Modeling Duchenne Muscular Dystrophy Cardiomyopathy with Patients’ Induced Pluripotent Stem-Cell-Derived Cardiomyocytes
暂无分享,去创建一个
[1] D. Mack,et al. Calcium handling maturation and adaptation to increased substrate stiffness in human iPSC-derived cardiomyocytes: The impact of full-length dystrophin deficiency , 2022, Frontiers in Physiology.
[2] E. Gottlieb,et al. Bioenergetic and Metabolic Impairments in Induced Pluripotent Stem Cell-Derived Cardiomyocytes Generated from Duchenne Muscular Dystrophy Patients , 2022, International journal of molecular sciences.
[3] Y. C. Chai,et al. Long-term culture of patient-derived cardiac organoids recapitulated Duchenne muscular dystrophy cardiomyopathy and disease progression , 2022, bioRxiv.
[4] C. de la Torre,et al. RhoA/ROCK2 signalling is enhanced by PDGF‐AA in fibro‐adipogenic progenitor cells: implications for Duchenne muscular dystrophy , 2022, Journal of cachexia, sarcopenia and muscle.
[5] K. Desloovere,et al. Human iPSC model reveals a central role for NOX4 and oxidative stress in Duchenne cardiomyopathy , 2022, Stem cell reports.
[6] D. Mack,et al. Full-length dystrophin deficiency leads to contractile and calcium transient defects in human engineered heart tissues , 2022, Journal of tissue engineering.
[7] G. Viglietto,et al. From Spheroids to Organoids: The Next Generation of Model Systems of Human Cardiac Regeneration in a Dish , 2021, International journal of molecular sciences.
[8] H. Roderick,et al. Incomplete Assembly of the Dystrophin-Associated Protein Complex in 2D and 3D-Cultured Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes , 2021, Frontiers in Cell and Developmental Biology.
[9] H. Sakurai,et al. Orai1–STIM1 Regulates Increased Ca2+ Mobilization, Leading to Contractile Duchenne Muscular Dystrophy Phenotypes in Patient-Derived Induced Pluripotent Stem Cells , 2021, Biomedicines.
[10] E. Olson,et al. Cardiac Myoediting Attenuates Cardiac Abnormalities in Human and Mouse Models of Duchenne Muscular Dystrophy , 2021, Circulation research.
[11] H. Baharvand,et al. Organoids: a novel modality in disease modeling , 2021, Bio-Design and Manufacturing.
[12] M. Lawlor,et al. Cardiomyocyte-produced miR-339-5p mediates pathology in Duchenne muscular dystrophy cardiomyopathy. , 2021, Human molecular genetics.
[13] M. Zatz,et al. Sarcoglycanopathies: an update , 2021, Neuromuscular Disorders.
[14] E. Olson,et al. A consolidated AAV system for single-cut CRISPR correction of a common Duchenne muscular dystrophy mutation , 2021, Molecular therapy. Methods & clinical development.
[15] F. Fieschi,et al. NADPH Oxidases (NOX): An Overview from Discovery, Molecular Mechanisms to Physiology and Pathology , 2021, Antioxidants.
[16] I. Komuro,et al. Decreased YAP activity reduces proliferative ability in human induced pluripotent stem cell of duchenne muscular dystrophy derived cardiomyocytes , 2021, Scientific Reports.
[17] H. Blau,et al. Increased tissue stiffness triggers contractile dysfunction and telomere shortening in dystrophic cardiomyocytes , 2021, Stem cell reports.
[18] E. Olson,et al. Precise correction of Duchenne muscular dystrophy exon deletion mutations by base and prime editing , 2021, Science Advances.
[19] J. Chamberlain,et al. Micro-dystrophin gene therapy prevents heart failure in an improved Duchenne muscular dystrophy cardiomyopathy mouse model , 2021, JCI insight.
[20] E. Gottlieb,et al. Depressed β‐adrenergic inotropic responsiveness and intracellular calcium handling abnormalities in Duchenne Muscular Dystrophy patients’ induced pluripotent stem cell–derived cardiomyocytes , 2021, Journal of cellular and molecular medicine.
[21] S. Remy,et al. Cardiovascular phenotype of the Dmdmdx rat – a suitable animal model for Duchenne muscular dystrophy , 2021, Disease Models & Mechanisms.
[22] S. A. Shariati,et al. Microtechnology-based methods for organoid models , 2020, Microsystems & Nanoengineering.
[23] C. G. Perry,et al. Mitochondrial bioenergetic dysfunction in the D2.mdx model of Duchenne muscular dystrophy is associated with microtubule disorganization in skeletal muscle , 2020, PloS one.
[24] C. Gersbach,et al. In Vivo Gene Editing of Muscle Stem Cells with Adeno-Associated Viral Vectors in a Mouse Model of Duchenne Muscular Dystrophy , 2020, Molecular therapy. Methods & clinical development.
[25] Yu-Sheng Chen,et al. Development of a Cardiac Sarcomere Functional Genomics Platform to Enable Scalable Interrogation of Human TNNT2 Variants , 2020, Circulation.
[26] P. Skládal,et al. DMD Pluripotent Stem Cell Derived Cardiac Cells Recapitulate in vitro Human Cardiac Pathophysiology , 2020, Frontiers in Bioengineering and Biotechnology.
[27] T. Luongo,et al. The debate continues - What is the role of MCU and mitochondrial calcium uptake in the heart? , 2020, Journal of molecular and cellular cardiology.
[28] Jianyi(Jay) Zhang,et al. Stem Cell-Derived Cardiomyocytes and Beta-Adrenergic Receptor Blockade in Duchenne Muscular Dystrophy Cardiomyopathy. , 2020, Journal of the American College of Cardiology.
[29] E. Murphy,et al. Role of Mitochondrial Calcium and the Permeability Transition Pore in Regulating Cell Death. , 2020, Circulation research.
[30] A. Nakamura,et al. Amelioration of intracellular Ca2+ regulation by exon-45 skipping in Duchenne muscular dystrophy-induced pluripotent stem cell-derived cardiomyocytes. , 2019, Biochemical and biophysical research communications.
[31] S. Nelson,et al. Targeting RyR Activity Boosts Antisense Exon 44 and 45 Skipping in Human DMD Skeletal or Cardiac Muscle Culture Models , 2019, Molecular therapy. Nucleic acids.
[32] R. Moritz,et al. Absence of full-length dystrophin impairs normal maturation and contraction of cardiomyocytes derived from human induced pluripotent stem cells. , 2019, Cardiovascular research.
[33] E. Marbán,et al. Disease-modifying bioactivity of intravenous cardiosphere-derived cells and exosomes in mdx mice. , 2019, JCI insight.
[34] J. Takita,et al. The intracellular Ca2+ concentration is elevated in cardiomyocytes differentiated from hiPSCs derived from a Duchenne muscular dystrophy patient , 2019, PloS one.
[35] G. Comi,et al. Fibrosis Rescue Improves Cardiac Function in Dystrophin-Deficient Mice and Duchenne Patient-Specific Cardiomyocytes by Immunoproteasome Modulation. , 2019, The American journal of pathology.
[36] P. Skládal,et al. Non-invasive electromechanical cell-based biosensors for improved investigation of 3D cardiac models. , 2019, Biosensors & bioelectronics.
[37] D. Freimark,et al. Electrophysiological abnormalities in induced pluripotent stem cell‐derived cardiomyocytes generated from Duchenne muscular dystrophy patients , 2019, Journal of cellular and molecular medicine.
[38] Charles A. Gersbach,et al. Long-term Evaluation of AAV-CRISPR Genome Editing for Duchenne Muscular Dystrophy , 2018, Nature Medicine.
[39] J. Ervasti,et al. Dystrophinopathy-associated dysfunction of Krebs cycle metabolism , 2018, Human molecular genetics.
[40] Melanie Gartz,et al. Exosomes exert cardioprotection in dystrophin-deficient cardiomyocytes via ERK1/2-p38/MAPK signaling , 2018, Scientific Reports.
[41] M. Scheinman,et al. Post-Translational Modifications and Diastolic Calcium Leak Associated to the Novel RyR2-D3638A Mutation Lead to CPVT in Patient-Specific hiPSC-Derived Cardiomyocytes , 2018, Journal of clinical medicine.
[42] D. Freimark,et al. Functional abnormalities in induced Pluripotent Stem Cell-derived cardiomyocytes generated from titin-mutated patients with dilated cardiomyopathy , 2018, PloS one.
[43] S. Messina,et al. Hippo signaling pathway is altered in Duchenne muscular dystrophy , 2018, PloS one.
[44] Jia Li,et al. Genetic Modulation of RNA Splicing with a CRISPR-Guided Cytidine Deaminase. , 2018, Molecular cell.
[45] L. Terada,et al. ROS signaling and ER stress in cardiovascular disease. , 2018, Molecular aspects of medicine.
[46] Daniel J. Garry,et al. Integrative effects of dystrophin loss on metabolic function of the mdx mouse , 2018, Scientific Reports.
[47] E. Ashley,et al. Telomere shortening is a hallmark of genetic cardiomyopathies , 2018, Proceedings of the National Academy of Sciences.
[48] E. Derivery,et al. A Fluorescent Membrane Tension Probe , 2018, Nature Chemistry.
[49] Qian Fan,et al. lncRNA ENSMUST00000134285 Increases MAPK11 Activity, Regulating Aging-Related Myocardial Apoptosis , 2018, The journals of gerontology. Series A, Biological sciences and medical sciences.
[50] H. Zhang,et al. Comprehensive genetic characteristics of dystrophinopathies in China , 2018, Orphanet Journal of Rare Diseases.
[51] Steven B Marston. The Molecular Mechanisms of Mutations in Actin and Myosin that Cause Inherited Myopathy , 2018, International journal of molecular sciences.
[52] K. Adachi,et al. Female dystrophinopathy: Review of current literature , 2018, Neuromuscular Disorders.
[53] Gang Bao,et al. A high-fidelity Cas9 mutant delivered as a ribonucleoprotein complex enables efficient gene editing in human haematopoietic stem and progenitor cells , 2018, Nature Medicine.
[54] G. Raposo,et al. Exosomes and extracellular vesicles: the path forward. , 2018, Essays in biochemistry.
[55] Rinat Meir,et al. Placenta-derived mesenchymal stromal cells and their exosomes exert therapeutic effects in Duchenne muscular dystrophy. , 2018, Biomaterials.
[56] O. Binah,et al. Generation of Duchenne muscular dystrophy patient-specific induced pluripotent stem cell line lacking exons 45-50 of the dystrophin gene (IITi001-A). , 2018, Stem cell research.
[57] K. Mamchaoui,et al. miR-708-5p and miR-34c-5p are involved in nNOS regulation in dystrophic context , 2018, Skeletal muscle.
[58] K. Mamchaoui,et al. miR-708-5p and miR-34c-5p are involved in nNOS regulation in dystrophic context , 2018, Skeletal Muscle.
[59] O. Binah,et al. Genome Editing in Induced Pluripotent Stem Cells using CRISPR/Cas9 , 2018, Stem Cell Reviews and Reports.
[60] N. Sniadecki,et al. Afterload promotes maturation of human induced pluripotent stem cell derived cardiomyocytes in engineered heart tissues. , 2018, Journal of molecular and cellular cardiology.
[61] S. Pandya,et al. Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and neuromuscular, rehabilitation, endocrine, and gastrointestinal and nutritional management , 2018, The Lancet Neurology.
[62] Malte Tiburcy,et al. Correction of diverse muscular dystrophy mutations in human engineered heart muscle by single-site genome editing , 2018, Science Advances.
[63] Joe Z. Zhang,et al. Molecular and functional resemblance of differentiated cells derived from isogenic human iPSCs and SCNT-derived ESCs , 2017, Proceedings of the National Academy of Sciences.
[64] G. Ronzitti,et al. Emerging Issues in AAV-Mediated In Vivo Gene Therapy , 2017, Molecular therapy. Methods & clinical development.
[65] H. Mandel,et al. Investigating the cardiac pathology of SCO2‐mediated hypertrophic cardiomyopathy using patients induced pluripotent stem cell–derived cardiomyocytes , 2017, Journal of cellular and molecular medicine.
[66] Z. Giricz,et al. Alternative Splicing of NOX4 in the Failing Human Heart , 2017, Front. Physiol..
[67] Dipali G. Sashital,et al. The Revolution Continues: Newly Discovered Systems Expand the CRISPR-Cas Toolkit. , 2017, Molecular cell.
[68] D. Ando,et al. Gene Editing: Regulatory and Translation to Clinic. , 2017, Hematology/oncology clinics of North America.
[69] R. Finkel,et al. Ataluren in patients with nonsense mutation Duchenne muscular dystrophy (ACT DMD): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial , 2017, The Lancet.
[70] Malte Tiburcy,et al. Functional correction of dystrophin actin binding domain mutations by genome editing. , 2017, JCI insight.
[71] D. Freimark,et al. CRISPR correction of the PRKAG2 gene mutation in the patient's induced pluripotent stem cell-derived cardiomyocytes eliminates electrophysiological and structural abnormalities. , 2017, Heart rhythm.
[72] B. Kennedy,et al. Genome editing: the breakthrough technology for inherited retinal disease? , 2017, Expert opinion on biological therapy.
[73] Dominique Rasoloson,et al. Precision genome editing using synthesis-dependent repair of Cas9-induced DNA breaks , 2017, Proceedings of the National Academy of Sciences.
[74] J. Kleijnen,et al. The burden, epidemiology, costs and treatment for Duchenne muscular dystrophy: an evidence review , 2017, Orphanet Journal of Rare Diseases.
[75] E. Olson,et al. Publication : CRISPR-Cpf1 correction of muscular dystrophy mutations in human cardiomyocytes and mice , 2018 .
[76] Hao Yin,et al. Delivery technologies for genome editing , 2017, Nature Reviews Drug Discovery.
[77] David R. Liu,et al. CRISPR-Based Technologies for the Manipulation of Eukaryotic Genomes , 2017, Cell.
[78] Anton Salykin,et al. Atomic force microscopy combined with human pluripotent stem cell derived cardiomyocytes for biomechanical sensing. , 2016, Biosensors & bioelectronics.
[79] Gaelen T. Hess,et al. Directed evolution using dCas9-targeted somatic hypermutation in mammalian cells , 2016, Nature Methods.
[80] M. Mahfouz,et al. Genome editing: the road of CRISPR/Cas9 from bench to clinic , 2016, Experimental & Molecular Medicine.
[81] K. Bielawski,et al. Real-Time Force and Frequency Analysis of Engineered Human Heart Tissue Derived from Induced Pluripotent Stem Cells Using Magnetic Sensing. , 2016, Tissue engineering. Part C, Methods.
[82] K. Hilber,et al. Decreased inward rectifier potassium current IK1 in dystrophin-deficient ventricular cardiomyocytes , 2016, Channels.
[83] H. Topaloglu,et al. Practice guideline update summary: Corticosteroid treatment of Duchenne muscular dystrophy , 2016, Neurology.
[84] Lei S. Qi,et al. CRISPR/Cas9 in Genome Editing and Beyond. , 2016, Annual review of biochemistry.
[85] M. Oshimura,et al. Concordant but Varied Phenotypes among Duchenne Muscular Dystrophy Patient-Specific Myoblasts Derived using a Human iPSC-Based Model. , 2016, Cell reports.
[86] Xiao-ming Meng,et al. TGF-β: the master regulator of fibrosis , 2016, Nature Reviews Nephrology.
[87] D. Garry,et al. Dystrophin-Deficient Cardiomyopathy. , 2016, Journal of the American College of Cardiology.
[88] F. Chrétien,et al. The Female mdx Mouse: An Unexpected Vascular Story , 2016 .
[89] A. Manzur,et al. Corticosteroids for the treatment of Duchenne muscular dystrophy. , 2016, The Cochrane database of systematic reviews.
[90] J. Keith Joung,et al. 731. High-Fidelity CRISPR-Cas9 Nucleases with No Detectable Genome-Wide Off-Target Effects , 2016 .
[91] David R. Liu,et al. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage , 2016, Nature.
[92] Atsushi Nakano,et al. A Single CRISPR-Cas9 Deletion Strategy that Targets the Majority of DMD Patients Restores Dystrophin Function in hiPSC-Derived Muscle Cells. , 2016, Cell stem cell.
[93] D. Mack,et al. Nicorandil, a Nitric Oxide Donor and ATP-Sensitive Potassium Channel Opener, Protects Against Dystrophin-Deficient Cardiomyopathy , 2016, Journal of cardiovascular pharmacology and therapeutics.
[94] Renzhi Han,et al. CRISPR-mediated Genome Editing Restores Dystrophin Expression and Function in mdx Mice. , 2016, Molecular therapy : the journal of the American Society of Gene Therapy.
[95] R. Moxley,et al. Practice guideline update summary: Corticosteroid treatment of Duchenne muscular dystrophy , 2016, Neurology.
[96] H. Kurahashi,et al. Next-generation sequencing discloses a nonsense mutation in the dystrophin gene from long preserved dried umbilical cord and low-level somatic mosaicism in the proband mother , 2016, Journal of Human Genetics.
[97] J. Joung,et al. High-fidelity CRISPR-Cas9 variants with undetectable genome-wide off-targets , 2015, Nature.
[98] B. Byrne,et al. Longitudinal effect of eteplirsen versus historical control on ambulation in Duchenne muscular dystrophy , 2015, Annals of neurology.
[99] Xuan Guan,et al. Nanopatterned Human iPSC-Based Model of a Dystrophin-Null Cardiomyopathic Phenotype , 2015, Cellular and molecular bioengineering.
[100] R. Janicek,et al. Mitochondrial dysfunctions during progression of dystrophic cardiomyopathy. , 2015, Cell calcium.
[101] J. Pu,et al. Common Variants in TRDN and CALM1 Are Associated with Risk of Sudden Cardiac Death in Chronic Heart Failure Patients in Chinese Han Population , 2015, PloS one.
[102] Michael Eldar,et al. Functional abnormalities in iPSC-derived cardiomyocytes generated from CPVT1 and CPVT2 patients carrying ryanodine or calsequestrin mutations , 2015, Journal of cellular and molecular medicine.
[103] Zhiping Tan,et al. Ehlers-Danlos syndrome type IV is associated with a novel G984R COL3A1 mutation. , 2015, Molecular medicine reports.
[104] E. Hoffman,et al. Contemporary Cardiac Issues in Duchenne Muscular Dystrophy , 2015, Circulation.
[105] G. Bett,et al. Modeling and study of the mechanism of dilated cardiomyopathy using induced pluripotent stem cells derived from individuals with Duchenne muscular dystrophy , 2015, Disease Models & Mechanisms.
[106] Katherine A James,et al. Prevalence of Duchenne and Becker Muscular Dystrophies in the United States , 2015, Pediatrics.
[107] E. Urbina,et al. Autonomic Dysfunction: A Driving Force for Myocardial Fibrosis in Young Duchenne Muscular Dystrophy Patients? , 2015, Pediatric Cardiology.
[108] N. Kouprina,et al. Recent advances in chromosome engineering , 2015, Chromosome Research.
[109] M. Oshimura,et al. A pathway from chromosome transfer to engineering resulting in human and mouse artificial chromosomes for a variety of applications to bio-medical challenges , 2015, Chromosome Research.
[110] Andrew D. Williams,et al. Defects in Mitochondrial ATP Synthesis in Dystrophin-Deficient Mdx Skeletal Muscles May Be Caused by Complex I Insufficiency , 2014, PloS one.
[111] D. Pisano,et al. Telomerase expression confers cardioprotection in the adult mouse heart after acute myocardial infarction , 2014, Nature Communications.
[112] Frédérique Magdinier,et al. Telomere position effect: regulation of gene expression with progressive telomere shortening over long distances , 2014, Genes & development.
[113] Jan Vrbsky,et al. Forced aggregation and defined factors allow highly uniform-sized embryoid bodies and functional cardiomyocytes from human embryonic and induced pluripotent stem cells , 2014, Heart and Vessels.
[114] J. Renaud,et al. Characterization of Dystrophin Deficient Rats: A New Model for Duchenne Muscular Dystrophy , 2014, PloS one.
[115] Xue Zhang,et al. Screening of Duchenne Muscular Dystrophy (DMD) Mutations and Investigating Its Mutational Mechanism in Chinese Patients , 2014, PloS one.
[116] A. De Luca,et al. Gene expression in mdx mouse muscle in relation to age and exercise: aberrant mechanical-metabolic coupling and implications for pre-clinical studies in Duchenne muscular dystrophy. , 2014, Human molecular genetics.
[117] A. Jacobsen,et al. miR-339-5p regulates the p53 tumor-suppressor pathway by targeting MDM2 , 2014, Oncogene.
[118] E. Marbán,et al. Exosomes as Critical Agents of Cardiac Regeneration Triggered by Cell Therapy , 2014, Stem cell reports.
[119] D. Stapleton,et al. Dysfunctional Muscle and Liver Glycogen Metabolism in mdx Dystrophic Mice , 2014, PloS one.
[120] H. Ruohola-Baker,et al. Dystrophin-deficient cardiomyocytes derived from human urine: new biologic reagents for drug discovery. , 2014, Stem cell research.
[121] G. Obermair,et al. Enhanced currents through L-type calcium channels in cardiomyocytes disturb the electrophysiology of the dystrophic heart. , 2014, American journal of physiology. Heart and circulatory physiology.
[122] N. Elvassore,et al. Complete restoration of multiple dystrophin isoforms in genetically corrected Duchenne muscular dystrophy patient–derived cardiomyocytes , 2014, Molecular therapy. Methods & clinical development.
[123] Alexandra M. Greiner,et al. Cyclic Tensile Strain Controls Cell Shape and Directs Actin Stress Fiber Formation and Focal Adhesion Alignment in Spreading Cells , 2013, PloS one.
[124] David A. Scott,et al. Genome engineering using the CRISPR-Cas9 system , 2013, Nature Protocols.
[125] S. Kaveri,et al. Development of inhibitory antibodies to therapeutic factor VIII in severe hemophilia A is associated with microsatellite polymorphisms in the HMOX1 promoter , 2013, Haematologica.
[126] Natsuyo Aoyama,et al. Phenotypic Screening with Human iPS Cell–Derived Cardiomyocytes , 2013, Journal of biomolecular screening.
[127] David A. Scott,et al. Double Nicking by RNA-Guided CRISPR Cas9 for Enhanced Genome Editing Specificity , 2013, Cell.
[128] Hanns Lochmüller,et al. Exon skipping and gene transfer restore dystrophin expression in human induced pluripotent stem cells-cardiomyocytes harboring DMD mutations. , 2013, Stem cells and development.
[129] Min Zhang,et al. NADPH oxidases in heart failure: poachers or gamekeepers? , 2013, Antioxidants & redox signaling.
[130] N. Dahdah,et al. All-cause mortality and cardiovascular outcomes with prophylactic steroid therapy in Duchenne muscular dystrophy. , 2013, Journal of the American College of Cardiology.
[131] S. Raimondo,et al. Exosomes as Intercellular Signaling Organelles Involved in Health and Disease: Basic Science and Clinical Applications , 2013, International journal of molecular sciences.
[132] Rachel Salmon,et al. Newborn bloodspot screening for Duchenne Muscular Dystrophy: 21 years experience in Wales (UK) , 2013, European Journal of Human Genetics.
[133] R. Schleip,et al. The role of fibrosis in Duchenne muscular dystrophy , 2012, Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology.
[134] G. Piluso,et al. Improvement of survival in Duchenne Muscular Dystrophy: retrospective analysis of 835 patients , 2012, Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology.
[135] K. Serizawa,et al. Nicorandil prevents endothelial dysfunction due to antioxidative effects via normalisation of NADPH oxidase and nitric oxide synthase in streptozotocin diabetic rats , 2011, Cardiovascular diabetology.
[136] Kumaraswamy Nanthakumar,et al. Role of KATP Channels in the Maintenance of Ventricular Fibrillation in Cardiomyopathic Human Hearts , 2011, Circulation research.
[137] D. Kass,et al. Pathophysiology and Therapy of Cardiac Dysfunction in Duchenne Muscular Dystrophy , 2011, American journal of cardiovascular drugs : drugs, devices, and other interventions.
[138] T. Okano,et al. Hippo pathway regulation by cell morphology and stress fibers , 2011, Development.
[139] J. Bourke,et al. Exon skipping and dystrophin restoration in patients with Duchenne muscular dystrophy after systemic phosphorodiamidate morpholino oligomer treatment: an open-label, phase 2, dose-escalation study , 2011, The Lancet.
[140] Samira Kiani,et al. Genetic engineering of human ES and iPS cells using TALE nucleases , 2011, Nature Biotechnology.
[141] Susan Carpenter,et al. Modularly assembled designer TAL effector nucleases for targeted gene knockout and gene replacement in eukaryotes , 2011, Nucleic acids research.
[142] Elo Leung,et al. A TALE nuclease architecture for efficient genome editing , 2011, Nature Biotechnology.
[143] D. Cacchiarelli,et al. miR‐31 modulates dystrophin expression: new implications for Duchenne muscular dystrophy therapy , 2011, EMBO reports.
[144] S. Delp,et al. Short Telomeres and Stem Cell Exhaustion Model Duchenne Muscular Dystrophy in mdx/mTR Mice , 2010, Cell.
[145] D. Duan,et al. Gender influences cardiac function in the mdx model of duchenne cardiomyopathy , 2010, Muscle & nerve.
[146] E. McNally,et al. Cardiac Assessment in Duchenne and Becker Muscular Dystrophies , 2010, Current heart failure reports.
[147] Richard A Young,et al. Chromatin structure and gene expression programs of human embryonic and induced pluripotent stem cells. , 2010, Cell stem cell.
[148] J. Bach,et al. Duchenne Muscular Dystrophy: The Effect of Glucocorticoids on Ventilator Use and Ambulation , 2010, American journal of physical medicine & rehabilitation.
[149] Zhujun Zhang,et al. Mutation spectrum of the dystrophin gene in 442 Duchenne/Becker muscular dystrophy cases from one Japanese referral center , 2010, Journal of Human Genetics.
[150] J. Sadoshima,et al. Upregulation of Nox4 by Hypertrophic Stimuli Promotes Apoptosis and Mitochondrial Dysfunction in Cardiac Myocytes , 2010, Circulation research.
[151] Hongli,et al. Upregulation of Nox4 by Hypertrophic Stimuli Promotes Apoptosis and Mitochondrial Dysfunction in Cardiac Myocytes , 2010 .
[152] S. Matecki,et al. Leaky RyR2 trigger ventricular arrhythmias in Duchenne muscular dystrophy , 2010, Proceedings of the National Academy of Sciences.
[153] M. Oshimura,et al. Complete Genetic Correction of iPS Cells From Duchenne Muscular Dystrophy , 2009, Molecular therapy : the journal of the American Society of Gene Therapy.
[154] Livija Medne,et al. Mutational spectrum of DMD mutations in dystrophinopathy patients: application of modern diagnostic techniques to a large cohort , 2009, Human mutation.
[155] B. Aronow,et al. Calcium influx is sufficient to induce muscular dystrophy through a TRPC-dependent mechanism , 2009, Proceedings of the National Academy of Sciences.
[156] Robin Goland,et al. Generation of pluripotent stem cells from patients with type 1 diabetes , 2009, Proceedings of the National Academy of Sciences.
[157] F. Leturcq,et al. Endomysial Fibrosis in Duchenne Muscular Dystrophy: A Marker of Poor Outcome Associated With Macrophage Alternative Activation , 2009, Journal of neuropathology and experimental neurology.
[158] P. Mattila,et al. Contractility-dependent actin dynamics in cardiomyocyte sarcomeres , 2009, Journal of Cell Science.
[159] A. Grant,et al. Cardiac ion channels. , 2009, Circulation. Arrhythmia and electrophysiology.
[160] Rudolf Jaenisch,et al. Parkinson's Disease Patient-Derived Induced Pluripotent Stem Cells Free of Viral Reprogramming Factors , 2009, Cell.
[161] J. Tidball,et al. NO may prompt calcium leakage in dystrophic muscle , 2009, Nature Medicine.
[162] G. van Ommen,et al. Theoretic applicability of antisense‐mediated exon skipping for Duchenne muscular dystrophy mutations , 2009, Human mutation.
[163] S. Matecki,et al. Hypernitrosylated ryanodine receptor/calcium release channels are leaky in dystrophic muscle , 2009, Nature Medicine.
[164] E. Niggli,et al. Pathways of abnormal stress-induced Ca2+ influx into dystrophic mdx cardiomyocytes. , 2009, Cell calcium.
[165] G. Thiene,et al. Journal of Cardiovascular Magnetic Resonance Open Access Cardiac Involvement in Patients with Becker Muscular Dystrophy: New Diagnostic and Pathophysiological Insights by a Cmr Approach , 2022 .
[166] J. Finsterer,et al. Cardiac involvement in Becker muscular dystrophy. , 2008, The Canadian journal of cardiology.
[167] Hynek Wichterle,et al. Induced Pluripotent Stem Cells Generated from Patients with ALS Can Be Differentiated into Motor Neurons , 2008, Science.
[168] E. Hoffman,et al. Dystrophin-deficient cardiomyopathy in mouse: Expression of Nox4 and Lox are associated with fibrosis and altered functional parameters in the heart , 2008, Neuromuscular Disorders.
[169] E. Niggli,et al. Dystrophic cardiomyopathy: amplification of cellular damage by Ca2+ signalling and reactive oxygen species-generating pathways. , 2008, Cardiovascular research.
[170] T. Ichisaka,et al. Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors , 2007, Cell.
[171] John Bourke,et al. Managing Duchenne muscular dystrophy – The additive effect of spinal surgery and home nocturnal ventilation in improving survival , 2007, Neuromuscular Disorders.
[172] S. Yamanaka,et al. Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors , 2006, Cell.
[173] Patrick Ruchat,et al. Cardiac Sodium Channel Nav1.5 Is Regulated by a Multiprotein Complex Composed of Syntrophins and Dystrophin , 2006, Circulation research.
[174] G. van Ommen,et al. Entries in the Leiden Duchenne muscular dystrophy mutation database: An overview of mutation types and paradoxical cases that confirm the reading‐frame rule , 2006, Muscle & nerve.
[175] Kenneth R. Laurita,et al. Cellular Mechanism of Calcium-Mediated Triggered Activity in the Heart , 2005, Circulation research.
[176] B. Wong,et al. Evolution of the mdx mouse cardiomyopathy: physiological and morphological findings , 2004, Neuromuscular Disorders.
[177] Teiji Wada,et al. Mitogen-activated protein kinases in apoptosis regulation , 2004, Oncogene.
[178] Yuan-Ping Pang,et al. ABCC9 mutations identified in human dilated cardiomyopathy disrupt catalytic KATP channel gating , 2004, Nature Genetics.
[179] J. D. Porter,et al. Temporal gene expression profiling of dystrophin-deficient (mdx) mouse diaphragm identifies conserved and muscle group-specific mechanisms in the pathogenesis of muscular dystrophy. , 2004, Human molecular genetics.
[180] Mei Han,et al. Gene expression profiling of Duchenne muscular dystrophy skeletal muscle , 2003, Neurogenetics.
[181] L. Kunkel,et al. Gene expression comparison of biopsies from Duchenne muscular dystrophy (DMD) and normal skeletal muscle , 2002, Proceedings of the National Academy of Sciences of the United States of America.
[182] H. Debaix,et al. Involvement of TRPC in the abnormal calcium influx observed in dystrophic (mdx) mouse skeletal muscle fibers , 2002, The Journal of cell biology.
[183] B. Allard,et al. Ca(2+) influx and opening of Ca(2+)-activated K(+) channels in muscle fibers from control and mdx mice. , 2002, Biophysical journal.
[184] Andrew P. Weir,et al. Function and genetics of dystrophin and dystrophin-related proteins in muscle. , 2002, Physiological reviews.
[185] S. Smaili,et al. Bax translocation to mitochondria subsequent to a rapid loss of mitochondrial membrane potential , 2001, Cell Death and Differentiation.
[186] M. Entman,et al. Telomerase reverse transcriptase promotes cardiac muscle cell proliferation, hypertrophy, and survival , 2001, Proceedings of the National Academy of Sciences of the United States of America.
[187] T. Pozzan,et al. Alteration in Calcium Handling at the Subcellular Level inmdx Myotubes* , 2001, The Journal of Biological Chemistry.
[188] J. Ervasti,et al. The Dystrophin Complex Forms a Mechanically Strong Link between the Sarcolemma and Costameric Actin , 2000, The Journal of cell biology.
[189] Robert L Moritz,et al. Identification of DIABLO, a Mammalian Protein that Promotes Apoptosis by Binding to and Antagonizing IAP Proteins , 2000, Cell.
[190] K. Fujiwara,et al. Scavenging effect of nicorandil on free radicals and lipid peroxide in streptozotocin-induced diabetic rats. , 2000, Metabolism: clinical and experimental.
[191] R. Steinhardt,et al. Calcium Influx through Calcium Leak Channels Is Responsible for the Elevated Levels of Calcium-dependent Proteolysis in Dystrophic Myotubes* , 2000, The Journal of Biological Chemistry.
[192] K. Campbell,et al. Dystrophic phenotype induced in vitro by antibody blockade of muscle alpha-dystroglycan-laminin interaction. , 1999, Journal of cell science.
[193] D. Bredt,et al. Interaction of Nitric Oxide Synthase with the Postsynaptic Density Protein PSD-95 and α1-Syntrophin Mediated by PDZ Domains , 1996, Cell.
[194] J. Tidball,et al. Calpains Are Activated in Necrotic Fibers from mdx Dystrophic Mice (*) , 1995, The Journal of Biological Chemistry.
[195] H. Klamut,et al. The human dystrophin gene requires 16 hours to be transcribed and is cotranscriptionally spliced , 1995, Nature Genetics.
[196] L. Kunkel,et al. The structural and functional diversity of dystrophin , 1993, Nature Genetics.
[197] A. Jauch,et al. Somatic mosaicism for a deletion of the dystrophin gene in a carrier of Becker muscular dystrophy , 1992, European Journal of Pediatrics.
[198] R. Strohman,et al. Fiber regeneration is not persistent in dystrophic (MDX) mouse skeletal muscle. , 1991, Developmental biology.
[199] A. Pestronk,et al. Prednisone in Duchenne dystrophy. A randomized, controlled trial defining the time course and dose response. Clinical Investigation of Duchenne Dystrophy Group. , 1991, Archives of neurology.
[200] W. Denetclaw,et al. Increased activity of calcium leak channels in myotubes of Duchenne human and mdx mouse origin. , 1990, Science.
[201] N. Taira,et al. Nicorandil as a hybrid between nitrates and potassium channel activators. , 1989, The American journal of cardiology.
[202] R. Steinhardt,et al. Increased protein degradation results from elevated free calcium levels found in muscle from mdx mice , 1988, Nature.
[203] D. Shotton,et al. Muscular dystrophy in the mdx mouse: Histopathology of the soleus and extensor digitorum longus muscles , 1987, Journal of the Neurological Sciences.
[204] G. Vrbóva,et al. Muscle development in mdx mutant mice , 1984, Muscle & nerve.
[205] K. Moore,et al. X chromosome-linked muscular dystrophy (mdx) in the mouse. , 1984, Proceedings of the National Academy of Sciences of the United States of America.
[206] J. Bodensteiner,et al. Intracellular calcium accumulation in Duchenne dystrophy and other myopathies , 1978, Neurology.
[207] N. Sniadecki,et al. Engineered Heart Tissues for Contractile, Structural, and Transcriptional Assessment of Human Pluripotent Stem Cell-Derived Cardiomyocytes in a Three-Dimensional, Auxotonic Environment. , 2022, Methods in molecular biology.
[208] Gaetano Santulli,et al. New Insights in Cardiac Calcium Handling and Excitation-Contraction Coupling. , 2018, Advances in experimental medicine and biology.
[209] D. Sorriento,et al. Functional Role of Mitochondria in Arrhythmogenesis. , 2017, Advances in experimental medicine and biology.
[210] Z. Giricz,et al. Alternative Splicing of NOX 4 in the Failing Human Heart , 2017 .
[211] C. Lutz,et al. Effect of genetic background on the dystrophic phenotype in mdx mice. , 2016, Human molecular genetics.
[212] John K. Hall,et al. Animal models of muscular dystrophy. , 2012, Progress in molecular biology and translational science.
[213] C. Bourcier-Lucas,et al. Stress-induced opening of the permeability transition pore in the dystrophin-deficient heart is attenuated by acute treatment with sildenafil. , 2011, American journal of physiology. Heart and circulatory physiology.
[214] D. Annane,et al. Cardiomyopathy in Duchenne muscular dystrophy: pathogenesis and therapeutics , 2009, Heart Failure Reviews.
[215] M. Bianchi,et al. Dystroglycan and muscular dystrophies related to the dystrophin-glycoprotein complex. , 2003, Annali dell'Istituto superiore di sanita.
[216] T. Kawada,et al. Dystrophin disruption might be related to myocardial cell apoptosis caused by isoproterenol. , 2000, Journal of cardiovascular pharmacology.
[217] A. Monaco,et al. An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. , 1988, Genomics.