author: Randy J. Chandler, PhD, MB Organic Acid Research Section Metabolic Medicine Branch National Human Genome Research Institute National Institutes of Health Bldg 10, Room 5B39 Bethesda, MD 20892, USA.
| |
Collapse
|
6
|
Rossi A, Assunto A, Rosano C, Tucci S, Ruoppolo M, Caterino M, Pirozzi F, Strisciuglio P, Parenti G, Melis D. Mitochondrial reprogramming in peripheral blood mononuclear cells of patients with glycogen storage disease type Ia. GENES & NUTRITION 2023; 18:10. [PMID: 37280548 DOI: 10.1186/s12263-023-00729-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 05/05/2023] [Indexed: 06/08/2023]
Abstract
BACKGROUND Glycogen storage disease type Ia (GSDIa) is an inborn metabolic disorder caused by the deficiency of glucose-6-phospatase-α (G6Pase-α) leading to mitochondrial dysfunction. It remains unclear whether mitochondrial dysfunction is present in patients' peripheral blood mononuclear cells (PBMC) and whether dietary treatment can play a role. The aim of this study was to investigate mitochondrial function in PBMC of GSDIa patients. METHODS Ten GSDIa patients and 10 age-, sex- and fasting-time matched controls were enrolled. Expression of genes involved in mitochondrial function and activity of key fatty acid oxidation (FAO) and Krebs cycle proteins were assessed in PBMC. Targeted metabolomics and assessment of metabolic control markers were also performed. RESULTS Adult GSDIa patients showed increased CPT1A, SDHB, TFAM, mTOR expression (p < 0.05) and increased VLCAD, CPT2 and citrate synthase activity in PBMC (p < 0.05). VLCAD activity directly correlated with WC (p < 0.01), BMI (p < 0.05), serum malonycarnitine levels (p < 0.05). CPT2 activity directly correlated with BMI (p < 0.05). CONCLUSION Mitochondrial reprogramming is detectable in PBMC of GSDIa patients. This feature may develop as an adaptation to the liver enzyme defect and may be triggered by dietary (over)treatment in the frame of G6Pase-α deficiency. PBMC can represent an adequate mean to assess (diet-induced) metabolic disturbances in GSDIa.
Collapse
Affiliation(s)
- Alessandro Rossi
- Department of Translational Medicine, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Antonia Assunto
- Department of Translational Medicine, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Carmen Rosano
- Department of Translational Medicine, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Sara Tucci
- Pharmacy, Medical Center - University of Freiburg, Hugstetterstr. 55, D-79106, Freiburg, Germany
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
- CEINGE Advanced Biotechnologies, Naples, Italy
| | - Marianna Caterino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
- CEINGE Advanced Biotechnologies, Naples, Italy
| | - Francesca Pirozzi
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
- CEINGE Advanced Biotechnologies, Naples, Italy
| | - Pietro Strisciuglio
- Department of Translational Medicine, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Giancarlo Parenti
- Department of Translational Medicine, Section of Pediatrics, University of Naples Federico II, Naples, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Daniela Melis
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Section of Pediatrics, University of Salerno, Via Salvador Allende, 43 84081, Baronissi (Salerno), Italy.
| |
Collapse
|
7
|
Seminotti B, Grings M, Glänzel NM, Vockley J, Leipnitz G. Peroxisome proliferator-activated receptor (PPAR) agonists as a potential therapy for inherited metabolic disorders. Biochem Pharmacol 2023; 209:115433. [PMID: 36709926 DOI: 10.1016/j.bcp.2023.115433] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023]
Abstract
Inherited metabolic disorders (IMDs) are genetic disorders that cause a disruption of a specific metabolic pathway leading to biochemical, clinical and pathophysiological sequelae. While the metabolite abnormalities in body fluids and tissues can usually be defined by directed or broad-spectrum metabolomic analysis, the pathophysiology of these changes is often not obvious. Mounting evidence has revealed that secondary mitochondrial dysfunction, mainly oxidative phosphorylation impairment and elevated reactive oxygen species, plays a pivotal role in many disorders. Peroxisomal proliferator-activated receptors (PPARs) consist of a group of nuclear hormone receptors (PPARα, PPARβ/δ, and PPARγ) that regulate multiple cellular functions and processes, including response to oxidative stress, inflammation, lipid metabolism, and mitochondrial bioenergetics and biogenesis. In this context, the activation of PPARs has been shown to stimulate oxidative phosphorylation and reduce reactive species levels. Thus, pharmacological treatment with PPAR activators, such as fibrates, has gained much attention in the last 15 years. This review summarizes preclinical (animal models and patient-derived cells) and clinical data on the effect of PPARs in IMDs.
Collapse
Affiliation(s)
- Bianca Seminotti
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil; Division of Genetic and Genomic Medicine, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mateus Grings
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil
| | - Nícolas Manzke Glänzel
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil
| | - Jerry Vockley
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guilhian Leipnitz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, CEP 90035-190, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil.
| |
Collapse
|
8
|
Petrova IO, Smirnikhina SA. Studies on glycogen storage disease type 1a animal models: a brief perspective. Transgenic Res 2022; 31:593-606. [PMID: 36006546 DOI: 10.1007/s11248-022-00325-7] [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: 01/25/2022] [Accepted: 08/09/2022] [Indexed: 01/20/2023]
Abstract
Glycogen storage disease type 1 (GSD1) is a rare hereditary monogenic disease characterized by the disturbed glucose metabolism. The most widespread variant of GSD1 is GSD1a, which is a deficiency of glucose-6-phosphatase-ɑ. Glucose-6-phosphatase-ɑ is expressed only in liver, kidney, and intestine, and these organs are primarily affected by its deficiency, and long-term complications of GSD1a include hepatic tumors and chronic liver disease. This article is a brief overview of existing animal models for GSD1a, from the first mouse model of 1996 to modern CRISPR/Cas9-generated ones. First whole-body murine models demonstrated exact metabolic symptoms of GSD1a, but the animals did not survive weaning. The protocol for glucose treatment allowed prolonged survival of affected animals, but long-term complications, such as hepatic tumorigenesis, could not be investigated. Next, organ-specific knockout models were developed, and most of the metabolic research was performed on liver glucose-6-phosphate-deficient mice. Naturally occuring mutation was also discovered in dogs. All these models are widely used to study GSD1a from metabolic and physiological standpoints and to develop possible treatments involving gene therapy. Research performed using these models helped elucidate the role of glycogen and lipid accumulation, hypoxia, mitochondrial dysfunction, and autophagy impairment in long-term complications of GSD1a, including hepatic tumorigenesis. Recently, gene replacement therapy and genome editing were tested on described models, and some of the developed approaches have reached clinical trials.
Collapse
Affiliation(s)
- Irina O Petrova
- Laboratory of Genome Editing, Research Center for Medical Genetics, Moskvorechye 1, Moscow, Russia, 115478.
| | - Svetlana A Smirnikhina
- Laboratory of Genome Editing, Research Center for Medical Genetics, Moskvorechye 1, Moscow, Russia, 115478
| |
Collapse
|
9
|
Tang Q, Liu W, Yang X, Tian Y, Chen J, Hu Y, Fu N. ATG5-Mediated Autophagy May Inhibit Pyroptosis to Ameliorate Oleic Acid-Induced Hepatocyte Steatosis. DNA Cell Biol 2022; 41:1038-1052. [PMID: 36473201 DOI: 10.1089/dna.2022.0265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Despite activated autophagy ameliorating hepatocyte steatosis and metabolic associated fatty liver disease (MAFLD), mechanisms underlying the beneficial roles of autophagy in hepatic deregulation of lipid metabolism remain undefined. We explored whether autophagy can ameliorate oleic acid (OA)-induced hepatic steatosis by suppressing pyroptosis. Pyroptosis is involved in hepatocyte steatosis induced by OA. In addition, autophagy flux was blocked in OA-treated hepatocytes. Treatment with OA induced lipid accumulation in liver cell line L-02, which was attenuated by rapamycin (Rap), an autophagy agonist, while aggravated by autophagy inhibitor bafilomycin A1 (Baf A1). Inversely, treatment with pyroptotic agonist Nigericin aggravated OA-induced hepatic steatosis, while pyroptosis antagonist disulfiram ameliorated this effect. Mechanistically, treatment with Rap downregulated the expression of pyroptosis-related proteins, including NLRP3, Caspase-1, IL-18, GSDMD expression evoked by OA, thus improving pyroptosis in hepatic steatosis. Significantly, overexpression of ATG5 obviously downregulated cleaved caspase-1 expressions without altering the total caspase1 expressions in hepatic cell steatosis. Taken together, our studies strongly demonstrated that the activation of ATG5 inhibits pyroptosis to improve hepatic steatosis and suggest autophagy activation as a potential therapeutic strategy for pyroptosis-mediated MAFLD.
Collapse
Affiliation(s)
- Qianyu Tang
- Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, The Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Wenhui Liu
- Department of Intensive Care Unit, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), Hengyang, China
| | - Xuefeng Yang
- Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, The Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Yaying Tian
- Department of Infectious and Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Jiacheng Chen
- Department of Intensive Care Unit, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Yang Hu
- Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, The Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Nian Fu
- Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, The Affiliated Nanhua Hospital, University of South China, Hengyang, China.,Clinical Research Institute, Hengyang Medical School, The Affiliated Nanhua Hospital, University of South China, Hengyang, China
| |
Collapse
|
10
|
Young EN, Dogan M, Watkins C, Bajwa A, Eason JD, Kuscu C, Kuscu C. A Review of Defatting Strategies for Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2022; 23:ijms231911805. [PMID: 36233107 PMCID: PMC9569609 DOI: 10.3390/ijms231911805] [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: 09/10/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/30/2022] Open
Abstract
Non-alcoholic fatty liver disease is a huge cause of chronic liver failure around the world. This condition has become more prevalent as rates of metabolic syndrome, type 2 diabetes, and obesity have also escalated. The unfortunate outcome for many people is liver cirrhosis that warrants transplantation or being unable to receive a transplant since many livers are discarded due to high levels of steatosis. Over the past several years, however, a great deal of work has gone into understanding the pathophysiology of this disease as well as possible treatment options. This review summarizes various defatting strategies including in vitro use of pharmacologic agents, machine perfusion of extracted livers, and genomic approaches targeting specific proteins. The goal of the field is to reduce the number of necessary transplants and expand the pool of organs available for use.
Collapse
|
11
|
da Rosa-Junior NT, Parmeggiani B, Glänzel NM, de Moura Alvorcem L, Brondani M, Britto R, Grings M, Ortiz VD, Turck P, da Rosa Araujo AS, Wajner M, Leipnitz G. Antioxidant system disturbances and mitochondrial dysfunction induced by 3-methyglutaric acid in rat heart are prevented by bezafibrate. Eur J Pharmacol 2022; 924:174950. [DOI: 10.1016/j.ejphar.2022.174950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 03/25/2022] [Accepted: 04/05/2022] [Indexed: 11/30/2022]
|
12
|
Mathis T, Poms M, Köfeler H, Gautschi M, Plecko B, Baumgartner MR, Hochuli M. Untargeted plasma metabolomics identifies broad metabolic perturbations in glycogen storage disease type I. J Inherit Metab Dis 2022; 45:235-247. [PMID: 34671989 PMCID: PMC9299190 DOI: 10.1002/jimd.12451] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND The metabolic defect in glycogen storage disease type I (GSDI) results in fasting hypoglycemia and typical secondary metabolic abnormalities (eg, hypertriglyceridemia, hyperlactatemia, hyperuricemia). The aim of this study was to assess further perturbations of the metabolic network in GSDI patients under ongoing treatment. METHODS In this prospective observational study, plasma samples of 14 adult patients (11 GSDIa, 3 GSDIb. Mean age 26.4 years, range 16-46 years) on standard treatment were compared to a cohort of 31 healthy controls utilizing ultra-high performance liquid chromatography (UHPLC) in combination with high resolution tandem mass spectrometry (HR-MS/MS) and subsequent statistical multivariate analysis. In addition, plasma fatty acid profiling was performed by GC/EI-MS. RESULTS The metabolomic profile showed alterations of metabolites in different areas of the metabolic network in both GSD subtypes, including pathways of fuel metabolism and energy generation, lipids and fatty acids, amino acid and methyl-group metabolism, the urea cycle, and purine/pyrimidine metabolism. These alterations were present despite adequate dietary treatment, did not correlate with plasma triglycerides or lactate, both parameters typically used to assess the quality of metabolic control in clinical practice, and were not related to the presence or absence of complications (ie, nephropathy or liver adenomas). CONCLUSION The metabolic defect of GSDI has profound effects on a variety of metabolic pathways in addition to the known typical abnormalities. These alterations are present despite optimized dietary treatment, which may contribute to the risk of developing long-term complications, an inherent problem of GSDI which appears to be only partly modified by current therapy.
Collapse
Affiliation(s)
- Tamara Mathis
- Division of Endocrinology, Diabetes, and Clinical NutritionUniversity Hospital Zurich, University of ZurichZurichSwitzerland
| | - Martin Poms
- Department of Clinical Chemistry and BiochemistryUniversity Children's Hospital Zurich, University of ZurichZurichSwitzerland
| | - Harald Köfeler
- Core Facility Mass SpectrometryMedical University of GrazGrazAustria
| | - Matthias Gautschi
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics and Institute of Clinical ChemistryUniversity Hospital Bern, InselspitalBernSwitzerland
| | - Barbara Plecko
- Department of Pediatrics and Adolescent MedicineMedical University of GrazGrazAustria
| | - Matthias R. Baumgartner
- Division of Metabolism and Children's Research Center (CRC)University Children's Hospital, Zurich, University of ZurichZurichSwitzerland
- radiz—Rare Disease Initiative Zurich, Clinical Research Priority Program for Rare DiseasesUniversity of ZurichZurichSwitzerland
| | - Michel Hochuli
- Division of Endocrinology, Diabetes, and Clinical NutritionUniversity Hospital Zurich, University of ZurichZurichSwitzerland
- radiz—Rare Disease Initiative Zurich, Clinical Research Priority Program for Rare DiseasesUniversity of ZurichZurichSwitzerland
- Department of Diabetes, Endocrinology, Nutritional Medicine and MetabolismInselspital, Bern University Hospital and University of BernBernSwitzerland
| |
Collapse
|
13
|
Dard L, Hubert C, Esteves P, Blanchard W, Bou About G, Baldasseroni L, Dumon E, Angelini C, Delourme M, Guyonnet-Duperat V, Claverol S, Bonneu M, Fontenille L, Kissa K, Séguéla PE, Thambo JB, Levy N, Herault Y, Bellance N, Dias Amoedo N, Magdinier F, Sorg T, Lacombe D, Rossignol R. HRAS germline mutations impair LKB1/AMPK signaling and mitochondrial homeostasis in Costello syndrome models. J Clin Invest 2022; 132:131053. [PMID: 35230976 PMCID: PMC9012293 DOI: 10.1172/jci131053] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 02/23/2022] [Indexed: 11/17/2022] Open
Abstract
Germline mutations that activate genes in the canonical RAS/MAPK signaling pathway are responsible for rare human developmental disorders known as RASopathies. Here, we analyzed the molecular determinants of Costello syndrome (CS) using a mouse model expressing HRAS p.G12S, patient skin fibroblasts, hiPSC-derived human cardiomyocytes, a HRAS p.G12V zebrafish model and human fibroblasts expressing lentiviral constructs carrying HRAS p.G12S or HRAS p.G12A mutations. The findings revealed alteration of mitochondrial proteostasis and defective oxidative phosphorylation in the heart and skeletal muscle of Costello mice that were also found in the cell models of the disease. The underpinning mechanisms involved the inhibition of the AMPK signaling pathway by mutant forms of HRAS, leading to alteration of mitochondrial proteostasis and bioenergetics. Pharmacological activation of mitochondrial bioenergetics and quality control restored organelle function in HRAS p.G12A and p.G12S cell models, reduced left ventricle hypertrophy in the CS mice and diminished the occurrence of developmental defects in the CS zebrafish model. Collectively, these findings highlight the importance of mitochondrial proteostasis in the pathophysiology of RASopathies and suggest that patients with Costello syndrome may benefit from treatment with mitochondrial modulators.
Collapse
Affiliation(s)
| | | | | | | | - Ghina Bou About
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch, France
| | | | - Elodie Dumon
- INSERM U688, University of Bordeaux, Bordeaux, France
| | | | | | | | | | - Marc Bonneu
- Plateforme Proteome, University of Bordeaux, Bordeaux, France
| | | | | | | | | | - Nicolas Levy
- Marseille Medical Genetics, INSERM, Marseille, France
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch, France
| | | | | | | | - Tania Sorg
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch, France
| | - Didier Lacombe
- Medical Genetics Department, Bordeaux University Hospital CHU Bordeaux, INSERM U121, Bordeaux, France
| | | |
Collapse
|
14
|
Berling É, Laforêt P, Wahbi K, Labrune P, Petit F, Ronzitti G, O'Brien A. Narrative review of glycogen storage disorder type III with a focus on neuromuscular, cardiac and therapeutic aspects. J Inherit Metab Dis 2021; 44:521-533. [PMID: 33368379 DOI: 10.1002/jimd.12355] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/19/2020] [Accepted: 12/22/2020] [Indexed: 12/26/2022]
Abstract
Glycogen storage disorder type III (GSDIII) is a rare inborn error of metabolism due to loss of glycogen debranching enzyme activity, causing inability to fully mobilize glycogen stores and its consequent accumulation in various tissues, notably liver, cardiac and skeletal muscle. In the pediatric population, it classically presents as hepatomegaly with or without ketotic hypoglycemia and failure to thrive. In the adult population, it should also be considered in the differential diagnosis of left ventricular hypertrophy or hypertrophic cardiomyopathy, myopathy, exercise intolerance, as well as liver cirrhosis or fibrosis with subsequent liver failure. In this review article, we first present an overview of the biochemical and clinical aspects of GSDIII. We then focus on the recent findings regarding cardiac and neuromuscular impairment associated with the disease. We review new insights into the pathophysiology and clinical picture of this disorder, including symptomatology, imaging and electrophysiology. Finally, we discuss current and upcoming treatment strategies such as gene therapy aimed at the replacement of the malfunctioning enzyme to provide a stable and long-term therapeutic option for this debilitating disease.
Collapse
Affiliation(s)
- Édouard Berling
- Généthon, Evry, France
- Université Paris-Saclay, Univ Evry, INSERM, Généthon, Integrare Research Unit UMR_S951, Evry, France
| | - Pascal Laforêt
- APHP, Department of Neurology, Raymond Poincaré Hospital, Centre de Référence de Pathologie Neuromusculaire Nord-Est-Ile-de-France, Garches, France
- INSERM U 1179, Université Versailles Saint Quentin en Yvelines, Paris-Saclay, France
| | - Karim Wahbi
- APHP, Cochin Hospital, Cardiology Department, FILNEMUS, Paris-Descartes, Sorbonne Paris Cité University, Paris, France
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France
- INSERM Unit 970, Paris Cardiovascular Research Centre (PARCC), Paris, France
| | - Philippe Labrune
- APHP, Université Paris-Saclay, Hôpital Antoine Béclère, Centre de Référence Maladies Héréditaires du Métabolisme Hépatique, Service de Pédiatrie, 92141 Clamart cedex, France
- INSERM U1195, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - François Petit
- Department of Genetics, APHP, Université Paris Saclay, Hôpital Antoine Béclère, Clamart, France
| | - Giuseppe Ronzitti
- Généthon, Evry, France
- Université Paris-Saclay, Univ Evry, INSERM, Généthon, Integrare Research Unit UMR_S951, Evry, France
| | - Alan O'Brien
- Généthon, Evry, France
- Service de Médecine Génique, Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Quebec, Canada
| |
Collapse
|
15
|
Zhou J, Singh BK, Ho JP, Lim A, Bruinstroop E, Ohba K, Sinha RA, Yen PM. MED1 mediator subunit is a key regulator of hepatic autophagy and lipid metabolism. Autophagy 2021; 17:4043-4061. [PMID: 33734012 DOI: 10.1080/15548627.2021.1899691] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatic macroautophagy/autophagy and fatty acid metabolism are transcriptionally regulated by nuclear receptors (NRs); however, it is not known whether their transcriptional co-activators are involved in autophagy. We thus examined MED1 (mediator complex subunit 1), a key component of the Mediator Complex that directly interacts with NRs, on these processes. We found that MED1 knockdown (KD) in cultured hepatic cells decreased autophagy and mitochondrial activity that was accompanied by decreased transcription of genes involved in these processes. Lipophagy and fatty acid β-oxidation also were impaired. These effects also occurred after thyroid hormone stimulation, nutrient-replete or -deplete conditions, and in liver-specific Med1 KD (Med1 LKD) mice under fed and fasting conditions. Together, these findings showed that Med1 played a key role in hepatic autophagy, mitochondria function, and lipid metabolism under these conditions. Additionally, we identified downregulated hepatic genes in Med1 LKD mice, and subjected them to ChIP Enrichment Analysis. Our findings showed that the transcriptional activity of several NRs and transcription factors (TFs), including PPARA and FOXO1, likely were affected by Med1 LKD. Finally, Med1 expression and autophagy also were decreased in two mouse models of nonalcoholic fatty liver disease (NAFLD) suggesting that decreased Med1 may contribute to hepatosteatosis. In summary, MED1 plays an essential role in regulating hepatic autophagy and lipid oxidation during different hormonal and nutrient conditions. Thus, MED1 may serve as an integrator of multiple transcriptional pathways involved in these metabolic processes.Abbreviations: BAF: bafilomycin A1; db/db mice; Leprdb/db mice; ECAR: extracellular acidification rate; KD: knockdown; MED1: mediator complex subunit 1; NAFLD: nonalcoholic fatty liver disease; OCR: oxygen consumption rate; PPARA/PPARα: peroxisomal proliferator activated receptor alpha; TF: transcription factor; TFEB: transcription factor EB; tf-LC3: tandem fluorescence RFP-GFP-LC3; TG: triglyceride; TH: Thyroid hormone; TR: thyroid hormone receptors; V-ATPase: vacuolar-type H+-ATPase; WDF: Western diet with 15% fructose in drinking water.
Collapse
Affiliation(s)
- Jin Zhou
- Program of Cardiovascular & Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore
| | - Brijesh K Singh
- Program of Cardiovascular & Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore
| | - Jia Pei Ho
- Program of Cardiovascular & Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore
| | - Andrea Lim
- Program of Cardiovascular & Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore
| | - Eveline Bruinstroop
- Department of Endocrinology and Metabolism, Amsterdam UMC, Amsterdam, The Netherlands
| | - Kenji Ohba
- Program of Cardiovascular & Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore
| | - Rohit A Sinha
- Program of Cardiovascular & Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore.,Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow India
| | - Paul M Yen
- Program of Cardiovascular & Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore.,Duke Molecular Physiology Institute and Department of Medicine, Duke University Medical Center, Durham, NC USA
| |
Collapse
|
16
|
Cho JH, Weinstein DA, Lee YM. Emerging roles of autophagy in hepatic tumorigenesis and therapeutic strategies in glycogen storage disease type Ia: A review. J Inherit Metab Dis 2021; 44:118-128. [PMID: 32474930 DOI: 10.1002/jimd.12267] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/19/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022]
Abstract
Glycogen storage disease type Ia (GSD-Ia) is an inherited metabolic disease caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC) which plays a critical role in blood glucose homeostasis by catalyzing the hydrolysis of glucose-6-phosphate (G6P) to glucose and phosphate in the terminal step of glycogenolysis and gluconeogenesis. Patients with GSD-Ia manifest life-threatening fasting hypoglycemia along with the excessive accumulation of hepatic glycogen and triglycerides which results in hepatomegaly and a risk of long-term complications such as hepatocellular adenoma and carcinoma (HCA/HCC). The etiology of HCA/HCC development in GSD-Ia, however, is unknown. Recent studies have shown that the livers in model animals of GSD-Ia display impairment of autophagy, a cellular recycling process which is critical for energy metabolism and cellular homeostasis. However, molecular mechanisms of autophagy impairment and its involvement in pathogenesis in GSD-Ia are still under investigation. Here, we summarize the latest advances for signaling pathways implicated in hepatic autophagy impairment and the roles of autophagy in hepatic tumorigenesis in GSD-Ia. In addition, recent evidence has illustrated that autophagy plays an important role in hepatic metabolism and liver-directed gene therapy mediated by recombinant adeno-associated virus (rAAV). Therefore, we highlight the possible role of hepatic autophagy in metabolic control and rAAV-mediated gene therapy for GSD-Ia. In this review, we also provide potential therapeutic strategies for GSD-Ia on the basis of molecular mechanisms underlying hepatic autophagy impairment in GSD-Ia.
Collapse
Affiliation(s)
- Jun-Ho Cho
- Glycogen Storage Disease Program, Department of Pediatrics, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - David A Weinstein
- Glycogen Storage Disease Program, Department of Pediatrics, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Glycogen Storage Disease Program, Connecticut Children's Medical Center, Hartford, Connecticut, USA
| | - Young Mok Lee
- Glycogen Storage Disease Program, Department of Pediatrics, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
17
|
Aoudjehane L, Gautheron J, Le Goff W, Goumard C, Gilaizeau J, Nget CS, Savier E, Atif M, Lesnik P, Morichon R, Chrétien Y, Calmus Y, Scatton O, Housset C, Conti F. Novel defatting strategies reduce lipid accumulation in primary human culture models of liver steatosis. Dis Model Mech 2020; 13:dmm042663. [PMID: 32094147 PMCID: PMC7197711 DOI: 10.1242/dmm.042663] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/13/2020] [Indexed: 12/13/2022] Open
Abstract
Normothermic perfusion provides a means to rescue steatotic liver grafts, including by pharmacological defatting. In this study, we tested the potential of new drug combinations to trigger defatting in three human culture models, primary hepatocytes with induced steatosis, primary hepatocytes isolated from steatotic liver, and precision-cut liver slices (PCLS) of steatotic liver. Forskolin, L-carnitine and a PPARα agonist were all combined with rapamycin, an immunosuppressant that induces autophagy, in a D-FAT cocktail. D-FAT was tested alone or in combination with necrosulfonamide, an inhibitor of mixed lineage kinase domain like pseudokinase involved in necroptosis. Within 24 h, in all three models, D-FAT induced a decrease in triglyceride content by 30%, attributable to an upregulation of genes involved in free fatty acid β-oxidation and autophagy, and a downregulation of those involved in lipogenesis. Defatting was accompanied by a decrease in endoplasmic reticulum stress and in the production of reactive oxygen species. The addition of necrosulfonamide increased the efficacy of defatting by 8%-12% in PCLS, with a trend towards increased autophagy. In conclusion, culture models, notably PCLS, are insightful to design strategies for liver graft rescue. Defatting can be rapidly achieved by combinations of drugs targeting mitochondrial oxidative metabolism, macro-autophagy and lipogenesis.
Collapse
Affiliation(s)
- Lynda Aoudjehane
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris 75012, France
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris 75013, France
| | - Jérémie Gautheron
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris 75012, France
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris 75013, France
| | - Wilfried Le Goff
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris 75012, France
| | - Claire Goumard
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris 75012, France
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris 75013, France
- Department of Hepatobiliary and Liver Transplantation Surgery, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, Paris 75013, France
| | - Julia Gilaizeau
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris 75012, France
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris 75013, France
| | - Chan Sonavine Nget
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris 75012, France
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris 75013, France
| | - Eric Savier
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris 75012, France
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris 75013, France
- Department of Hepatobiliary and Liver Transplantation Surgery, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, Paris 75013, France
| | - Muhammad Atif
- Centre d'immunologie et maladies infectieuses, Sorbonne Université, INSERM, U1135, Paris 75013, France
| | - Philippe Lesnik
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris 75012, France
| | - Romain Morichon
- Production et Analyse des données en Sciences de la vie et en Santé (PASS), Sorbonne Université, INSERM, UMS 37, Paris 75013, France
| | - Yves Chrétien
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris 75012, France
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris 75013, France
| | - Yvon Calmus
- Department of Medical Liver Transplantation, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, Paris 75013, France
| | - Olivier Scatton
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris 75012, France
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris 75013, France
- Department of Hepatobiliary and Liver Transplantation Surgery, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, Paris 75013, France
| | - Chantal Housset
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris 75012, France
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris 75013, France
- Department of Hepatology, Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Paris 75012, France
| | - Filomena Conti
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris 75012, France
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris 75013, France
- Department of Medical Liver Transplantation, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, Paris 75013, France
| |
Collapse
|
18
|
Sinha RA, Rajak S, Singh BK, Yen PM. Hepatic Lipid Catabolism via PPARα-Lysosomal Crosstalk. Int J Mol Sci 2020; 21:ijms21072391. [PMID: 32244266 PMCID: PMC7170715 DOI: 10.3390/ijms21072391] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/01/2020] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors which belong to the nuclear hormone receptor superfamily. They regulate key aspects of energy metabolism within cells. Recently, PPARα has been implicated in the regulation of autophagy-lysosomal function, which plays a key role in cellular energy metabolism. PPARα transcriptionally upregulates several genes involved in the autophagy-lysosomal degradative pathway that participates in lipolysis of triglycerides within the hepatocytes. Interestingly, a reciprocal regulation of PPARα nuclear action by autophagy-lysosomal activity also exists with implications in lipid metabolism. This review succinctly discusses the unique relationship between PPARα nuclear action and lysosomal activity and explores its impact on hepatic lipid homeostasis under pathological conditions such as non-alcoholic fatty liver disease (NAFLD).
Collapse
Affiliation(s)
- Rohit A. Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India;
- Correspondence: or
| | - Sangam Rajak
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India;
| | - Brijesh K. Singh
- Program of Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore 169587, Singapore (P.M.Y.)
| | - Paul M. Yen
- Program of Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore 169587, Singapore (P.M.Y.)
| |
Collapse
|
19
|
Douiev L, Sheffer R, Horvath G, Saada A. Bezafibrate Improves Mitochondrial Fission and Function in DNM1L-Deficient Patient Cells. Cells 2020; 9:cells9020301. [PMID: 32012656 PMCID: PMC7072316 DOI: 10.3390/cells9020301] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 12/13/2022] Open
Abstract
Mitochondria are involved in many cellular processes and their main role is cellular energy production. They constantly undergo fission and fusion, and these counteracting processes are under strict balance. The cytosolic dynamin-related protein 1, Drp1, or dynamin-1-like protein (DNM1L) mediates mitochondrial and peroxisomal division. Defects in the DNM1L gene result in a complex neurodevelopmental disorder with heterogeneous symptoms affecting multiple organ systems. Currently there is no curative treatment available for this condition. We have previously described a patient with a de novo heterozygous c.1084G>A (p.G362S) DNM1L mutation and studied the effects of a small molecule, bezafibrate, on mitochondrial functions in this patient’s fibroblasts compared to controls. Bezafibrate normalized growth on glucose-free medium, as well as ATP production and oxygen consumption. It improved mitochondrial morphology in the patient’s fibroblasts, although causing a mild increase in ROS production at the same time. A human foreskin fibroblast cell line overexpressing the p.G362S mutation showed aberrant mitochondrial morphology, which normalized in the presence of bezafibrate. Further studies would be needed to show the consistency of the response to bezafibrate, possibly using fibroblasts from patients with different mutations in DNM1L, and this treatment should be confirmed in clinical trials. However, taking into account the favorable effects in our study, we suggest that bezafibrate could be offered as a treatment option for patients with certain DNM1L mutations.
Collapse
Affiliation(s)
- Liza Douiev
- Department of Genetic and Metabolic Diseases and Jacques Roboh Department of Genetic Research, Hadassah Medical Center, Jerusalem 9112001, Israel;
- Correspondence: (L.D.); or (A.S.); Tel.: +972-26776844 (L.D. & A.S.); Fax: +972-26779018 (L.D. & A.S.)
| | - Ruth Sheffer
- Department of Genetic and Metabolic Diseases and Jacques Roboh Department of Genetic Research, Hadassah Medical Center, Jerusalem 9112001, Israel;
| | - Gabriella Horvath
- Division of Biochemical Diseases, Department of Pediatrics, University of British Columbia, Vancouver, BC BC V6H 3V4, Canada;
| | - Ann Saada
- Department of Genetic and Metabolic Diseases and Jacques Roboh Department of Genetic Research, Hadassah Medical Center, Jerusalem 9112001, Israel;
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel
- Correspondence: (L.D.); or (A.S.); Tel.: +972-26776844 (L.D. & A.S.); Fax: +972-26779018 (L.D. & A.S.)
| |
Collapse
|
20
|
Yavarow ZA, Kang HR, Waskowicz LR, Bay BH, Young SP, Yen PM, Koeberl DD. Fenofibrate rapidly decreases hepatic lipid and glycogen storage in neonatal mice with glycogen storage disease type Ia. Hum Mol Genet 2020; 29:286-294. [PMID: 31816064 PMCID: PMC7003036 DOI: 10.1093/hmg/ddz290] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/20/2019] [Accepted: 12/02/2019] [Indexed: 12/26/2022] Open
Abstract
Glycogen storage disease type Ia (GSD Ia) is caused by autosomal mutations in glucose-6-phosphatase α catalytic subunit (G6PC) and can present with severe hypoglycemia, lactic acidosis and hypertriglyceridemia. In both children and adults with GSD Ia, there is over-accumulation of hepatic glycogen and triglycerides that can lead to steatohepatitis and a risk for hepatocellular adenoma or carcinoma. Here, we examined the effects of the commonly used peroxisomal proliferated activated receptor α agonist, fenofibrate, on liver and kidney autophagy and lipid metabolism in 5-day-old G6pc -/- mice serving as a model of neonatal GSD Ia. Five-day administration of fenofibrate decreased the elevated hepatic and renal triglyceride and hepatic glycogen levels found in control G6pc -/- mice. Fenofibrate also induced autophagy and promoted β-oxidation of fatty acids and stimulated gene expression of acyl-CoA dehydrogenases in the liver. These findings show that fenofibrate can rapidly decrease hepatic glycogen and triglyceride levels and renal triglyceride levels in neonatal G6pc -/- mice. Moreover, since fenofibrate is an FDA-approved drug that has an excellent safety profile, our findings suggest that fenofibrate could be a potential pharmacological therapy for GSD Ia in neonatal and pediatric patients as well as for adults. These findings may also apply to non-alcoholic fatty liver disease, which shares similar pathological and metabolic changes with GSD Ia.
Collapse
Affiliation(s)
- Zollie A Yavarow
- Department of Pharmacology, Duke University, Durham NC 27710, USA
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham NC 27710, USA
| | - Hye-Ri Kang
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham NC 27710, USA
| | - Lauren R Waskowicz
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham NC 27710, USA
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore
| | - Sarah P Young
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham NC 27710, USA
| | - Paul M Yen
- Cardiovascular and Metabolic Disorders Program, Duke—National University of Singapore Graduate Medical School Singapore, Singapore 169547, Singapore
| | - Dwight D Koeberl
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| |
Collapse
|
21
|
Rajas F, Gautier-Stein A, Mithieux G. Glucose-6 Phosphate, A Central Hub for Liver Carbohydrate Metabolism. Metabolites 2019; 9:metabo9120282. [PMID: 31756997 PMCID: PMC6950410 DOI: 10.3390/metabo9120282] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/14/2019] [Accepted: 11/18/2019] [Indexed: 12/23/2022] Open
Abstract
Cells efficiently adjust their metabolism according to the abundance of nutrients and energy. The ability to switch cellular metabolism between anabolic and catabolic processes is critical for cell growth. Glucose-6 phosphate is the first intermediate of glucose metabolism and plays a central role in the energy metabolism of the liver. It acts as a hub to metabolically connect glycolysis, the pentose phosphate pathway, glycogen synthesis, de novo lipogenesis, and the hexosamine pathway. In this review, we describe the metabolic fate of glucose-6 phosphate in a healthy liver and the metabolic reprogramming occurring in two pathologies characterized by a deregulation of glucose homeostasis, namely type 2 diabetes, which is characterized by fasting hyperglycemia; and glycogen storage disease type I, where patients develop severe hypoglycemia during short fasting periods. In these two conditions, dysfunction of glucose metabolism results in non-alcoholic fatty liver disease, which may possibly lead to the development of hepatic tumors. Moreover, we also emphasize the role of the transcription factor carbohydrate response element-binding protein (ChREBP), known to link glucose and lipid metabolisms. In this regard, comparing these two metabolic diseases is a fruitful approach to better understand the key role of glucose-6 phosphate in liver metabolism in health and disease.
Collapse
Affiliation(s)
- Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213, F-69008 Lyon, France; (A.G.-S.); (G.M.)
- Université de Lyon, F-69008 Lyon, France
- Université Lyon 1, F-69622 Villeurbanne, France
- Correspondence:
| | - Amandine Gautier-Stein
- Institut National de la Santé et de la Recherche Médicale, U1213, F-69008 Lyon, France; (A.G.-S.); (G.M.)
- Université de Lyon, F-69008 Lyon, France
- Université Lyon 1, F-69622 Villeurbanne, France
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, F-69008 Lyon, France; (A.G.-S.); (G.M.)
- Université de Lyon, F-69008 Lyon, France
- Université Lyon 1, F-69622 Villeurbanne, France
| |
Collapse
|
22
|
Pathogenesis of Hepatic Tumors following Gene Therapy in Murine and Canine Models of Glycogen Storage Disease. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 15:383-391. [PMID: 31890731 PMCID: PMC6909089 DOI: 10.1016/j.omtm.2019.10.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 10/30/2019] [Indexed: 12/15/2022]
Abstract
Glycogen storage disease type Ia (GSD Ia) is caused by mutations in the glucose-6-phosphatase (G6Pase) catalytic subunit gene (G6PC). GSD Ia complications include hepatocellular adenomas (HCA) with a risk for hepatocellular carcinoma (HCC) formation. Genome editing with adeno-associated virus (AAV) vectors containing a zinc-finger nuclease (ZFN) and a G6PC donor transgene was evaluated in adult mice with GSD Ia. Although mouse livers expressed G6Pase, HCA and HCC occurred following AAV vector administration. Interestingly, vector genomes were almost undetectable in the tumors but remained relatively high in adjacent liver (p < 0.01). G6Pase activity was decreased in tumors, in comparison with adjacent liver (p < 0.01). Furthermore, AAV-G6Pase vector-treated dogs with GSD Ia developed HCC with lower G6Pase activity (p < 0.01) in comparison with adjacent liver. AAV integration and tumor marker analysis in mice revealed that tumors arose from the underlying disorder, not from vector administration. Similarly to human GSD Ia-related HCA and HCC, mouse and dog tumors did not express elevated α-fetoprotein. Taken together, these results suggest that AAV-mediated gene therapy not only corrects hepatic G6Pase deficiency, but also has potential to suppress HCA and HCC in the GSD Ia liver.
Collapse
|
23
|
Wei XY, Yang YJ, Zhu XH. The effect of bezafibrate in preventing glucolipid abnormalities induced by the antipsychotic risperidone. Psychiatry Res 2019; 281:112584. [PMID: 31586837 DOI: 10.1016/j.psychres.2019.112584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 11/19/2022]
Abstract
The present study aimed to investigate the effect of bezafibrate on glucolipid abnormalities induced by antipsychotics in schizophrenia. Patients in the treatment group (group A) were treated with antipsychotics and a daily dose of 200 mg bezafibrate for 12 weeks, and patients in the control group (group B) were treated with antipsychotics; sugar, fat and weight changes before and after the treatment were compared between the two groups. Before treatment the differences in TG, TC, LDL-C, HDL-C, body weight and blood glucose between groups A and B were not statistically significant. However, in group B, levels of TG, TC, LDL-C, body weight and blood glucose after treatment showed statistically significant increases, although levels of HDL-C did not register any statistically significant change. By contract, in group A, there were no statistically significant changes in any of the variables measured. Bezafibrate can prevent an increase in sugar, fat and weight gain in treating schizophrenia patients with antipsychotics, and low doses of bezafibrate are safe in the antipsychotic treatment for schizophrenia.
Collapse
Affiliation(s)
- Xian-Yu Wei
- Department of Psychology, Xuzhou Oriental People's Hospital, Xuzhou 221004, China.
| | - Yong-Jie Yang
- Department of Psychology, Xuzhou Oriental People's Hospital, Xuzhou 221004, China
| | - Xiang-Hua Zhu
- Department of Psychology, Xuzhou Oriental People's Hospital, Xuzhou 221004, China
| |
Collapse
|
24
|
Kishnani PS, Sun B, Koeberl DD. Gene therapy for glycogen storage diseases. Hum Mol Genet 2019; 28:R31-R41. [PMID: 31227835 PMCID: PMC6796997 DOI: 10.1093/hmg/ddz133] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/02/2019] [Accepted: 06/07/2019] [Indexed: 12/17/2022] Open
Abstract
The focus of this review is the development of gene therapy for glycogen storage diseases (GSDs). GSD results from the deficiency of specific enzymes involved in the storage and retrieval of glucose in the body. Broadly, GSDs can be divided into types that affect liver or muscle or both tissues. For example, glucose-6-phosphatase (G6Pase) deficiency in GSD type Ia (GSD Ia) affects primarily the liver and kidney, while acid α-glucosidase (GAA) deficiency in GSD II causes primarily muscle disease. The lack of specific therapy for the GSDs has driven efforts to develop new therapies for these conditions. Gene therapy needs to replace deficient enzymes in target tissues, which has guided the planning of gene therapy experiments. Gene therapy with adeno-associated virus (AAV) vectors has demonstrated appropriate tropism for target tissues, including the liver, heart and skeletal muscle in animal models for GSD. AAV vectors transduced liver and kidney in GSD Ia and striated muscle in GSD II mice to replace the deficient enzyme in each disease. Gene therapy has been advanced to early phase clinical trials for the replacement of G6Pase in GSD Ia and GAA in GSD II (Pompe disease). Other GSDs have been treated in proof-of-concept studies, including GSD III, IV and V. The future of gene therapy appears promising for the GSDs, promising to provide more efficacious therapy for these disorders in the foreseeable future.
Collapse
Affiliation(s)
- Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Baodong Sun
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Dwight D Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| |
Collapse
|
25
|
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
|
26
|
Jauze L, Monteillet L, Mithieux G, Rajas F, Ronzitti G. Challenges of Gene Therapy for the Treatment of Glycogen Storage Diseases Type I and Type III. Hum Gene Ther 2019; 30:1263-1273. [PMID: 31319709 DOI: 10.1089/hum.2019.102] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glycogen storage diseases (GSDs) type I (GSDI) and type III (GSDIII), the most frequent hepatic GSDs, are due to defects in glycogen metabolism, mainly in the liver. In addition to hypoglycemia and liver pathology, renal, myeloid, or muscle complications affect GSDI and GSDIII patients. Currently, patient management is based on dietary treatment preventing severe hypoglycemia and increasing the lifespan of patients. However, most of the patients develop long-term pathologies. In the past years, gene therapy for GSDI has generated proof of concept for hepatic GSDs. This resulted in a recent clinical trial of adeno-associated virus (AAV)-based gene replacement for GSDIa. However, the current limitations of AAV-mediated gene transfer still represent a challenge for successful gene therapy in GSDI and GSDIII. Indeed, transgene loss over time was observed in GSDI liver, possibly due to the degeneration of hepatocytes underlying the physiopathology of both GSDI and GSDIII and leading to hepatic tumor development. Moreover, multitissue targeting requires high vector doses to target nonpermissive tissues such as muscle and kidney. Interestingly, recent pharmacological interventions or dietary regimen aiming at the amelioration of the hepatocyte abnormalities before the administration of gene therapy demonstrated improved efficacy in GSDs. In this review, we describe the advances in gene therapy and the limitations to be overcome to achieve efficient and safe gene transfer in GSDs.
Collapse
Affiliation(s)
- Louisa Jauze
- INTEGRARE, Genethon, Inserm, Université d'Evry, Université Paris-Saclay, Evry, France.,Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, France.,Université de Lyon, Lyon, France.,Université Lyon I, Villeurbanne, France
| | - Laure Monteillet
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, France.,Université de Lyon, Lyon, France.,Université Lyon I, Villeurbanne, France
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, France.,Université de Lyon, Lyon, France.,Université Lyon I, Villeurbanne, France
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, France.,Université de Lyon, Lyon, France.,Université Lyon I, Villeurbanne, France
| | - Giuseppe Ronzitti
- INTEGRARE, Genethon, Inserm, Université d'Evry, Université Paris-Saclay, Evry, France
| |
Collapse
|
27
|
Zhou J, Waskowicz LR, Lim A, Liao XH, Lian B, Masamune H, Refetoff S, Tran B, Koeberl DD, Yen PM. A Liver-Specific Thyromimetic, VK2809, Decreases Hepatosteatosis in Glycogen Storage Disease Type Ia. Thyroid 2019; 29:1158-1167. [PMID: 31337282 PMCID: PMC6707038 DOI: 10.1089/thy.2019.0007] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Glycogen storage disease type Ia (GSD Ia), also known as von Gierke disease, is the most common glycogen storage disorder. It is caused by the deficiency of glucose-6-phosphatase, the enzyme that catalyzes the final step of gluconeogenesis and glycogenolysis. The accumulation of glucose-6-phosphate leads to increased glycogen and triglyceride levels in the liver. Patients with GSD Ia can develop steatohepatitis, cirrhosis, and increased risk for hepatocellular adenomas and carcinomas. We previously showed that animal models of GSD Ia had defective autophagy and dysfunctional mitochondria. In this study, we examined the effect of VK2809, a liver-specific thyroid hormone receptor β agonist, on hepatic steatosis, autophagy, and mitochondrial biogenesis in a mouse model of GSD Ia. Methods:G6pc-/--deficient (GSD Ia) mice were treated with VK2809 or vehicle control by daily intraperitoneal injection for four days. The hepatic triglyceride and glycogen were determined by biochemical assays. Autophagy and mitochondrial biogenesis were measured by Western blotting for key autophagy and mitochondrial markers. Results: VK2809 treatment decreased hepatic mass and triglyceride content in GSD Ia mice. VK2809 stimulated hepatic autophagic flux as evidenced by increased microtubule-associated protein light chain 3-II (LC3B-II), decreased p62 protein levels, activation of AMP-activated protein kinase (AMPK), inhibition of the mammalian target of rapamycin (mTOR) signaling, enhancement of protein levels of ATG5-ATG12, and increased lysosomal protein expression. VK2809 also increased the expression of carnitine palmitoyltransferase 1a (CPT1α) and fibroblast growth factor 21 (FGF21), as well as mitochondrial biogenesis to promote mitochondrial β-oxidation. Conclusions: In summary, VK2809 treatment decreased hepatic triglyceride levels in GSD Ia mice through its simultaneous restoration of autophagy, mitochondrial biogenesis, and β-oxidation of fatty acids. Liver-specific thyromimetics represent a potential therapy for hepatosteatosis in GSD Ia as well as nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Jin Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Lauren R. Waskowicz
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina
| | - Andrea Lim
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Xiao-Hui Liao
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Brian Lian
- Viking Therapeutics, San Diego, California
| | | | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, Illinois
- Department of Pediatrics and Committee on Genetics, The University of Chicago, Chicago, Illinois
| | - Brian Tran
- Viking Therapeutics, San Diego, California
| | - Dwight D. Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina
- Address correspondence to: Dwight D. Koeberl, MD, PhD, Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, DUMC Box 103856, Durham, NC, 27710
| | - Paul M. Yen
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina
- Paul M. Yen, MD, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, 8 College Road, Singapore 169587, Singapore
| |
Collapse
|
28
|
Kang HR, Waskowicz L, Seifts AM, Landau DJ, Young SP, Koeberl DD. Bezafibrate Enhances AAV Vector-Mediated Genome Editing in Glycogen Storage Disease Type Ia. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 13:265-273. [PMID: 30859111 PMCID: PMC6395830 DOI: 10.1016/j.omtm.2019.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 02/06/2019] [Indexed: 01/06/2023]
Abstract
Glycogen storage disease type Ia (GSD Ia) is a rare inherited disease caused by mutations in the glucose-6-phosphatase (G6Pase) catalytic subunit gene (G6PC). Absence of G6Pase causes life-threatening hypoglycemia and long-term complications because of the accumulations of metabolic intermediates. Bezafibrate, a pan-peroxisome proliferator-activated receptor (PPAR) agonist, was administered in the context of genome editing with a zinc-finger nuclease-containing vector (AAV-ZFN) and a G6Pase donor vector (AAV-RoG6P). Bezafibrate treatment increased survival and decreased liver size (liver/body mass, p < 0.05) in combination with genome editing. Blood glucose has higher (p < 0.05) after 4 h of fasting, and liver glycogen accumulation (p < 0.05) was lower in association with higher G6Pase activity (p < 0.05). Furthermore, bezafibrate-treated mice had increased numbers of G6PC transgenes (p < 0.05) and higher ZFN activity (p < 0.01) in the liver compared with controls. PPAR-α expression was increased and PPAR-γ expression was decreased in bezafibrate-treated mice. Therefore, bezafibrate improved hepatocellular abnormalities and increased the transduction efficiency of AAV vector-mediated genome editing in liver, whereas higher expression of G6Pase corrected molecular signaling in GSD Ia. Taken together, bezafibrate shows promise as a drug for increasing AAV vector-mediated genome editing.
Collapse
Affiliation(s)
- Hye-Ri Kang
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Lauren Waskowicz
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Andrea M. Seifts
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Dustin J. Landau
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Sarah P. Young
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Dwight D. Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
- Corresponding author: Dwight D. Koeberl, Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Box 103856, Durham, NC 27710, USA.
| |
Collapse
|