51
|
Urine glucose tetrasaccharide: A good biomarker for glycogenoses type II and III? A study of the French cohort. Mol Genet Metab Rep 2020; 23:100583. [PMID: 32382504 PMCID: PMC7200937 DOI: 10.1016/j.ymgmr.2020.100583] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/19/2020] [Accepted: 03/21/2020] [Indexed: 12/25/2022] Open
Key Words
- ACN, Acetonitrile
- BAB, Butyl-4-aminobenzoate
- CRIM, Cross Immune Reactive Material
- ERT, Enzyme Replacement Therapy
- GSD, Glycogen Storage Disease
- GVUS, Genetic Variant of Unknown Significance
- Glc4, Glcα1-6Glcα1-4Glcα1-4Glc, tetraglucose,
- IOPD, Infantile-Onset Pompe disease
- IS, Internal Standard
- LOD, Limit of Detection
- LOPD, Late-Onset Pompe disease
- LOQ, Limit of Quantification
- NaBH3CN, Sodium Cyanoborohydride
- PD, Pompe Disease
- QC, Quality Control
- SPE, Solid Phase Extraction
- del ex 18, c.2481+102_2646+31 del
Collapse
|
52
|
Fusco AF, McCall AL, Dhindsa JS, Zheng L, Bailey A, Kahn AF, ElMallah MK. The Respiratory Phenotype of Pompe Disease Mouse Models. Int J Mol Sci 2020; 21:ijms21062256. [PMID: 32214050 PMCID: PMC7139647 DOI: 10.3390/ijms21062256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 01/10/2023] Open
Abstract
Pompe disease is a glycogen storage disease caused by a deficiency in acid α-glucosidase (GAA), a hydrolase necessary for the degradation of lysosomal glycogen. This deficiency in GAA results in muscle and neuronal glycogen accumulation, which causes respiratory insufficiency. Pompe disease mouse models provide a means of assessing respiratory pathology and are important for pre-clinical studies of novel therapies that aim to treat respiratory dysfunction and improve quality of life. This review aims to compile and summarize existing manuscripts that characterize the respiratory phenotype of Pompe mouse models. Manuscripts included in this review were selected utilizing specific search terms and exclusion criteria. Analysis of these findings demonstrate that Pompe disease mouse models have respiratory physiological defects as well as pathologies in the diaphragm, tongue, higher-order respiratory control centers, phrenic and hypoglossal motor nuclei, phrenic and hypoglossal nerves, neuromuscular junctions, and airway smooth muscle. Overall, the culmination of these pathologies contributes to severe respiratory dysfunction, underscoring the importance of characterizing the respiratory phenotype while developing effective therapies for patients.
Collapse
|
53
|
Darios F, Stevanin G. Impairment of Lysosome Function and Autophagy in Rare Neurodegenerative Diseases. J Mol Biol 2020; 432:2714-2734. [PMID: 32145221 PMCID: PMC7232018 DOI: 10.1016/j.jmb.2020.02.033] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023]
Abstract
Rare genetic diseases affect a limited number of patients, but their etiology is often known, facilitating the development of reliable animal models and giving the opportunity to investigate physiopathology. Lysosomal storage disorders are a group of rare diseases due to primary alteration of lysosome function. These diseases are often associated with neurological symptoms, which highlighted the importance of lysosome in neurodegeneration. Likewise, other groups of rare neurodegenerative diseases also present lysosomal alteration. Lysosomes fuse with autophagosomes and endosomes to allow the degradation of their content thanks to hydrolytic enzymes. It has emerged that alteration of the autophagy–lysosome pathway could play a critical role in neuronal death in many neurodegenerative diseases. Using a repertoire of selected rare neurodegenerative diseases, we highlight that a variety of alterations of the autophagy–lysosome pathway are associated with neuronal death. Yet, in most cases, it is still unclear why alteration of this pathway can lead to neurodegeneration. Lysosome function is impaired in many rare neurodegenerative diseases, making it a convergent point for these diseases. Impaired lysosome function is associated with alteration of the autophagy pathway. Autophagy–lysosome pathway can be impaired at various steps in different rare neurodegenerative diseases. The mechanisms linking impaired autophagy–lysosome pathway to neurodegeneration are still not fully elucidated.
Collapse
Affiliation(s)
- Frédéric Darios
- Sorbonne Université, F-75013, Paris, France; Inserm, U1127, F-75013 Paris, France; CNRS, UMR 7225, F-75013 Paris, France; Institut du Cerveau et de la Moelle Epinière, ICM, F-75013 Paris, France.
| | - Giovanni Stevanin
- Sorbonne Université, F-75013, Paris, France; Inserm, U1127, F-75013 Paris, France; CNRS, UMR 7225, F-75013 Paris, France; Institut du Cerveau et de la Moelle Epinière, ICM, F-75013 Paris, France; PSL Research University, Ecole Pratique des Hautes Etudes, Laboratoire de Neurogénétique, F-75013 Paris, France
| |
Collapse
|
54
|
Neuropathophysiology of Lysosomal Storage Diseases: Synaptic Dysfunction as a Starting Point for Disease Progression. J Clin Med 2020; 9:jcm9030616. [PMID: 32106459 PMCID: PMC7141115 DOI: 10.3390/jcm9030616] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/21/2020] [Accepted: 02/21/2020] [Indexed: 12/11/2022] Open
Abstract
About two thirds of the patients affected with lysosomal storage diseases (LSD) experience neurological manifestations, such as developmental delay, seizures, or psychiatric problems. In order to develop efficient therapies, it is crucial to understand the neuropathophysiology underlying these symptoms. How exactly lysosomal storage affects biogenesis and function of neurons is still under investigation however recent research highlights a substantial role played by synaptic defects, such as alterations in synaptic spines, synaptic proteins, postsynaptic densities, and synaptic vesicles that might lead to functional impairments in synaptic transmission and neurodegeneration, finally culminating in massive neuronal death and manifestation of cognitive symptoms. Unveiling how the synaptic components are affected in neurological LSD will thus enable a better understanding of the complexity of disease progression as well as identify crucial targets of therapeutic relevance and optimal time windows for targeted intervention.
Collapse
|
55
|
Abstract
Objective To review all clinical studies and experience gained with icodextrin to date; primarily its use in peritoneal dialysis in patients with end-stage renal failure, but also its use as an intraperitoneal vehicle. Data sources Peer-reviewed original research articles in the literature; abstracts from international scientific meetings; data generated from the compassionate use programme. Study selection All published studies to date are included, some 10–20 studies being included in this review. Data extraction Data have not been specifically extracted from studies; results have been described in the context of overall experience. Results Over ten years of clinical experience with icodextrin have now been accumulated, in both continuous ambulatory peritoneal dialysis (CAPD) and automated peritoneal dialysis (APD). A small number of patients have received icodextrin for over five years, with no loss of effect. Icodextrin produces sustained ultrafiltration over long dwells while being iso-osmolar, by the process of colloid osmosis. Conclusion Icodextrin represents the first viable alternative osmotic agent to glucose, for use in solutions for peritoneal dialysis. It also has a potential use as a vehicle solution for intraperitoneal drug delivery.
Collapse
Affiliation(s)
- Elizabeth Peers
- ML Laboratories pIc, St. Albans, and Manchester RoyalInfirmary, Manchester, England
| | - Ram Gokal
- ML Laboratories pIc, St. Albans, and Manchester RoyalInfirmary, Manchester, England
| |
Collapse
|
56
|
Aung-Htut MT, Ham KA, Tchan MC, Fletcher S, Wilton SD. Novel Mutations Found in Individuals with Adult-Onset Pompe Disease. Genes (Basel) 2020; 11:genes11020135. [PMID: 32012848 PMCID: PMC7073677 DOI: 10.3390/genes11020135] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/19/2019] [Accepted: 01/23/2020] [Indexed: 11/16/2022] Open
Abstract
Pompe disease, or glycogen storage disease II is a rare, progressive disease leading to skeletal muscle weakness due to deficiency of the acid α-1,4-glucosidase enzyme (GAA). The severity of disease and observed time of onset is subject to the various combinations of heterozygous GAA alleles. Here we have characterized two novel mutations: c.2074C>T and c.1910_1918del, and a previously reported c.1082C>G mutation of uncertain clinical significance. These mutations were found in three unrelated patients with adult-onset Pompe disease carrying the common c.-32-13T>G mutation. The c.2074 C>T nonsense mutation has obvious consequences on GAA expression but the c.1910_1918del (deletion of 3 amino acids) and c.1082C>G missense variants are more subtle DNA changes with catastrophic consequences on GAA activity. Molecular and clinical analyses from the three patients corresponded with the anticipated pathogenicity of each mutation.
Collapse
Affiliation(s)
- May T. Aung-Htut
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia; (M.T.A.-H.); (K.A.H.); (S.F.)
- Perron Institute for Neurological and Translational Science and The University of Western Australia, Perth 6009, Australia
| | - Kristin A. Ham
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia; (M.T.A.-H.); (K.A.H.); (S.F.)
| | - Michel C. Tchan
- Genetic Medicine, Westmead Hospital, Sydney 2145, Australia;
- Sydney Medical School, The University of Sydney, Sydney 2006, Australia
| | - Sue Fletcher
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia; (M.T.A.-H.); (K.A.H.); (S.F.)
| | - Steve D. Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia; (M.T.A.-H.); (K.A.H.); (S.F.)
- Perron Institute for Neurological and Translational Science and The University of Western Australia, Perth 6009, Australia
- Correspondence:
| |
Collapse
|
57
|
d'Azzo A, Annunziata I. Transcription factor competition regulates lysosomal biogenesis and autophagy. Mol Cell Oncol 2020; 7:1685840. [PMID: 32158913 PMCID: PMC7051135 DOI: 10.1080/23723556.2019.1685840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 10/01/2019] [Accepted: 10/23/2019] [Indexed: 11/10/2022]
Abstract
“In the field of observation, chance favours only the prepared mind” (Louis Pasteur). This motto seems to have guided our unexpected results published recently in Nature Communications, where we describe an epigenetic rheostat that regulates expression of the constituents of the lysosomal and autophagic systems.
Collapse
Affiliation(s)
- Alessandra d'Azzo
- Department of Genetics, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ida Annunziata
- Department of Genetics, St Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
58
|
Farah BL, Yen PM, Koeberl DD. Links between autophagy and disorders of glycogen metabolism - Perspectives on pathogenesis and possible treatments. Mol Genet Metab 2020; 129:3-12. [PMID: 31787497 PMCID: PMC7836271 DOI: 10.1016/j.ymgme.2019.11.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 01/17/2023]
Abstract
The glycogen storage diseases are a group of inherited metabolic disorders that are characterized by specific enzymatic defects involving the synthesis or degradation of glycogen. Each disorder presents with a set of symptoms that are due to the underlying enzyme deficiency and the particular tissues that are affected. Autophagy is a process by which cells degrade and recycle unneeded or damaged intracellular components such as lipids, glycogen, and damaged mitochondria. Recent studies showed that several of the glycogen storage disorders have abnormal autophagy which can disturb normal cellular metabolism and/or mitochondrial function. Here, we provide a clinical overview of the glycogen storage disorders, a brief description of autophagy, and the known links between specific glycogen storage disorders and autophagy.
Collapse
Affiliation(s)
- Benjamin L Farah
- Department of Pathology, Singapore General Hospital, Singapore, Singapore.
| | - Paul M Yen
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Dwight D Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, NC, USA; Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA..
| |
Collapse
|
59
|
Yoshida T, Jonouchi T, Osafune K, Takita J, Sakurai H. A Liver Model of Infantile-Onset Pompe Disease Using Patient-Specific Induced Pluripotent Stem Cells. Front Cell Dev Biol 2019; 7:316. [PMID: 31850350 PMCID: PMC6895003 DOI: 10.3389/fcell.2019.00316] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/18/2019] [Indexed: 11/13/2022] Open
Abstract
Infantile-onset Pompe disease (IOPD) is a life-threatening multi-organ disease caused by an inborn defect of lysosomal acid α-glucosidase (GAA), which can degrade glycogen into glucose. Lack of GAA causes abnormal accumulation of glycogen in the lysosomes, particularly in the skeletal muscle, liver, and heart. Enzyme replacement therapy (ERT) with recombinant human GAA (rhGAA) is the only available treatment; however, its effect varies by organ. Thus, to fully understand the pathomechanism of IOPD, organ-specific disease models are necessary. We previously generated induced pluripotent stem cells (iPSCs) from three unrelated patients with IOPD and establish a skeletal muscle model of IOPD. Here, we used the same iPSC lines as the previous study and differentiated them into hepatocytes. As a result, hepatocytes differentiated from iPSC of IOPD patients showed abnormal accumulation of lysosomal glycogen, the hallmark of Pompe disease. Using this model, we also demonstrated that glycogen accumulation was dose-dependently restored by rhGAA treatment. In conclusion, we have successfully established an in vitro liver model of IOPD using patient-specific iPSCs. This model can be a platform to elucidate the underlying disease mechanism or to be applied to drug-screening. Moreover, our study also suggest that an iPSC-based approach is suitable for modeling of diseases that affect multiple organs like Pompe disease.
Collapse
Affiliation(s)
- Takeshi Yoshida
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tatsuya Jonouchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Junko Takita
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
60
|
Esmail S, Danter WR. DeepNEU: Artificially Induced Stem Cell (aiPSC) and Differentiated Skeletal Muscle Cell (aiSkMC) Simulations of Infantile Onset POMPE Disease (IOPD) for Potential Biomarker Identification and Drug Discovery. Front Cell Dev Biol 2019; 7:325. [PMID: 31867331 PMCID: PMC6909925 DOI: 10.3389/fcell.2019.00325] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/25/2019] [Indexed: 12/17/2022] Open
Abstract
Infantile onset Pompe disease (IOPD) is a rare and lethal genetic disorder caused by the deletion of the acid alpha-glucosidase (GAA) gene. This gene encodes an essential lysosomal enzyme that converts glycogen to glucose. While enzyme replacement therapy helps some, our understanding of disease pathophysiology is limited. In this project we develop computer simulated stem cells (aiPSC) and differentiated skeletal muscle cells (aiSkMC) to empower IOPD research and drug discovery. Our Artificial Intelligence (AI) platform, DeepNEU v3.6 was used to generate aiPSC and aiSkMC simulations with and without GAA expression. These simulations were validated using peer reviewed results from the recent literature. Once the aiSkMC simulations (IOPD and WT) were validated they were used to evaluate calcium homeostasis and mitochondrial function in IOPD. Lastly, we used aiSkMC IOPD simulations to identify known and novel biomarkers and potential therapeutic targets. The aiSkMC simulations of IOPD correctly predicted genotypic and phenotypic features that were reported in recent literature. The probability that these features were accurately predicted by chance alone using the binomial test is 0.0025. The aiSkMC IOPD simulation correctly identified L-type calcium channels (VDCC) as a biomarker and confirmed the positive effects of calcium channel blockade (CCB) on calcium homeostasis and mitochondrial function. These published data were extended by the aiSkMC simulations to identify calpain(s) as a novel potential biomarker and therapeutic target for IOPD. This is the first time that computer simulations of iPSC and differentiated skeletal muscle cells have been used to study IOPD. The simulations are robust and accurate based on available published literature. We also demonstrated that the IOPD simulations can be used for potential biomarker identification leading to targeted drug discovery. We will continue to explore the potential for calpain inhibitors with and without CCB as effective therapy for IOPD.
Collapse
|
61
|
Respiratory complications of metabolic disease in the paediatric population: A review of presentation, diagnosis and therapeutic options. Paediatr Respir Rev 2019; 32:55-65. [PMID: 31101546 DOI: 10.1016/j.prrv.2019.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 12/21/2022]
Abstract
Inborn errors of metabolism (IEMs) whilst individually rare, as a group constitute a field which is increasingly demands on pulmonologists. With the advent of new therapies such as enzyme replacement and gene therapy, early diagnosis and treatment of these conditions can impact on long term outcome, making their timely recognition and appropriate investigation increasingly important. Conversely, with improved treatment, survival of these patients is increasing, with the emergence of previously unknown respiratory phenotypes. It is thus important that pulmonologists are aware of and appropriately monitor and manage these complications. This review aims to highlight the respiratory manifestations which can occur. It isdivided into conditions resulting primarily in obstructive airway and lung disease, restrictive lung disease such as interstitial lung disease or pulmonary alveolar proteinosis and pulmonary hypertension, whilst acknowledging that some diseases have the potential to cause all three. The review focuses on general phenotypes of IEMs, their known respiratory complications and the basic metabolic investigations which should be performed where an IEM is suspected.
Collapse
|
62
|
Lagler FB, Moder A, Rohrbach M, Hennermann J, Mengel E, Gökce S, Hundsberger T, Rösler KM, Karabul N, Huemer M. Extent, impact, and predictors of diagnostic delay in Pompe disease: A combined survey approach to unveil the diagnostic odyssey. JIMD Rep 2019; 49:89-95. [PMID: 31497486 PMCID: PMC6718115 DOI: 10.1002/jmd2.12062] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Early diagnosis is of substantial benefit for patients with Pompe disease. Yet underdiagnosing and substantial diagnostic delay are still frequent and the determinants of this are unknown. This study is the first to systematically investigate the diagnostic odyssey in Pompe disease from patients', parents', and physicians' perspectives. METHODS Patients with infantile or late onset Pompe disease, their parents as well as their metabolic experts were invited to fill in respective surveys. The survey addressed perceived disease symptoms at onset and during the course of the disease, specialties of involved physicians, activities of patient-initiated search for diagnosis and the perceived impact of time to diagnosis on outcome. Results of experts' and patients'/parents' surveys were compared and expressed by descriptive statistics. RESULTS AND DISCUSSION We collected data on 15 males and 17 females including 9 infantile and 23 late onset Pompe patients. All received the correct diagnosis at a metabolic or musculoskeletal expert center. Patients with direct referral to the expert center had the lowest diagnostic delay, while patients who were seen by several physicians, received the correct diagnosis after 44%-200% longer delay. The proportion of direct referral varied strongly between pediatricians (57%) and other disciplines (18%-36%). CONCLUSION Our study highlights a substantially larger diagnostic delay in Pompe patients that are not directly referred to expert centers for diagnostic work. Our findings may be used to develop more successful strategies for early diagnosis. SYNOPSIS Diagnostic delay in Pompe disease is substantial particularly in patients that are not directly referred to expert centers for diagnostic workup, so facilitating direct referral may be a new strategy for early diagnosis.
Collapse
Affiliation(s)
- Florian B. Lagler
- Institute for Inborn Errors of Metabolism, Paracelsus Medical UniversitySalzburgAustria
- Department of PaediatricsParacelsus Medical UniversitySalzburgAustria
| | - Angelika Moder
- Institute for Inborn Errors of Metabolism, Paracelsus Medical UniversitySalzburgAustria
| | - Marianne Rohrbach
- Division of Metabolism and Children's Research CentreUniversity Children's HospitalZurichSwitzerland
| | - Julia Hennermann
- Division of Metabolic Diseases (Villa Metabolica)Center for Diseases in Childhood and Adolescence, Mainz Medical UniversityMainzGermany
| | - Eugen Mengel
- Division of Metabolic Diseases (Villa Metabolica)Center for Diseases in Childhood and Adolescence, Mainz Medical UniversityMainzGermany
| | - Seyfullah Gökce
- Division of Metabolic Diseases (Villa Metabolica)Center for Diseases in Childhood and Adolescence, Mainz Medical UniversityMainzGermany
| | | | - Kai M. Rösler
- Department of Neurology, InselspitalUniversity HospitalBernSwitzerland
| | - Nesrin Karabul
- Center of Endocrinology and Metabolism, Rheumatology and NeurologyEndokrinologikum FrankfurtFrankfurt a. M.Germany
| | - Martina Huemer
- Division of Metabolism and Children's Research CentreUniversity Children's HospitalZurichSwitzerland
- Radiz – Rare Disease Initiative Zürich, Clinical Research Priority ProgramUniversity of ZürichZürichSwitzerland
- Department of PaediatricsLandeskrankenhausBregenzAustria
| |
Collapse
|
63
|
Do HV, Khanna R, Gotschall R. Challenges in treating Pompe disease: an industry perspective. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:291. [PMID: 31392203 DOI: 10.21037/atm.2019.04.15] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pompe disease is a rare inherited metabolic disorder of defective lysosomal glycogen catabolism due to a deficiency in acid alpha-glucosidase (GAA). Alglucosidase alfa enzyme replacement therapy (ERT) using recombinant human GAA (rhGAA ERT) is the only approved treatment for Pompe disease. Alglucosidase alfa has provided irrefutable clinical benefits, but has not been an optimal treatment primarily due to poor drug targeting of ERT to skeletal muscles. Several critical factors contribute to this inefficiency. Some are inherent to the anatomy of the body that cannot be altered, while others may be addressed with better drug design and engineering. The knowledge gained from alglucosidase alfa ERT over the past 2 decades has allowed us to better understand the challenges that hinder its effectiveness. In this review, we detail the problems which must be overcome for improving drug targeting and clinical efficacy. These same issues may also impact therapeutic enzymes derived from gene therapies, and thus, have important implications for the development of next generation therapies for Pompe.
Collapse
Affiliation(s)
- Hung V Do
- Amicus Therapeutics, Inc., Cranbury, NJ, USA
| | | | | |
Collapse
|
64
|
McCall AL, Stankov SG, Cowen G, Cloutier D, Zhang Z, Yang L, Clement N, Falk DJ, Byrne BJ. Reduction of Autophagic Accumulation in Pompe Disease Mouse Model Following Gene Therapy. Curr Gene Ther 2019; 19:197-207. [PMID: 31223086 DOI: 10.2174/1566523219666190621113807] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/31/2019] [Accepted: 06/11/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Pompe disease is a fatal neuromuscular disorder caused by a deficiency in acid α-glucosidase, an enzyme responsible for glycogen degradation in the lysosome. Currently, the only approved treatment for Pompe disease is enzyme replacement therapy (ERT), which increases patient survival, but does not fully correct the skeletal muscle pathology. Skeletal muscle pathology is not corrected with ERT because low cation-independent mannose-6-phosphate receptor abundance and autophagic accumulation inhibits the enzyme from reaching the lysosome. Thus, a therapy that more efficiently targets skeletal muscle pathology, such as adeno-associated virus (AAV), is needed for Pompe disease. OBJECTIVE The goal of this project was to deliver a rAAV9-coGAA vector driven by a tissue restrictive promoter will efficiently transduce skeletal muscle and correct autophagic accumulation. METHODS Thus, rAAV9-coGAA was intravenously delivered at three doses to 12-week old Gaa-/- mice. 1 month after injection, skeletal muscles were biochemically and histologically analyzed for autophagy-related markers. RESULTS At the highest dose, GAA enzyme activity and vacuolization scores achieved therapeutic levels. In addition, resolution of autophagosome (AP) accumulation was seen by immunofluorescence and western blot analysis of autophagy-related proteins. Finally, mice treated at birth demonstrated persistence of GAA expression and resolution of lysosomes and APs compared to those treated at 3 months. CONCLUSION In conclusion, a single systemic injection of rAAV9-coGAA ameliorates vacuolar accumulation and prevents autophagic dysregulation.
Collapse
Affiliation(s)
- Angela L McCall
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Sylvia G Stankov
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Gabrielle Cowen
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Denise Cloutier
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Zizhao Zhang
- Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL, United States
| | - Lin Yang
- Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL, United States
| | - Nathalie Clement
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Darin J Falk
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Barry J Byrne
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
65
|
Brewer MK, Gentry MS. Brain Glycogen Structure and Its Associated Proteins: Past, Present and Future. ADVANCES IN NEUROBIOLOGY 2019; 23:17-81. [PMID: 31667805 PMCID: PMC7239500 DOI: 10.1007/978-3-030-27480-1_2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This chapter reviews the history of glycogen-related research and discusses in detail the structure, regulation, chemical properties and subcellular distribution of glycogen and its associated proteins, with particular focus on these aspects in brain tissue.
Collapse
Affiliation(s)
- M Kathryn Brewer
- Department of Molecular and Cellular Biochemistry, Epilepsy and Brain Metabolism Center, Lafora Epilepsy Cure Initiative, and Center for Structural Biology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Matthew S Gentry
- Department of Molecular and Cellular Biochemistry, Epilepsy and Brain Metabolism Center, Lafora Epilepsy Cure Initiative, and Center for Structural Biology, University of Kentucky College of Medicine, Lexington, KY, USA.
| |
Collapse
|
66
|
Ghanbari F, Khaksari M, Vaezi G, Hojati V, Shiravi A. Hydrogen Sulfide Protects Hippocampal Neurons Against Methamphetamine Neurotoxicity Via Inhibition of Apoptosis and Neuroinflammation. J Mol Neurosci 2018; 67:133-141. [DOI: 10.1007/s12031-018-1218-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 11/11/2018] [Indexed: 12/28/2022]
|
67
|
Lagalice L, Pichon J, Gougeon E, Soussi S, Deniaud J, Ledevin M, Maurier V, Leroux I, Durand S, Ciron C, Franzoso F, Dubreil L, Larcher T, Rouger K, Colle MA. Satellite cells fail to contribute to muscle repair but are functional in Pompe disease (glycogenosis type II). Acta Neuropathol Commun 2018; 6:116. [PMID: 30382921 PMCID: PMC6211565 DOI: 10.1186/s40478-018-0609-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 09/29/2018] [Indexed: 12/26/2022] Open
Abstract
Pompe disease, which is due to acid alpha-glucosidase deficiency, is characterized by skeletal muscle dysfunction attributed to the accumulation of glycogen-filled lysosomes and autophagic buildup. Despite the extensive tissue damages, a failure of satellite cell (SC) activation and lack of muscle regeneration have been reported in patients. However, the origin of this defective program is unknown. Additionally, whether these deficits occur gradually over the disease course is unclear. Using a longitudinal pathophysiological study of two muscles in a Pompe mouse model, here, we report that the enzymatic defect results in a premature saturating glycogen overload and a high number of enlarged lysosomes. The muscles gradually display profound remodeling as the number of autophagic vesicles, centronucleated fibers, and split fibers increases and larger fibers are lost. Only a few regenerated fibers were observed regardless of age, although the SC pool was preserved. Except for the early age, during which higher numbers of activated SCs and myoblasts were observed, no myogenic commitment was observed in response to the damage. Following in vivo injury, we established that muscle retains regenerative potential, demonstrating that the failure of SC participation in repair is related to an activation signal defect. Altogether, our findings provide new insight into the pathophysiology of Pompe disease and highlight that the activation signal defect of SCs compromises muscle repair, which could be related to the abnormal energetic supply following autophagic flux impairment.
Collapse
Affiliation(s)
- Lydie Lagalice
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Julien Pichon
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
- INSERM UMR1089, Université de Nantes, Centre Hospitalier Universitaire, Nantes, France
| | - Eliot Gougeon
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Salwa Soussi
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Johan Deniaud
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Mireille Ledevin
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Virginie Maurier
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Isabelle Leroux
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Sylvie Durand
- BIA, INRA, Centre INRA Pays de la Loire, Nantes, F-44300 France
| | - Carine Ciron
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Francesca Franzoso
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Laurence Dubreil
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Thibaut Larcher
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Karl Rouger
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| | - Marie-Anne Colle
- PAnTher, INRA, École Nationale Vétérinaire, Agro-alimentaire et de l’alimentation Nantes-Atlantique (Oniris), Université Bretagne Loire (UBL), Nantes, F-44307 France
| |
Collapse
|
68
|
Abstract
Pompe disease is a rare and deadly muscle disorder. As a clinical entity, the disease has been known for over 75 years. While an optimist might be excited about the advances made during this time, a pessimist would note that we have yet to find a cure. However, both sides would agree that many findings in basic science-such as the Nobel prize-winning discoveries of glycogen metabolism, the lysosome, and autophagy-have become the foundation of our understanding of Pompe disease. The disease is a glycogen storage disorder, a lysosomal disorder, and an autophagic myopathy. In this review, we will discuss how these past discoveries have guided Pompe research and impacted recent therapeutic developments.
Collapse
Affiliation(s)
- Lara Kohler
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rosa Puertollano
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Nina Raben
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
69
|
Shafahi M, Vaezi G, Shajiee H, Sharafi S, Khaksari M. Crocin Inhibits Apoptosis and Astrogliosis of Hippocampus Neurons Against Methamphetamine Neurotoxicity via Antioxidant and Anti-inflammatory Mechanisms. Neurochem Res 2018; 43:2252-2259. [PMID: 30259275 DOI: 10.1007/s11064-018-2644-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/11/2018] [Accepted: 09/19/2018] [Indexed: 01/19/2023]
Abstract
Methamphetamine (METH) is a stimulant drug, which can cause neurotoxicity and increase the risk of neurodegenerative disorders. The mechanisms of acute METH intoxication comprise intra-neuronal events including oxidative stress, dopamine oxidation, and excitotoxicity. According to recent studies, crocin protects neurons by functioning as an anti-oxidant, anti-inflammatory, and anti-apoptotic compound. Accordingly, this study aimed to determine if crocin can protect against METH-induced neurotoxicity. Seventy-two male Wistar rats that weighed 260-300 g were randomly allocated to six groups of control (n = 12), crocin 90 mg/kg group (n = 12), METH (n = 12), METH + crocin 30 mg/kg (n = 12), METH + crocin 60 mg/kg (n = 12), and METH + crocin 90 mg/kg (n = 12). METH neurotoxicity was induced by 40 mg/kg of METH in four injections (e.g., 4 × 10 mg/kg q. 2 h, IP). Crocin was intraperitoneally (IP) injected at 30 min, 24 h, and 48 h after the final injection of METH. Seven days after METH injection, the rats' brains were removed for biochemical assessment using the ELISA technique, and immunohistochemistry staining was used for caspase-3 and glial fibrillary acidic protein (GFAP) detection. Crocin treatment could significantly increase superoxide dismutase (P < 0.05) and glutathione (P < 0.01) levels and reduce malondialdehyde and TNF-α in comparison with the METH group (P < 0.05). Moreover, crocin could significantly decline the level of caspase-3 and GFAP-positive cells in the CA1 region (P < 0.01). According to the results, crocin exerts neuroprotective effects on METH neurotoxicity via the inhibition of apoptosis and neuroinflammation.
Collapse
Affiliation(s)
- Monire Shafahi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Golamhassan Vaezi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Hooman Shajiee
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Shahram Sharafi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Mehdi Khaksari
- Addiction Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.
| |
Collapse
|
70
|
Cerebellar Ataxia in Children: A Clinical and MRI Approach to the Differential Diagnosis. Top Magn Reson Imaging 2018; 27:275-302. [PMID: 30086112 DOI: 10.1097/rmr.0000000000000175] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
: The cerebellum has long been recognized as a fundamental structure in motor coordination. Structural cerebellar abnormalities and diseases involving the cerebellum are relatively common in children. The not always specific clinical presentation of ataxia, incoordination, and balance impairment can often be a challenge to attain a precise diagnosis. Continuous advances in genetic research and moreover the constant development in neuroimaging modalities, particularly in the field of magnetic resonance imaging, have promoted a better understanding of cerebellar diseases and led to several modifications in their classification in recent years. Thorough clinical and neuroimaging investigation is recommended for proper diagnosis. This review outlines an update of causes of cerebellar disorders that present clinically with ataxia in the pediatric population. These conditions were classified in 2 major groups, namely genetic malformations and acquired or disruptive disorders recognizable by neuroimaging and subsequently according to their features during the prenatal and postnatal periods.
Collapse
|
71
|
Enzymatic replacement therapy in patients with late-onset Pompe disease – 6-Year follow up. Neurol Neurochir Pol 2018; 52:465-469. [DOI: 10.1016/j.pjnns.2018.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/29/2018] [Accepted: 05/02/2018] [Indexed: 11/20/2022]
|
72
|
Corti M, Liberati C, Smith BK, Lawson LA, Tuna IS, Conlon TJ, Coleman KE, Islam S, Herzog RW, Fuller DD, Collins SW, Byrne BJ. Safety of Intradiaphragmatic Delivery of Adeno-Associated Virus-Mediated Alpha-Glucosidase (rAAV1-CMV-hGAA) Gene Therapy in Children Affected by Pompe Disease. HUM GENE THER CL DEV 2018; 28:208-218. [PMID: 29160099 DOI: 10.1089/humc.2017.146] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A first-in-human trial of diaphragmatic gene therapy (AAV1-CMV-GAA) to treat respiratory and neural dysfunction in early-onset Pompe disease was conducted. The primary objective of this study was to assess the safety of rAAV1-CMV-hGAA vector delivered to the diaphragm muscle of Pompe disease subjects with ventilatory insufficiency. Safety was assessed by measurement of change in serum chemistries and hematology, urinalysis, and immune response to GAA and AAV, as well as change in level of health. The data demonstrate that the AAV treatment was safe and there were no adverse events related to the study agent. Adverse events related to the study procedure were observed in subjects with lower baseline neuromuscular function. All adverse events were resolved before the end of the study, except for one severe adverse event determined not to be related to either the study agent or the study procedure. In addition, an anti-capsid and anti-transgene antibody response was observed in all subjects who received rAAV1-CMV-hGAA, except for subjects who received concomitant immunomodulation to manage reaction to enzyme replacement therapy, as per their standard of care. This observation is significant for future gene therapy studies and serves to establish a clinically relevant approach to blocking immune responses to both the AAV capsid protein and transgene product.
Collapse
Affiliation(s)
- Manuela Corti
- 1 Department of Pediatrics, College of Medicine, University of Florida , Gainesville, Florida
| | - Cristina Liberati
- 1 Department of Pediatrics, College of Medicine, University of Florida , Gainesville, Florida
| | - Barbara K Smith
- 2 Department of Physical Therapy, College of Public Health and Health Profession, University of Florida , Gainesville, Florida
| | - Lee Ann Lawson
- 3 Department of Endocrinology, College of Medicine, University of Florida , Gainesville, Florida
| | - Ibrahim S Tuna
- 4 Department of Radiology, College of Medicine, University of Florida , Gainesville, Florida
| | - Thomas J Conlon
- 1 Department of Pediatrics, College of Medicine, University of Florida , Gainesville, Florida
| | - Kirsten E Coleman
- 1 Department of Pediatrics, College of Medicine, University of Florida , Gainesville, Florida
| | - Saleem Islam
- 1 Department of Pediatrics, College of Medicine, University of Florida , Gainesville, Florida
| | - Roland W Herzog
- 1 Department of Pediatrics, College of Medicine, University of Florida , Gainesville, Florida
| | - David D Fuller
- 2 Department of Physical Therapy, College of Public Health and Health Profession, University of Florida , Gainesville, Florida
| | - Shelley W Collins
- 1 Department of Pediatrics, College of Medicine, University of Florida , Gainesville, Florida
| | - Barry J Byrne
- 1 Department of Pediatrics, College of Medicine, University of Florida , Gainesville, Florida
| |
Collapse
|
73
|
Savarese M, Torella A, Musumeci O, Angelini C, Astrea G, Bello L, Bruno C, Comi GP, Di Fruscio G, Piluso G, Di Iorio G, Ergoli M, Esposito G, Fanin M, Farina O, Fiorillo C, Garofalo A, Giugliano T, Magri F, Minetti C, Moggio M, Passamano L, Pegoraro E, Picillo E, Sampaolo S, Santorelli FM, Semplicini C, Udd B, Toscano A, Politano L, Nigro V. Targeted gene panel screening is an effective tool to identify undiagnosed late onset Pompe disease. Neuromuscul Disord 2018; 28:586-591. [DOI: 10.1016/j.nmd.2018.03.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/12/2018] [Accepted: 03/29/2018] [Indexed: 10/17/2022]
|
74
|
Abstract
One of the fundamental properties of the cell is the capability to digest and remodel its own components according to metabolic and developmental needs. This is accomplished via the autophagy-lysosome system, a pathway of critical importance in the brain, where it contributes to neuronal plasticity and must protect nonreplaceable neurons from the potentially harmful accumulation of cellular waste. The study of lysosomal biogenesis and function in the context of common and rare neurodegenerative diseases has revealed that a dysfunctional autophagy-lysosome system is the shared nexus where multiple, interconnected pathogenic events take place. The characterization of pathways and mechanisms regulating the lysosomal system and autophagic clearance offers unprecedented opportunities for the development of polyvalent therapeutic strategies based on the enhancement of the autophagy-lysosome pathway to maintain cellular homeostasis and achieve neuroprotection.
Collapse
Affiliation(s)
- Jaiprakash Sharma
- Department of Molecular and Human Genetics, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA;
| | - Alberto di Ronza
- Department of Molecular and Human Genetics, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA;
| | - Parisa Lotfi
- Department of Molecular and Human Genetics, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA;
| | - Marco Sardiello
- Department of Molecular and Human Genetics, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA;
| |
Collapse
|
75
|
Schneider JL, Dingman RK, Balu-Iyer SV. Lipidic Nanoparticles Comprising Phosphatidylinositol Mitigate Immunogenicity and Improve Efficacy of Recombinant Human Acid Alpha-Glucosidase in a Murine Model of Pompe Disease. J Pharm Sci 2018; 107:831-837. [PMID: 29102549 PMCID: PMC5812781 DOI: 10.1016/j.xphs.2017.10.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 11/25/2022]
Abstract
Enzyme replacement therapy with recombinant human acid α-glucosidase (rhGAA) is complicated by the formation of anti-rhGAA antibodies, a short circulating half-life, instability in the plasma, and limited uptake into target tissue. Previously, we have demonstrated that phosphatidylinositol (PI) containing liposomes can reduce the immunogenicity and extend plasma survival of factor VIII (FVIII) in a mouse model of hemophilia A. In this article, we investigate the ability of PI liposomes to be used as a delivery vehicle to overcome the issues that complicate therapy with rhGAA. In a murine model of Pompe disease, administration of PI-rhGAA mitigated the immunogenicity of rhGAA, resulting in a significantly lower formation of anti-rhGAA antibodies. PI-rhGAA also showed minimal improvements to the pharmacokinetic parameters and efficacy measures compared to free rhGAA. Overall, these data suggest that PI-rhGAA may have the potential to be a useful therapeutic option for improving the treatment of Pompe disease.
Collapse
Affiliation(s)
- Jennifer L Schneider
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214
| | - Robert K Dingman
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214
| | - Sathy V Balu-Iyer
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214.
| |
Collapse
|
76
|
Keeling E, Lotery AJ, Tumbarello DA, Ratnayaka JA. Impaired Cargo Clearance in the Retinal Pigment Epithelium (RPE) Underlies Irreversible Blinding Diseases. Cells 2018; 7:E16. [PMID: 29473871 PMCID: PMC5850104 DOI: 10.3390/cells7020016] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 02/20/2018] [Accepted: 02/22/2018] [Indexed: 01/09/2023] Open
Abstract
Chronic degeneration of the Retinal Pigment Epithelium (RPE) is a precursor to pathological changes in the outer retina. The RPE monolayer, which lies beneath the neuroretina, daily internalises and digests large volumes of spent photoreceptor outer segments. Impaired cargo handling and processing in the endocytic/phagosome and autophagy pathways lead to the accumulation of lipofuscin and pyridinium bis-retinoid A2E aggregates and chemically modified compounds such as malondialdehyde and 4-hydroxynonenal within RPE. These contribute to increased proteolytic and oxidative stress, resulting in irreversible damage to post-mitotic RPE cells and development of blinding conditions such as age-related macular degeneration, Stargardt disease and choroideremia. Here, we review how impaired cargo handling in the RPE results in their dysfunction, discuss new findings from our laboratory and consider how newly discovered roles for lysosomes and the autophagy pathway could provide insights into retinopathies. Studies of these dynamic, molecular events have also been spurred on by recent advances in optics and imaging technology. Mechanisms underpinning lysosomal impairment in other degenerative conditions including storage disorders, α-synuclein pathologies and Alzheimer's disease are also discussed. Collectively, these findings help transcend conventional understanding of these intracellular compartments as simple waste disposal bags to bring about a paradigm shift in the way lysosomes are perceived.
Collapse
Affiliation(s)
- Eloise Keeling
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK.
| | - Andrew J Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK.
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK.
| | - David A Tumbarello
- Biological Sciences, Faculty of Natural & Environmental Sciences, Life Science Building 85, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK.
| | - J Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK.
| |
Collapse
|
77
|
McCall AL, Salemi J, Bhanap P, Strickland LM, Elmallah MK. The impact of Pompe disease on smooth muscle: a review. J Smooth Muscle Res 2018; 54:100-118. [PMID: 30787211 PMCID: PMC6380904 DOI: 10.1540/jsmr.54.100] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 12/26/2018] [Indexed: 12/24/2022] Open
Abstract
Pompe disease (OMIM 232300) is an autosomal recessive disorder caused by mutations in the gene encoding acid α-glucosidase (GAA) (EC 3.2.1.20), the enzyme responsible for hydrolyzing lysosomal glycogen. The primary cellular pathology is lysosomal glycogen accumulation in cardiac muscle, skeletal muscle, and motor neurons, which ultimately results in cardiorespiratory failure. However, the severity of pathology and its impact on clinical outcomes are poorly described in smooth muscle. The advent of enzyme replacement therapy (ERT) in 2006 has improved clinical outcomes in infantile-onset Pompe disease patients. Although ERT increases patient life expectancy and ventilator free survival, it is not entirely curative. Persistent motor neuron pathology and weakness of respiratory muscles, including airway smooth muscles, contribute to the need for mechanical ventilation by some patients on ERT. Some patients on ERT continue to experience life-threatening pathology to vascular smooth muscle, such as aneurysms or dissections within the aorta and cerebral arteries. Better characterization of the disease impact on smooth muscle will inform treatment development and help anticipate later complications. This review summarizes the published knowledge of smooth muscle pathology associated with Pompe disease in animal models and in patients.
Collapse
Affiliation(s)
- Angela L McCall
- Department of Pediatrics, School of Medicine, Duke University, Durham, NC, USA
| | - Jeffrey Salemi
- University of Massachusetts Medical School, Worcester, MA, USA
| | - Preeti Bhanap
- Department of Pediatrics, School of Medicine, Duke University, Durham, NC, USA
| | - Laura M Strickland
- Department of Pediatrics, School of Medicine, Duke University, Durham, NC, USA
| | - Mai K Elmallah
- Department of Pediatrics, School of Medicine, Duke University, Durham, NC, USA
| |
Collapse
|
78
|
Ramrakhiani N, Agarwal K, Bansal A. Walking with the ventilator: A rare case of Pompe's disease with a review of literature. Neurol India 2018; 66:545-547. [DOI: 10.4103/0028-3886.227264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
79
|
Nelson BC, Hashem SI, Adler ED. Human-Induced Pluripotent Stem Cell-Based Modeling of Cardiac Storage Disorders. Curr Cardiol Rep 2017; 19:26. [PMID: 28251514 DOI: 10.1007/s11886-017-0829-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW The aim of this study is to review the published human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) models of cardiac storage disorders and to evaluate the limitations and future applications of this technology. RECENT FINDINGS Several cardiac storage disorders (CSDs) have been modeled using patient-specific hiPSC-CMs, including Anderson-Fabry disease, Danon disease, and Pompe disease. These models have shown that patient-specific hiPSC-CMs faithfully recapitulate key phenotypic features of CSDs and respond predictably to pharmacologic manipulation. hiPSC-CMs generated from patients with CSDs are representative models of the patient disease state and can be used as an in vitro system for the study of human cardiomyocytes. While these models suffer from several limitations, they are likely to play an important role in future mechanistic studies of cardiac storage disorders and the development of targeted therapeutics for these diseases.
Collapse
Affiliation(s)
- Bradley C Nelson
- Department of Medicine, Division of Cardiology, University of California San Diego, 9500 Gilman Drive, Biomedical Research Facility, Room 1217 AA, La Jolla, CA, 92093, USA
| | - Sherin I Hashem
- Department of Medicine, Division of Cardiology, University of California San Diego, 9500 Gilman Drive, Biomedical Research Facility, Room 1217 AA, La Jolla, CA, 92093, USA
| | - Eric D Adler
- Department of Medicine, Division of Cardiology, University of California San Diego, 9500 Gilman Drive, Biomedical Research Facility, Room 1217 AA, La Jolla, CA, 92093, USA.
| |
Collapse
|
80
|
Platt FM. Emptying the stores: lysosomal diseases and therapeutic strategies. Nat Rev Drug Discov 2017; 17:133-150. [PMID: 29147032 DOI: 10.1038/nrd.2017.214] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lysosomal storage disorders (LSDs) - designated as 'orphan' diseases - are inborn errors of metabolism caused by defects in genes that encode proteins involved in various aspects of lysosomal homeostasis. For many years, LSDs were viewed as unattractive targets for the development of therapies owing to their low prevalence. However, the development and success of the first commercial biologic therapy for an LSD - enzyme replacement therapy for type 1 Gaucher disease - coupled with regulatory incentives rapidly catalysed commercial interest in therapeutically targeting LSDs. Despite ongoing challenges, various therapeutic strategies for LSDs now exist, with many agents approved, undergoing clinical trials or in preclinical development.
Collapse
Affiliation(s)
- Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| |
Collapse
|
81
|
Giacomelli E, Mummery CL, Bellin M. Human heart disease: lessons from human pluripotent stem cell-derived cardiomyocytes. Cell Mol Life Sci 2017; 74:3711-3739. [PMID: 28573431 PMCID: PMC5597692 DOI: 10.1007/s00018-017-2546-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 05/09/2017] [Accepted: 05/23/2017] [Indexed: 02/07/2023]
Abstract
Technical advances in generating and phenotyping cardiomyocytes from human pluripotent stem cells (hPSC-CMs) are now driving their wider acceptance as in vitro models to understand human heart disease and discover therapeutic targets that may lead to new compounds for clinical use. Current literature clearly shows that hPSC-CMs recapitulate many molecular, cellular, and functional aspects of human heart pathophysiology and their responses to cardioactive drugs. Here, we provide a comprehensive overview of hPSC-CMs models that have been described to date and highlight their most recent and remarkable contributions to research on cardiovascular diseases and disorders with cardiac traits. We conclude discussing immediate challenges, limitations, and emerging solutions.
Collapse
Affiliation(s)
- E Giacomelli
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - C L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, Building Zuidhorst, 7500 AE, Enschede, The Netherlands
| | - M Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands.
| |
Collapse
|
82
|
Tehrani KHN, Sakhaeyan E, Sakhaeyan E. Evaluation prevalence of Pompe disease in Iranian patients with myopathies of unknown etiology. Electron Physician 2017; 9:4886-4889. [PMID: 28894550 PMCID: PMC5587008 DOI: 10.19082/4886] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 09/26/2016] [Indexed: 11/23/2022] Open
Abstract
Background Pompe disease is a rare but potentially treatable metabolic disorder having an estimated worldwide incidence of one in forty thousand live births. While the introduction of enzyme replacement therapy (ERT) has considerably increased the awareness of the disease, the delay in diagnosis is still consistent and most patients go undetected. Objective This study aimed to determine the prevalence of late-onset Pompe disease (LOPD) in a high-risk population, using dried blood spot (DBS) as a main screening tool. Methods This cross-sectional study was performed on the 93 patients who attended to the neuromuscular center of Bu-ali hospital in Tehran, Iran, during 2014–2015. Inclusion criteria were: 1) age ≥1 years, 2) proximal myopathies of unknown etiology in lower limbs or symptoms of limb girdle muscle weakness (LGMW), and 3) unexplained elevated CPK (>174). Acid α-glucosidase (GAA) activity was measured separately on DBS by fluorometric method. For the final diagnosis, GAA deficiency was confirmed by a biochemical assay in skeletal muscle, whereas genotype was assessed by GAA molecular analysis. All statistical tests were performed using the SPSS version 16. Results are presented as mean (SD) or median (IQR), as appropriate. Results In a 12-month period, we studied 93 cases: 5 positive samples (5.3%) were detected by DBS screening, biochemical and molecular genetic studies finally confirmed LOPD diagnosis in 3 cases (3.22%). Among the 93 patients, 100% showed hyperCKemia, 89 patients (95.7%) showed LGMW and 4 patients had symptoms of proximal myopathies in the lower limb. Conclusions Results from the LOPED study suggest that GAA activity requires accurate screening by DBS in all patients referred for hyperCKemia and/or LGMW.
Collapse
Affiliation(s)
- Khadijeh Haji Naghi Tehrani
- M.D., Neurologist, Assistant Professor, Department of Neurology, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Elmira Sakhaeyan
- M.D., Graduated from Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Elnaz Sakhaeyan
- Pharm.D., Graduated from Tehran University of Medical Science (TUMS), Tehran, Iran
| |
Collapse
|
83
|
McIntosh P, Austin S, Sullivan J, Bailey L, Bailey C, Viskochil D, Kishnani PS. Three cases of multi-generational Pompe disease: Are current practices missing diagnostic and treatment opportunities? Am J Med Genet A 2017; 173:2628-2634. [PMID: 28763149 DOI: 10.1002/ajmg.a.38369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/08/2017] [Accepted: 06/25/2017] [Indexed: 11/07/2022]
Abstract
Pompe disease (Glycogen storage disease type II, GSDII, or acid maltase deficiency) is an autosomal recessive metabolic myopathy with a broad clinical spectrum, ranging from infantile to late-onset presentations. In 2015, Pompe disease was added as a core condition to the Recommended Uniform Screening Panel for state newborn screening (NBS). The clinical importance of Pompe disease is evolving with the use of NBS, increasing awareness of the disease, and higher than previously reported disease prevalence; however, current practices miss additional diagnostic and potential treatment opportunities in close relatives of the family proband. In this report, we describe three families with multiple individuals in multiple generations affected by both infantile and late-onset clinical presentations of Pompe disease. The presence of multi-generational disease within these families highlights the importance of subsequent risk assessment through medical history and physical examination, with a low threshold for the screening of a proband's family members. We recommend enzymology (GAA activity assay) as the first screening method, as opposed to targeted mutation analysis, for at-risk family members. Given that the initial symptoms of the slowly progressive late-onset presentation of Pompe disease may be mild or non-specific, enzymatic testing of all parents of affected infants should be considered.
Collapse
Affiliation(s)
- Paul McIntosh
- Duke University Medical Center, Durham, North Carolina
| | | | | | - Lauren Bailey
- Duke University Medical Center, Durham, North Carolina
| | | | | | | |
Collapse
|
84
|
Rapidly Progressive White Matter Involvement in Early Childhood: The Expanding Phenotype of Infantile Onset Pompe? JIMD Rep 2017; 39:55-62. [PMID: 28726123 DOI: 10.1007/8904_2017_46] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 05/31/2017] [Accepted: 06/30/2017] [Indexed: 01/01/2023] Open
Abstract
Glycogen accumulation in the central nervous system of patients with classical infantile onset Pompe disease (IOPD) has been a consistent finding on the few post-mortems performed. While delays in myelination and a possible reduction in processing speed have previously been noted, it has only been recently that the potential for clinically significant progressive white matter disease has been noted. The limited reports thus far published infer that in some IOPD patients, this manifests as intellectual decline in the second decade of life. We present a CRIM negative patient, immunomodulated with rituximab and methotrexate at birth, who despite an initial good clinical response to ERT, at the age of just under 4 years, presented with evolving spasticity in the lower limbs. The investigation of which revealed progressive central nervous system involvement. Given both the earlier onset of the symptoms and consanguineous familial pedigree, extensive biochemical and genetic investigation was undertaken to ensure no alternative pathology was elucidated. In light of these findings, we review the radiology and post-mortems of previous cases and discuss the potential mechanisms that may underlie this presentation.
Collapse
|
85
|
Rigante D, Cipolla C, Basile U, Gulli F, Savastano MC. Overview of immune abnormalities in lysosomal storage disorders. Immunol Lett 2017; 188:79-85. [PMID: 28687233 DOI: 10.1016/j.imlet.2017.07.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/28/2017] [Accepted: 07/03/2017] [Indexed: 01/01/2023]
Abstract
The critical relevance of the lysosomal compartment for normal cellular function can be proved by numbering the clinical phenotypes that arise in lysosomal storage disorders (LSDs), a group of around 70 different monogenic autosomal or X-linked syndromes, caused by specific lysosomal enzyme deficiencies: all LSDs are characterized by progressive accumulation of heterogeneous biologic materials in the lysosomes of various parts of the body such as viscera, skeleton, skin, heart, and central nervous system. At least a fraction of LSDs has been associated with mixed abnormalities involving the immune system, while some patients with LSDs may result more prone to autoimmune phenomena. A large production of proinflammatory cytokines has been observed in Gaucher and Fabry diseases, and wide different autoantibody production has been also reported in both. Many immune-mediated reactions are crucial to the pathogenesis of different inflammatory signs in mucopolysaccharidoses, and subverted heparan sulphate catabolism might dysregulate cellular homeostasis in the brain of these patients. Furthermore, an inappropriate activation of microglia is implicated in the neurodegenerative foci of Niemann-Pick disease, in which abnormal signalling pathways are activated by impaired sphingolipid metabolism. In addition, not the simple impaired catabolism of gangliosides per se, but also the production of anti-ganglioside autoantibodies contributes to the neurological disease of gangliosidoses. Even if the exact relationship between the modification of lysosomal activities and modulation of the immune system remains obscure, there is emerging evidence of different impaired immunity responses in a variety of LSDs: in this review we investigate and summarize the immune abnormalities and/or clinical data about immune system irregularities which have been described in a subset of LSDs.
Collapse
Affiliation(s)
- Donato Rigante
- Institute of Pediatrics, Fondazione Policlinico Universitario A. Gemelli, Università Cattolica Sacro Cuore, Rome, Italy.
| | - Clelia Cipolla
- Institute of Pediatrics, Fondazione Policlinico Universitario A. Gemelli, Università Cattolica Sacro Cuore, Rome, Italy
| | - Umberto Basile
- Department of Laboratory Medicine, Fondazione Policlinico Universitario A. Gemelli, Università Cattolica Sacro Cuore, Rome, Italy
| | - Francesca Gulli
- Laboratory of Clinical Pathology, Ospedale M.G. Vannini, Institute Figlie S. Camillo, Rome, Italy
| | | |
Collapse
|
86
|
Abstract
Subspecialty pediatric practice provides comprehensive medical care for a range of ages, from premature infants to children, and often includes adults with complex medical and surgical issues that warrant multidisciplinary care. Normal physiologic variations involving different body systems occur during sleep and these vary with age, stage of sleep, and underlying health conditions. This article is a concise review of the cardiovascular (CV) physiology and pathophysiology in children, sleep-disordered breathing (SDB) contributing to CV morbidity, congenital and acquired CV pathology resulting in SDB, and the relationship between SDB and CV morbidity in different clinical syndromes and systemic diseases in the expanded pediatric population.
Collapse
Affiliation(s)
- Grace R Paul
- Division of Pulmonary and Sleep Medicine, Nationwide Children's Hospital, The Ohio State University, 700 Children's Drive, Columbus, OH 43205, USA.
| | - Swaroop Pinto
- Division of Pulmonary and Sleep Medicine, Nationwide Children's Hospital, The Ohio State University, 700 Children's Drive, Columbus, OH 43205, USA
| |
Collapse
|
87
|
Chen X, Liu T, Huang M, Wu J, Zhu J, Guo Y, Xu X, Li F, Wang J, Fu L. Clinical and Molecular Characterization of Infantile-Onset Pompe Disease in Mainland Chinese Patients: Identification of Two Common Mutations. Genet Test Mol Biomarkers 2017; 21:391-396. [PMID: 28394184 DOI: 10.1089/gtmb.2016.0424] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS We sought to understand the clinical course and molecular defects of infantile-onset Pompe disease (IOPD) among mainland Chinese patients. MATERIALS AND METHODS Twenty-five Chinese patients with IOPD were enrolled and clinical data were retrospectively reviewed. The entire coding region of the GAA gene was amplified by polymerase chain reaction and analyzed by direct sequencing. RESULTS The median age at symptom onset was 3.4 months (range: 1.0-7.1 months) and 4.9 months (range: 2.7-8.3 months) at diagnosis. Only one patient received enzyme replacement therapy (ERT) and this child survived beyond the age of 2 years. Of the 24 patients not receiving ERT, all, but one patient, died at a median age of 8.3 months (range: 4.0-12.2 months). Thirteen novel and two common GAA mutations were identified in this study. The allelic frequency of c.2662G > T (p.Glu888X) was 23.1% in northern Chinese patients and 4.2% in southern Chinese patients, whereas the allelic frequency of c.1935C > A (p.Asp645Glu) was 20.8% in southern and 3.8% in northern Chinese patients. CONCLUSIONS We identified the most common mutations in southern and northern Chinese patients with IOPD.
Collapse
Affiliation(s)
- Xi Chen
- 1 Department of Cardiology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Tingliang Liu
- 1 Department of Cardiology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Meirong Huang
- 1 Department of Cardiology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Jinjin Wu
- 1 Department of Cardiology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Junxue Zhu
- 1 Department of Cardiology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Ying Guo
- 1 Department of Cardiology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Xinyi Xu
- 1 Department of Cardiology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Fen Li
- 1 Department of Cardiology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Jian Wang
- 2 Research Division of Birth Defects, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine , Shanghai, China
| | - Lijun Fu
- 1 Department of Cardiology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine , Shanghai, China .,3 Research Division of Cardiovascular Disease, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine , Shanghai, China
| |
Collapse
|
88
|
Delbridge LMD, Mellor KM, Taylor DJ, Gottlieb RA. Myocardial stress and autophagy: mechanisms and potential therapies. Nat Rev Cardiol 2017; 14:412-425. [PMID: 28361977 DOI: 10.1038/nrcardio.2017.35] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Autophagy is a ubiquitous cellular catabolic process responsive to energy stress. Research over the past decade has revealed that cardiomyocyte autophagy is a prominent homeostatic pathway, important in adaptation to altered myocardial metabolic demand. The cellular machinery of autophagy involves targeted direction of macromolecules and organelles for lysosomal degradation. Activation of autophagy has been identified as cardioprotective in some settings (that is, ischaemia and ischaemic preconditioning). In other situations, sustained autophagy has been linked with cardiopathology (for example, sustained pressure overload and heart failure). Perturbation of autophagy in diabetic cardiomyopathy has also been observed and is associated with both adaptive and maladaptive responses to stress. Emerging research findings indicate that various forms of selective autophagy operate in parallel to manage various types of catabolic cellular cargo including mitochondria, large proteins, glycogen, and stored lipids. In this Review, induction of autophagy associated with cardiac benefit or detriment is considered. The various static and dynamic approaches used to measure autophagy are critiqued, and current inconsistencies in the understanding of autophagy regulation in the heart are highlighted. The prospects for pharmacological intervention to achieve therapeutic manipulation of autophagic processes are also discussed.
Collapse
Affiliation(s)
- Lea M D Delbridge
- School of Biomedical Sciences, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Kimberley M Mellor
- Department of Physiology, Medical &Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand
| | - David J Taylor
- Heart Institute, Cedars-Sinai Hospital, 127 South San Vicente Boulevard, Los Angeles, California 90048, USA
| | - Roberta A Gottlieb
- Heart Institute, Cedars-Sinai Hospital, 127 South San Vicente Boulevard, Los Angeles, California 90048, USA
| |
Collapse
|
89
|
Whyte LS, Lau AA, Hemsley KM, Hopwood JJ, Sargeant TJ. Endo-lysosomal and autophagic dysfunction: a driving factor in Alzheimer's disease? J Neurochem 2017; 140:703-717. [PMID: 28027395 DOI: 10.1111/jnc.13935] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/19/2016] [Accepted: 12/19/2016] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, and its prevalence will increase significantly in the coming decades. Although important progress has been made, fundamental pathogenic mechanisms as well as most hereditary contributions to the sporadic form of the disease remain unknown. In this review, we examine the now substantial links between AD pathogenesis and lysosomal biology. The lysosome hydrolyses and processes cargo delivered by multiple pathways, including endocytosis and autophagy. The endo-lysosomal and autophagic networks are central to clearance of cellular macromolecules, which is important given there is a deficit in clearance of amyloid-β in AD. Numerous studies show prominent lysosomal dysfunction in AD, including perturbed trafficking of lysosomal enzymes and accumulation of the same substrates that accumulate in lysosomal storage disorders. Examination of the brain in lysosomal storage disorders shows the accumulation of amyloid precursor protein metabolites, which further links lysosomal dysfunction with AD. This and other evidence leads us to hypothesise that genetic variation in lysosomal genes modifies the disease course of sporadic AD.
Collapse
Affiliation(s)
- Lauren S Whyte
- Lysosomal Diseases Research Unit, Nutrition and Metabolism Theme, South Australian Health and Medical Research Institute, Adelaide, Australia.,School of Medicine, University of Adelaide, Adelaide, Australia
| | - Adeline A Lau
- Lysosomal Diseases Research Unit, Nutrition and Metabolism Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Kim M Hemsley
- Lysosomal Diseases Research Unit, Nutrition and Metabolism Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - John J Hopwood
- Lysosomal Diseases Research Unit, Nutrition and Metabolism Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Timothy J Sargeant
- Lysosomal Diseases Research Unit, Nutrition and Metabolism Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| |
Collapse
|
90
|
Abstract
PURPOSE OF REVIEW The article provides an overview of advances in the induced pluripotent stem cell field to model cardiomyopathies of inherited inborn errors of metabolism and acquired metabolic syndromes in vitro. RECENT FINDINGS Several inborn errors of metabolism have been studied using 'disease in a dish' models, including Pompe disease, Danon disease, Fabry disease, and Barth syndrome. Disease phenotypes of complex metabolic syndromes, such as diabetes mellitus and aldehyde dehydrogenase 2 deficiency, have also been observed. SUMMARY Differentiation of patient and disease-specific induced pluripotent stem cell-derived cardiomyocytes has provided the capacity to model deleterious cardiometabolic diseases to understand molecular mechanisms, perform drug screens, and identify novel drug targets.
Collapse
|
91
|
Wu J, Yang Y, Sun C, Sun S, Li Q, Yao Y, Fei F, Lu L, Chang Z, Zhang W, Wang X, Luo F. Disruption of the gaa Gene in Zebrafish Fails to Generate the Phenotype of Classical Pompe Disease. DNA Cell Biol 2017; 36:10-17. [PMID: 28045567 DOI: 10.1089/dna.2016.3459] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Jing Wu
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Yi Yang
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Chengjun Sun
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Shaoyang Sun
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qiang Li
- Translational Medical Center for Development and Disease, Shanghai Key Laboratory of Birth Defect, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Yuxiao Yao
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Fei Fei
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, School of Medicine, Yale Cancer Center, Yale University, New Haven, Connecticut
| | - Zhuo Chang
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Wenting Zhang
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Xu Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Feihong Luo
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
92
|
Bobillo Lobato J, Jiménez Hidalgo M, Jiménez Jiménez LM. Biomarkers in Lysosomal Storage Diseases. Diseases 2016; 4:diseases4040040. [PMID: 28933418 PMCID: PMC5456325 DOI: 10.3390/diseases4040040] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 12/04/2016] [Accepted: 12/12/2016] [Indexed: 12/22/2022] Open
Abstract
A biomarker is generally an analyte that indicates the presence and/or extent of a biological process, which is in itself usually directly linked to the clinical manifestations and outcome of a particular disease. The biomarkers in the field of lysosomal storage diseases (LSDs) have particular relevance where spectacular therapeutic initiatives have been achieved, most notably with the introduction of enzyme replacement therapy (ERT). There are two main types of biomarkers. The first group is comprised of those molecules whose accumulation is directly enhanced as a result of defective lysosomal function. These molecules represent the storage of the principal macro-molecular substrate(s) of a specific enzyme or protein, whose function is deficient in the given disease. In the second group of biomarkers, the relationship between the lysosomal defect and the biomarker is indirect. In this group, the biomarker reflects the effects of the primary lysosomal defect on cell, tissue, or organ functions. There is no “gold standard” among biomarkers used to diagnosis and/or monitor LSDs, but there are a number that exist that can be used to reasonably assess and monitor the state of certain organs or functions. A number of biomarkers have been proposed for the analysis of the most important LSDs. In this review, we will summarize the most promising biomarkers in major LSDs and discuss why these are the most promising candidates for screening systems.
Collapse
Affiliation(s)
- Joaquin Bobillo Lobato
- Servicio de Bioquímica Clínica, Unidad de Gestión Clínica de Laboratorios, Hospital Universitario Nuestra Señora de Valme, 41014-Sevilla, Spain.
| | - Maria Jiménez Hidalgo
- Servicio de Fisiopatología Celular y Bioenergética, Servicios Centrales de Investigación, Universidad Pablo de Olavide, 41013-Sevilla, Spain.
| | - Luis M Jiménez Jiménez
- Servicio de Fisiopatología Celular y Bioenergética, Servicios Centrales de Investigación, Universidad Pablo de Olavide, 41013-Sevilla, Spain.
| |
Collapse
|
93
|
|
94
|
Stütz AE, Wrodnigg TM. Carbohydrate-Processing Enzymes of the Lysosome: Diseases Caused by Misfolded Mutants and Sugar Mimetics as Correcting Pharmacological Chaperones. Adv Carbohydr Chem Biochem 2016; 73:225-302. [PMID: 27816107 DOI: 10.1016/bs.accb.2016.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lysosomal storage diseases are hereditary disorders caused by mutations on genes encoding for one of the more than fifty lysosomal enzymes involved in the highly ordered degradation cascades of glycans, glycoconjugates, and other complex biomolecules in the lysosome. Several of these metabolic disorders are associated with the absence or the lack of activity of carbohydrate-processing enzymes in this cell compartment. In a recently introduced therapy concept, for susceptible mutants, small substrate-related molecules (so-called pharmacological chaperones), such as reversible inhibitors of these enzymes, may serve as templates for the correct folding and transport of the respective protein mutant, thus improving its concentration and, consequently, its enzymatic activity in the lysosome. Carbohydrate-processing enzymes in the lysosome, related lysosomal diseases, and the scope and limitations of reported reversible inhibitors as pharmacological chaperones are discussed with a view to possibly extending and improving research efforts in this area of orphan diseases.
Collapse
Affiliation(s)
- Arnold E Stütz
- Glycogroup, Institute of Organic Chemistry, Graz University of Technology, Graz, Austria
| | - Tanja M Wrodnigg
- Glycogroup, Institute of Organic Chemistry, Graz University of Technology, Graz, Austria
| |
Collapse
|
95
|
Kuchař L, Asfaw B, Rybová J, Ledvinová J. Tandem Mass Spectrometry of Sphingolipids: Applications for Diagnosis of Sphingolipidoses. Adv Clin Chem 2016; 77:177-219. [PMID: 27717417 DOI: 10.1016/bs.acc.2016.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In recent years, mass spectrometry (MS) has become the dominant technology in lipidomic analysis. It is widely used in diagnosis and research of lipid metabolism disorders including those characterized by impairment of lysosomal functions and storage of nondegraded-degraded substrates. These rare diseases, which include sphingolipidoses, have severe and often fatal clinical consequences. Modern MS methods have contributed significantly to achieve a definitive diagnosis, which is essential in clinical practice to begin properly targeted patient care. Here we summarize MS and tandem MS methods used for qualitative and quantitative analysis of sphingolipids (SL) relative to the diagnostic process for sphingolipidoses and studies focusing on alterations in cell functions due to these disorders. This review covers the following topics: Tandem MS is sensitive and robust in determining the composition of sphingolipid classes in various biological materials. Its ability to establish SL metabolomic profiles using MS bench-top analyzers, significantly benefits the first stages of a diagnosis as well as metabolic studies of these disorders. It can thus contribute to a better understanding of the biological significance of SL.
Collapse
Affiliation(s)
- L Kuchař
- Charles University in Prague and General University Hospital, Prague, Czech Republic.
| | - B Asfaw
- Charles University in Prague and General University Hospital, Prague, Czech Republic
| | - J Rybová
- Charles University in Prague and General University Hospital, Prague, Czech Republic
| | - J Ledvinová
- Charles University in Prague and General University Hospital, Prague, Czech Republic.
| |
Collapse
|
96
|
Schneider JL, Balu-Iyer SV. Phosphatidylserine Converts Immunogenic Recombinant Human Acid Alpha-Glucosidase to a Tolerogenic Form in a Mouse Model of Pompe Disease. J Pharm Sci 2016; 105:3097-3104. [PMID: 27488899 PMCID: PMC5021602 DOI: 10.1016/j.xphs.2016.06.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 06/16/2016] [Accepted: 06/17/2016] [Indexed: 12/16/2022]
Abstract
Development of unwanted immune responses against therapeutic proteins is a major clinical complication. Recently, we have shown that exposure of Factor VIII in the presence of phosphatidylserine (PS) induces antigen-specific hyporesponsiveness to Factor VIII rechallenge, suggesting that PS is not immune suppressive, but rather immune regulatory in that PS converts an immunogen to a tolerogen. Since PS is exposed in the outer leaflet during apoptosis, we hypothesize that PS imparts tolerogenic activity to this natural process. Thus, immunization with PS containing liposomes would mimic this natural process. Here, we investigate the immune regulatory effects of PS in inducing tolerance toward recombinant human acid alpha-glucosidase (rhGAA). rhGAA was found to complex with PS liposomes through hydrophobic interactions, and incubation PS-rhGAA with dendritic cells resulted in the increased secretion of transforming growth factor-β. Immunization with PS-rhGAA or O-phospho-L-serine-rhGAA led to a reduction in anti-rhGAA antibody response which persisted despite rechallenge with free rhGAA. Importantly, the titer levels in a majority of these animals remained unchanged after rechallenge and can be considered nonresponders. These data provide evidence that PS liposomes can be used to induce tolerance toward therapeutic proteins, in general.
Collapse
Affiliation(s)
- Jennifer L Schneider
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214
| | - Sathy V Balu-Iyer
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214.
| |
Collapse
|
97
|
Bhattacharya K, Pontin J, Thompson S. Dietary Management of the Ketogenic Glycogen Storage Diseases. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2016. [DOI: 10.1177/2326409816661359] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Kaustuv Bhattacharya
- Genetic Metabolic Disorders Service, The Children’s Hospital at Westmead, Sydney, New South Wales, Australia
- Discipline of Paediatrics and Child Health, Sydney University, Sydney, New South Wales, Australia
| | - Jennifer Pontin
- Genetic Metabolic Disorders Service, The Children’s Hospital at Westmead, Sydney, New South Wales, Australia
| | - Sue Thompson
- Genetic Metabolic Disorders Service, The Children’s Hospital at Westmead, Sydney, New South Wales, Australia
| |
Collapse
|
98
|
van der Ploeg A, Carlier PG, Carlier RY, Kissel JT, Schoser B, Wenninger S, Pestronk A, Barohn RJ, Dimachkie MM, Goker-Alpan O, Mozaffar T, Pena LDM, Simmons Z, Straub V, Guglieri M, Young P, Boentert M, Baudin PY, Wens S, Shafi R, Bjartmar C, Thurberg BL. Prospective exploratory muscle biopsy, imaging, and functional assessment in patients with late-onset Pompe disease treated with alglucosidase alfa: The EMBASSY Study. Mol Genet Metab 2016; 119:115-23. [PMID: 27473031 DOI: 10.1016/j.ymgme.2016.05.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 11/23/2022]
Abstract
BACKGROUND Late-onset Pompe disease is characterized by progressive skeletal myopathy followed by respiratory muscle weakness, typically leading to loss of ambulation and respiratory failure. In this population, enzyme replacement therapy (ERT) with alglucosidase alfa has been shown to stabilize respiratory function and improve mobility and muscle strength. Muscle pathology and glycogen clearance from skeletal muscle in treatment-naïve adults after ERT have not been extensively examined. METHODS This exploratory, open-label, multicenter study evaluated glycogen clearance in muscle tissue samples collected pre- and post- alglucosidase alfa treatment in treatment-naïve adults with late-onset Pompe disease. The primary endpoint was the quantitative reduction in percent tissue area occupied by glycogen in muscle biopsies from baseline to 6months. Secondary endpoints included qualitative histologic assessment of tissue glycogen distribution, secondary pathology changes, assessment of magnetic resonance images (MRIs) for intact muscle and fatty replacement, and functional assessments. RESULTS Sixteen patients completed the study. After 6months of ERT, the percent tissue area occupied by glycogen in quadriceps and deltoid muscles decreased in 10 and 8 patients, respectively. No changes were detected on MRI from baseline to 6months. A majority of patients showed improvements on functional assessments after 6months of treatment. All treatment-related adverse events were mild or moderate. CONCLUSIONS This exploratory study provides novel insights into the histopathologic effects of ERT in late-onset Pompe disease patients. Ultrastructural examination of muscle biopsies demonstrated reduced lysosomal glycogen after ERT. Findings are consistent with stabilization of disease by ERT in treatment-naïve patients with late-onset Pompe disease.
Collapse
Affiliation(s)
- Ans van der Ploeg
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Pierre G Carlier
- Institut de Myologie, AIM and CEA NMR Laboratory - Spectroscopy Laboratory, Université Pierre et Marie Curie, Paris, France
| | - Robert-Yves Carlier
- Medical Imaging Department, Raymond Poincare University Hospital, Garches, France
| | - John T Kissel
- Department of Neurology, Division of Neuromuscular Medicine, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Benedikt Schoser
- Friedrich-Baur-Institut, Neurologische Klinik, Klinikum der Universität München, München, Germany
| | - Stephan Wenninger
- Friedrich-Baur-Institut, Neurologische Klinik, Klinikum der Universität München, München, Germany
| | - Alan Pestronk
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Richard J Barohn
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mazen M Dimachkie
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ozlem Goker-Alpan
- Lysosomal Disorders Unit and Center for Clinical Trials, O&O Alpan LLC, Fairfax, VA, USA
| | - Tahseen Mozaffar
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Loren D M Pena
- Division of Pediatric Medical Genetics, Duke University Medical Center, Durham, NC, USA
| | | | - Volker Straub
- Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Michela Guglieri
- Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Peter Young
- Department of Sleep Medicine and Neuromuscular Disorders, University Hospital of Münster, Münster, Germany
| | - Matthias Boentert
- Department of Sleep Medicine and Neuromuscular Disorders, University Hospital of Münster, Münster, Germany
| | | | - Stephan Wens
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | | | | | | |
Collapse
|
99
|
Chen MA, Weinstein DA. Glycogen storage diseases: Diagnosis, treatment and outcome. ACTA ACUST UNITED AC 2016. [DOI: 10.3233/trd-160006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
| | - David A. Weinstein
- Glycogen Storage Disease Program, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
100
|
Sato Y, Kobayashi H, Higuchi T, Shimada Y, Ida H, Ohashi T. Metabolomic Profiling of Pompe Disease-Induced Pluripotent Stem Cell-Derived Cardiomyocytes Reveals That Oxidative Stress Is Associated with Cardiac and Skeletal Muscle Pathology. Stem Cells Transl Med 2016; 6:31-39. [PMID: 28170191 PMCID: PMC5442755 DOI: 10.5966/sctm.2015-0409] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 06/13/2016] [Indexed: 12/25/2022] Open
Abstract
Pompe disease (PD) is a lysosomal storage disease that is caused by a deficiency of the acid α‐glucosidase, which results in glycogen accumulation in the lysosome. The major clinical symptoms of PD include skeletal muscle weakness, respiratory failure, and cardiac hypertrophy. Based on its severity and symptom onset, PD is classified into infantile and late‐onset forms. Lysosomal accumulation of glycogen can promote many types of cellular dysfunction, such as autophagic dysfunction, endoplasmic reticulum stress, and abnormal calcium signaling within skeletal muscle. However, the disease mechanism underlying PD cardiomyopathy is not fully understood. Several researchers have shown that PD induced pluripotent stem cell (iPSC)‐derived cardiomyocytes successfully replicate the disease phenotype and are useful disease models. We have analyzed the metabolomic profile of late‐onset PD iPSC‐derived cardiomyocytes and found that oxidative stress and mitochondrial dysfunction are likely associated with cardiac complications. Furthermore, we have validated that these disease‐specific changes were also observed in the cardiomyocytes and skeletal muscle of a genetically engineered murine PD model. Oxidative stress may contribute to skeletal muscle and cardiomyocyte dysfunction in PD mice; however, NF‐E2‐related factor 2 was downregulated in cardiomyocytes and skeletal muscle, despite evidence of oxidative stress. We hypothesized that oxidative stress and an impaired antioxidative stress response mechanism may underlie the molecular pathology of late‐onset PD. Stem Cells Translational Medicine2017;6:31–39
Collapse
Affiliation(s)
- Yohei Sato
- Department of Pediatrics, Jikei University School of Medicine, Tokyo, Japan
- Division of Gene Therapy, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| | - Hiroshi Kobayashi
- Department of Pediatrics, Jikei University School of Medicine, Tokyo, Japan
- Division of Gene Therapy, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Higuchi
- Division of Gene Therapy, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| | - Yohta Shimada
- Division of Gene Therapy, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| | - Hiroyuki Ida
- Department of Pediatrics, Jikei University School of Medicine, Tokyo, Japan
- Division of Gene Therapy, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| | - Toya Ohashi
- Department of Pediatrics, Jikei University School of Medicine, Tokyo, Japan
- Division of Gene Therapy, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|