Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Biol Open ; 13(9)2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39158383

ABSTRACT

Duchenne muscular dystrophy (DMD) is a progressive muscle wasting disorder affecting 1:3500 male births and is associated with myofiber degeneration, regeneration, and inflammation. Glucocorticoid treatments have been the standard of care due to immunomodulatory/immunosuppressive properties but novel genetic approaches, including exon skipping and gene replacement therapy, are currently being developed. The identification of additional biomarkers to assess DMD-related inflammatory responses and the potential efficacy of these therapeutic approaches are thus of critical importance. The current study uses RNA sequencing of skeletal muscle from two mdx mouse models to identify high mobility group box 1 (HMGB1) as a candidate biomarker potentially contributing to DMD-related inflammation. HMGB1 protein content was increased in a human iPSC-derived skeletal myocyte model of DMD and microdystrophin treatment decreased HMGB1 back to control levels. In vivo, HMGB1 protein levels were increased in vehicle treated B10-mdx skeletal muscle compared to B10-WT and significantly decreased in B10-mdx animals treated with adeno-associated virus (AAV)-microdystrophin. However, HMGB1 protein levels were not increased in D2-mdx skeletal muscle compared to D2-WT, demonstrating a strain-specific difference in DMD-related immunopathology.


Subject(s)
Biomarkers , Disease Models, Animal , HMGB1 Protein , Mice, Inbred mdx , Muscular Dystrophy, Duchenne , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/genetics , Animals , HMGB1 Protein/metabolism , HMGB1 Protein/genetics , Mice , Humans , Muscle, Skeletal/metabolism , Dystrophin/metabolism , Dystrophin/genetics , Induced Pluripotent Stem Cells/metabolism , Male
2.
Exp Cell Res ; 424(2): 113507, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36796746

ABSTRACT

Nemaline myopathies (NM) are a group of congenital myopathies that lead to muscle weakness and dysfunction. While 13 genes have been identified to cause NM, over 50% of these genetic defects are due to mutations in nebulin (NEB) and skeletal muscle actin (ACTA1), which are genes required for normal assembly and function of the thin filament. NM can be distinguished on muscle biopsies due to the presence of nemaline rods, which are thought to be aggregates of the dysfunctional protein. Mutations in ACTA1 have been associated with more severe clinical disease and muscle weakness. However, the cellular pathogenesis linking ACTA1 gene mutations to muscle weakness are unclear To evaluate cellular disease phenotypes, iPSC-derived skeletal myocytes (iSkM) harboring an ACTA1 H40Y point mutation were used to model NM in skeletal muscle. These were generated by Crispr-Cas9, and include one non-affected healthy control (C) and 2 NM iPSC clone lines, therefore representing isogenic controls. Fully differentiated iSkM were characterized to confirm myogenic status and subject to assays to evaluate nemaline rod formation, mitochondrial membrane potential, mitochondrial permeability transition pore (mPTP) formation, superoxide production, ATP/ADP/phosphate levels and lactate dehydrogenase release. C- and NM-iSkM demonstrated myogenic commitment as evidenced by mRNA expression of Pax3, Pax7, MyoD, Myf5 and Myogenin; and protein expression of Pax4, Pax7, MyoD and MF20. No nemaline rods were observed with immunofluorescent staining of NM-iSkM for ACTA1 or ACTN2, and these mRNA transcript and protein levels were comparable to C-iSkM. Mitochondrial function was altered in NM, as evidenced by decreased cellular ATP levels and altered mitochondrial membrane potential. Oxidative stress induction revealed the mitochondrial phenotype, as evidenced by collapsed mitochondrial membrane potential, early formation of the mPTP and increased superoxide production. Early mPTP formation was rescued with the addition of ATP to media. Together, these findings suggest that mitochondrial dysfunction and oxidative stress are disease phenotypes in the in vitro model of ACTA1 nemaline myopathy, and that modulation of ATP levels was sufficient to protect NM-iSkM mitochondria from stress-induced injury. Importantly, the nemaline rod phenotype was absent in our in vitro model of NM. We conclude that this in vitro model has the potential to recapitulate human NM disease phenotypes, and warrants further study.


Subject(s)
Induced Pluripotent Stem Cells , Myopathies, Nemaline , Humans , Myopathies, Nemaline/genetics , Myopathies, Nemaline/pathology , Induced Pluripotent Stem Cells/metabolism , Superoxides/metabolism , Muscle, Skeletal/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Weakness/genetics , Muscle Weakness/pathology , Actins/genetics , Actins/metabolism , Mutation , Mitochondria/metabolism , Adenosine Triphosphate/metabolism
3.
J Cardiovasc Pharmacol Ther ; 27: 10742484221088655, 2022.
Article in English | MEDLINE | ID: mdl-35353647

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is a neuromuscular disease caused by dystrophin gene mutations affecting striated muscle. Due to advances in skeletal muscle treatment, cardiomyopathy has emerged as a leading cause of death. Previously, nicorandil, a drug with antioxidant and nitrate-like properties, ameliorated cardiac damage and improved cardiac function in young, injured mdx mice. Nicorandil mitigated damage by stimulating antioxidant activity and limiting pro-oxidant expression. Here, we examined whether nicorandil was similarly cardioprotective in aged mdx mice. METHODS AND RESULTS: Nicorandil (6 mg/kg) was given over 15 months. Echocardiography of mdx mice showed some functional defects at 12 months compared to wild-type (WT) mice, but not at 15 months. Disease manifestation was evident in mdx mice via treadmill assays and survival, but not open field and grip strength assays. Cardiac levels of SOD2 and NOX4 were decreased in mdx vs. WT. Nicorandil increased survival in mdx but did not alter cardiac function, fibrosis, diaphragm function or muscle fatigue. CONCLUSIONS: In contrast to our prior work in young, injured mdx mice, nicorandil did not exert cardioprotective effects in 15 month aged mdx mice. Discordant findings may be explained by the lack of cardiac disease manifestation in aged mdx mice compared to WT, whereas significant cardiac dysfunction was previously seen with the sub-acute injury in young mice. Therefore, we are not able to conclude any cardioprotective effects with long-term nicorandil treatment in aging mdx mice.


Subject(s)
Cardiomyopathies , Muscular Dystrophy, Duchenne , Animals , Cardiomyopathies/drug therapy , Cardiomyopathies/etiology , Cardiomyopathies/prevention & control , Heart , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Nicorandil/pharmacology
4.
Methods Mol Biol ; 2319: 51-60, 2021.
Article in English | MEDLINE | ID: mdl-34331242

ABSTRACT

Cardiovascular disease is a worldwide health issue that affects millions of lives every year, and thus, researchers are in need of high-throughput model systems with which to investigate mechanisms of disease and to develop and test potential therapies. The use of human-derived induced pluripotent stem cells (iPSCs) differentiated into cardiomyocytes aims to address this need. While cardiac differentiation protocols have been established previously in iPSCs, optimization of cardiac differentiation remains crucial to obtaining high quality cardiomyocytes for future experimental analyses. Important factors to consider include cell density and rate of proliferation, temporal regulation of media changes throughout the differentiation process, and the concentration of the chemicals utilized. In this chapter, we present a detailed protocol to outline the process of differentiating cardiomyocytes from human iPSCs via modulation of Wnt signaling, characterization of cardiomyocytes by immunofluorescence, as well as guidelines for troubleshooting and optimizing these techniques.


Subject(s)
Cell Culture Techniques/methods , Culture Media/chemistry , Induced Pluripotent Stem Cells/cytology , Muscle Development , Myocytes, Cardiac/cytology , Wnt Signaling Pathway , Fluorescent Antibody Technique , Humans , In Vitro Techniques , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism
5.
Hum Mol Genet ; 30(23): 2347-2361, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34270708

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked genetic disease characterized by severe, progressive muscle wasting. Cardiomyopathy has emerged as a leading cause of death in patients with DMD. The mechanisms contributing to DMD cardiac disease remain under investigation and specific therapies available are lacking. Our prior work has shown that DMD-iPSC-derived cardiomyocytes (DMD-iCMs) are vulnerable to oxidative stress injury and chronic exposure to DMD-secreted exosomes impaired the cell's ability to protect against stress. In this study, we sought to examine a mechanism by which DMD cardiac exosomes impair cellular response through altering important stress-responsive genes in the recipient cells. Here, we report that DMD-iCMs secrete exosomes containing altered microRNA (miR) profiles in comparison to healthy controls. In particular, miR-339-5p was upregulated in DMD-iCMs, DMD exosomes and mdx mouse cardiac tissue. Restoring dystrophin in DMD-iCMs improved the cellular response to stress and was associated with downregulation of miR-339-5p, suggesting that it is disease-specific. Knockdown of miR-339-5p was associated with increased expression of MDM2, GSK3A and MAP2K3, which are genes involved in important stress-responsive signaling pathways. Finally, knockdown of miR-339-5p led to mitochondrial protection and a reduction in cell death in DMD-iCMs, indicating miR-339-5p is involved in direct modulation of stress-responsiveness. Together, these findings identify a potential mechanism by which exosomal miR-339-5p may be modulating cell signaling pathways that are important for robust stress responses. Additionally, these exosomal miRs may provide important disease-specific targets for future therapeutic advancements for the management and diagnosis of DMD cardiomyopathy.


Subject(s)
Cardiomyopathies/genetics , MicroRNAs/genetics , Muscular Dystrophy, Duchenne/complications , Myocytes, Cardiac/metabolism , Biomarkers , Cardiomyopathies/diagnosis , Disease Susceptibility , Dystrophin/genetics , Exosomes/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Stress, Physiological
6.
Dis Model Mech ; 13(11)2020 11 13.
Article in English | MEDLINE | ID: mdl-33188007

ABSTRACT

Cardiomyopathy is a leading cause of early mortality in Duchenne muscular dystrophy (DMD). There is a need to gain a better understanding of the molecular pathogenesis for the development effective therapies. Exosomes (exo) are secreted vesicles and exert effects via their RNA, lipid and protein cargo. The role of exosomes in disease pathology is unknown. Exosomes derived from stem cells have demonstrated cardioprotection in the murine DMD heart. However, it is unknown how the disease status of the donor cell type influences exosome function. Here, we sought to determine the phenotypic responses of DMD cardiomyocytes (DMD-iCMs) after long-term exposure to DMD cardiac exosomes (DMD-exo). DMD-iCMs were vulnerable to stress, evidenced by production of reactive oxygen species, the mitochondrial membrane potential and cell death levels. Long-term exposure to non-affected exosomes (N-exo) was protective. By contrast, long-term exposure to DMD-exo was not protective, and the response to stress improved with inhibition of DMD-exo secretion in vitro and in vivo The microRNA (miR) cargo, but not exosome surface peptides, was implicated in the pathological effects of DMD-exo. Exosomal surface profiling revealed N-exo peptides associated with PI3K-Akt signaling. Transcriptomic profiling identified unique changes with exposure to either N- or DMD-exo. Furthermore, DMD-exo miR cargo regulated injurious pathways, including p53 and TGF-beta. The findings reveal changes in exosomal cargo between healthy and diseased states, resulting in adverse outcomes. Here, DMD-exo contained miR changes, which promoted the vulnerability of DMD-iCMs to stress. Identification of these molecular changes in exosome cargo and effectual phenotypes might shed new light on processes underlying DMD cardiomyopathy.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Cardiomyopathies/pathology , Exosomes/metabolism , Muscular Dystrophy, Duchenne/pathology , Myocytes, Cardiac/metabolism , Animals , Cardiotonic Agents/metabolism , Cell Death , Cell Line , Female , Humans , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Myocytes, Cardiac/pathology , Paracrine Communication , Proteome/metabolism , Stress, Physiological , Transcription, Genetic
7.
J Muscle Res Cell Motil ; 41(4): 269-284, 2020 12.
Article in English | MEDLINE | ID: mdl-31564031

ABSTRACT

microRNAs are short, (18-22 nt) non-coding RNAs involved in important cellular processes due to their ability to regulate gene expression at the post-transcriptional level. Exosomes are small (50-200 nm) extracellular vesicles, naturally secreted from a variety of living cells and are believed to mediate cell-cell communication through multiple mechanisms, including uptake in destination cells. Circulating microRNAs and exosome-derived microRNAs can have key roles in regulating muscle cell development and differentiation. Several microRNAs are highly expressed in muscle and their regulation is important for myocyte homeostasis. Changes in muscle associated microRNA expression are associated with muscular diseases including muscular dystrophies, inflammatory myopathies, and congenital myopathies. In this review, we aim to highlight the biology of microRNAs and exosomes as well as their roles in muscle health and diseases. We also discuss the potential crosstalk between skeletal and cardiac muscle through exosomes and their contents.


Subject(s)
Exosomes/metabolism , MicroRNAs/genetics , Muscles/physiopathology , Humans
8.
Sci Rep ; 8(1): 16519, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30410044

ABSTRACT

As mediators of intercellular communication, exosomes containing molecular cargo are secreted by cells and taken up by recipient cells to influence cellular phenotype and function. Here we have investigated the effects of exosomes in dystrophin-deficient (Dys) induced pluripotent stem cell derived cardiomyocytes (iCMs). Our data demonstrate that exosomes secreted from either wild type (WT) or Dys-iCMs protect the Dys-iCM from stress-induced injury by decreasing reactive oxygen species and delaying mitochondrial permeability transition pore opening to maintain the mitochondrial membrane potential and decrease cell death. The protective effects of exosomes were dependent on the presence of exosomal surface proteins and activation of ERK1/2 and p38 MAPK signaling. Based on our findings, the acute effects of exosomes on recipient cells can be initiated from exosome membrane proteins and not necessarily their internal cargo.


Subject(s)
Dystrophin/deficiency , Exosomes/metabolism , Induced Pluripotent Stem Cells/cytology , MAP Kinase Signaling System , Mitochondria/metabolism , Myocytes, Cardiac/cytology , Cell Differentiation , Cell Survival , Cells, Cultured , Dystrophin/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Membrane Potential, Mitochondrial , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Sequence Deletion , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Stem Cell Res ; 18: 33-36, 2017 01.
Article in English | MEDLINE | ID: mdl-28395799

ABSTRACT

We have generated a human induced pluripotent stem cell (iPSC) line under feeder-free culture conditions using the urine derived cells (UCs) collected from non-affected control subjects to use as a comparison group for the iPSC lines containing a Plasminogen Activator Inhibitor-1 (PAI-1 homozygous/heterozygous) mutation. The Sendai Virus (SeV) vector encoding pluripotent Yamanaka transcription factors was used at a low multiplicity of infection to reprogram the UCs.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Urine/cytology , Base Sequence , Cell Culture Techniques/methods , Cell Line , Embryoid Bodies/metabolism , Embryoid Bodies/pathology , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Genotype , Heterozygote , Homozygote , Humans , Induced Pluripotent Stem Cells/metabolism , Karyotype , Microscopy, Fluorescence , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Sendai virus/genetics , Sequence Analysis, DNA , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Stem Cell Res ; 18: 41-44, 2017 01.
Article in English | MEDLINE | ID: mdl-28395801

ABSTRACT

We have generated a human induced pluripotent stem cell (iPSC) line under feeder-free culture conditions using the urine derived cells (UCs) collected from subjects heterozygous for a novel Plasminogen Activator Inhibitor-1 (PAI-1) mutation. The Sendai Virus (SeV) vector encoding pluripotent Yamanaka transcription factors was used at a low multiplicity of infection to reprogram the PAI-1 UCs.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Plasminogen Activator Inhibitor 1/genetics , Urine/cytology , Cell Culture Techniques , Cell Differentiation , Cell Line , Embryoid Bodies/metabolism , Embryoid Bodies/pathology , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Heterozygote , Humans , Induced Pluripotent Stem Cells/metabolism , Karyotype , Microscopy, Fluorescence , Polymorphism, Genetic , Sendai virus/genetics
12.
J Cardiovasc Transl Res ; 10(3): 295-304, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28032312

ABSTRACT

Heart failure with preserved left ventricular ejection fraction (HFpEF) has emerged as one of the largest unmet needs in cardiovascular medicine. HFpEF is increasing in prevalence and causes significant morbidity, mortality, and health care resource utilization. Patients have multiple co-morbidities which contribute to the disease complexity. To date, no effective treatment for HFpEF has been identified. The paucity of cardiac biopsies from this patient population and the absence of well-accepted animal models limit our understanding of the underlying molecular mechanisms of HFpEF. In this review, we discuss combining state-of-the-art technologies of microRNA profiling and human induced pluripotent cell-derived cardiomyocytes (iPSC-CMs) in order to uncover novel molecular pathways that may contribute to the development of HFpEF. Here, we focus the advantages and limitations of microRNA profiling and iPSC-CMs as a disease model system to discover molecular mechanisms in HFpEF.


Subject(s)
Heart Failure/metabolism , Induced Pluripotent Stem Cells/metabolism , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , Stroke Volume , Animals , Cell Line , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Induced Pluripotent Stem Cells/pathology , MicroRNAs/genetics , Myocytes, Cardiac/pathology , Phenotype , Signal Transduction
13.
Stem Cell Res ; 17(3): 657-660, 2016 11.
Article in English | MEDLINE | ID: mdl-27934602

ABSTRACT

We have generated a human induced pluripotent stem cell (iPSC) line under feeder-free culture conditions using the urine derived cells (UCs) collected from subject with a novel homozygous Plasminogen Activator Inhibitor-1 (PAI-1 null) mutation. The Sendai virus (SeV) vector encoding pluripotent Yamanaka transcription factors was used at a low multiplicity of infection to reprogram the PAI-1 UCs.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Plasminogen Activator Inhibitor 1/genetics , Base Sequence , Cell Differentiation , Cell Line , Cellular Reprogramming , DNA Mutational Analysis , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Homozygote , Humans , Induced Pluripotent Stem Cells/metabolism , Karyotype , Microscopy, Fluorescence , Mutagenesis, Insertional , Sendai virus/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Urine/cytology
SELECTION OF CITATIONS
SEARCH DETAIL