Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 133
Filter
1.
Cardiovasc Res ; 120(6): 630-643, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38230606

ABSTRACT

AIMS: Human pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) provide a platform to identify and characterize factors that regulate the maturation of CMs. The transition from an immature foetal to an adult CM state entails coordinated regulation of the expression of genes involved in myofibril formation and oxidative phosphorylation (OXPHOS) among others. Lysine demethylase 5 (KDM5) specifically demethylates H3K4me1/2/3 and has emerged as potential regulators of expression of genes involved in cardiac development and mitochondrial function. The purpose of this study is to determine the role of KDM5 in iPSC-CM maturation. METHODS AND RESULTS: KDM5A, B, and C proteins were mainly expressed in the early post-natal stages, and their expressions were progressively downregulated in the post-natal CMs and were absent in adult hearts and CMs. In contrast, KDM5 proteins were persistently expressed in the iPSC-CMs up to 60 days after the induction of myogenic differentiation, consistent with the immaturity of these cells. Inhibition of KDM5 by KDM5-C70 -a pan-KDM5 inhibitor, induced differential expression of 2372 genes, including upregulation of genes involved in fatty acid oxidation (FAO), OXPHOS, and myogenesis in the iPSC-CMs. Likewise, genome-wide profiling of H3K4me3 binding sites by the cleavage under targets and release using nuclease assay showed enriched of the H3K4me3 peaks at the promoter regions of genes encoding FAO, OXPHOS, and sarcomere proteins. Consistent with the chromatin and gene expression data, KDM5 inhibition increased the expression of multiple sarcomere proteins and enhanced myofibrillar organization. Furthermore, inhibition of KDM5 increased H3K4me3 deposits at the promoter region of the ESRRA gene and increased its RNA and protein levels. Knockdown of ESRRA in KDM5-C70-treated iPSC-CM suppressed expression of a subset of the KDM5 targets. In conjunction with changes in gene expression, KDM5 inhibition increased oxygen consumption rate and contractility in iPSC-CMs. CONCLUSION: KDM5 inhibition enhances maturation of iPSC-CMs by epigenetically upregulating the expressions of OXPHOS, FAO, and sarcomere genes and enhancing myofibril organization and mitochondrial function.


Subject(s)
Cell Differentiation , Fatty Acids , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Myofibrils , Oxidation-Reduction , Oxidative Phosphorylation , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , Humans , Fatty Acids/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/enzymology , Myofibrils/metabolism , Myofibrils/enzymology , Cells, Cultured , Histones/metabolism , Histones/genetics , Retinoblastoma-Binding Protein 2/metabolism , Retinoblastoma-Binding Protein 2/genetics , Gene Expression Regulation, Developmental , Mitochondria, Heart/enzymology , Mitochondria, Heart/metabolism , Mitochondria, Heart/genetics , Promoter Regions, Genetic
2.
Science ; 379(6628): 179-185, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36634166

ABSTRACT

CRISPR-Cas9 gene editing is emerging as a prospective therapy for genomic mutations. However, current editing approaches are directed primarily toward relatively small cohorts of patients with specific mutations. Here, we describe a cardioprotective strategy potentially applicable to a broad range of patients with heart disease. We used base editing to ablate the oxidative activation sites of CaMKIIδ, a primary driver of cardiac disease. We show in cardiomyocytes derived from human induced pluripotent stem cells that editing the CaMKIIδ gene to eliminate oxidation-sensitive methionine residues confers protection from ischemia/reperfusion (IR) injury. Moreover, CaMKIIδ editing in mice at the time of IR enables the heart to recover function from otherwise severe damage. CaMKIIδ gene editing may thus represent a permanent and advanced strategy for heart disease therapy.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Gene Editing , Heart Diseases , Animals , Humans , Mice , CRISPR-Cas Systems , Heart Diseases/genetics , Heart Diseases/therapy , Induced Pluripotent Stem Cells/enzymology , Myocytes, Cardiac/enzymology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics
3.
Stem Cell Res ; 65: 102971, 2022 12.
Article in English | MEDLINE | ID: mdl-36403546

ABSTRACT

Human dermal fibroblasts from a Leigh Syndrome (LS) patient harboring the heterozygous NDUFS1 R557X/D618N compound mutation were reprogrammed to generate integration-free induced pluripotent stem cells (iPSCs). The full characterization of IUFi002-A-iPSCs demonstrated that the line is free of exogenous reprogramming genes and maintains the genomic integrity. IUFi002-A-iPSCs' pluripotency was confirmed by the expression of pluripotency markers and embryoid body-based differentiation into cell types representative of each of the three germ layers. The generated iPSC line provides a powerful tool to investigate LS and analyze the molecular mechanisms underlying NDUFS1 mutations-induced pathology.


Subject(s)
Induced Pluripotent Stem Cells , Leigh Disease , NADH Dehydrogenase , Humans , Genomics , Induced Pluripotent Stem Cells/enzymology , Induced Pluripotent Stem Cells/pathology , Leigh Disease/genetics , Leigh Disease/pathology , Mutation , NADH Dehydrogenase/genetics , Cell Line
4.
Stem Cell Res ; 65: 102970, 2022 12.
Article in English | MEDLINE | ID: mdl-36399926

ABSTRACT

Cytochrome P450 (CYP) reaction phenotyping has become crucial for predicting drug reactions and side effects. Single nucleotide polymorphisms (SNPs) in CYP genes alter drug metabolism capacity and cause unexpected drug-related reactions. Here, we established two human induced pluripotent stem cell (hiPSC) lines with pharmacologically important SNPs in CYP2D6 in conjunction with CYP2C19 or CYP3A5 genes. These hiPSC lines can serve as valuable resources for expanding our understanding of the relationships between genotypes and drug reactions.


Subject(s)
Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2D6 , Cytochrome P-450 CYP3A , Induced Pluripotent Stem Cells , Pharmaceutical Preparations , Humans , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP3A/genetics , Induced Pluripotent Stem Cells/enzymology , Cell Line , Polymorphism, Single Nucleotide , Pharmaceutical Preparations/metabolism
5.
Sci Rep ; 12(1): 347, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013447

ABSTRACT

Cancer stem cells (CSCs) are capable of continuous proliferation, self-renewal and are proposed to play significant roles in oncogenesis, tumor growth, metastasis and cancer recurrence. We have established a model of CSCs that was originally developed from mouse induced pluripotent stem cells (miPSCs) by proposing miPSCs to the conditioned medium (CM) of cancer derived cells, which is a mimic of carcinoma microenvironment. Further research found that not only PI3K-Akt but also EGFR signaling pathway was activated during converting miPSCs into CSCs. In this study, we tried to observe both of PI3Kγ inhibitor Eganelisib and EGFR inhibitor Gefitinib antitumor effects on the models of CSCs derived from miPSCs (miPS-CSC) in vitro and in vivo. As the results, targeting these two pathways exhibited significant inhibition of cell proliferation, self-renewal, migration and invasion abilities in vitro. Both Eganelisib and Gefitinib showed antitumor effects in vivo while Eganelisib displayed more significant therapeutic efficacy and less side effects than Gefitinib on all miPS-CSC models. Thus, these data suggest that the inhibitiors of PI3K and EGFR, especially PI3Kγ, might be a promising therapeutic strategy against CSCs defeating cancer in the near future.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/metabolism , ErbB Receptors/antagonists & inhibitors , Gefitinib/pharmacology , Induced Pluripotent Stem Cells/drug effects , Neoplastic Stem Cells/drug effects , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , ErbB Receptors/metabolism , Female , Induced Pluripotent Stem Cells/enzymology , Induced Pluripotent Stem Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Signal Transduction , Tumor Burden/drug effects
6.
Cardiovasc Res ; 118(2): 517-530, 2022 01 29.
Article in English | MEDLINE | ID: mdl-33705529

ABSTRACT

AIMS: Hypertrophic cardiomyopathy (HCM) is characterized by cardiomyocyte hypertrophy and disarray, and myocardial stiffness due to interstitial fibrosis, which result in impaired left ventricular filling and diastolic dysfunction. The latter manifests as exercise intolerance, angina, and dyspnoea. There is currently no specific treatment for improving diastolic function in HCM. Here, we investigated whether myeloperoxidase (MPO) is expressed in cardiomyocytes and provides a novel therapeutic target for alleviating diastolic dysfunction in HCM. METHODS AND RESULTS: Human cardiomyocytes derived from control-induced pluripotent stem cells (iPSC-CMs) were shown to express MPO, with MPO levels being increased in iPSC-CMs generated from two HCM patients harbouring sarcomeric mutations in the MYBPC3 and MYH7 genes. The presence of cardiomyocyte MPO was associated with higher chlorination and peroxidation activity, increased levels of 3-chlorotyrosine-modified cardiac myosin binding protein-C (MYBPC3), attenuated phosphorylation of MYBPC3 at Ser-282, perturbed calcium signalling, and impaired cardiomyocyte relaxation. Interestingly, treatment with the MPO inhibitor, AZD5904, reduced 3-chlorotyrosine-modified MYBPC3 levels, restored MYBPC3 phosphorylation, and alleviated the calcium signalling and relaxation defects. Finally, we found that MPO protein was expressed in healthy adult murine and human cardiomyocytes, and MPO levels were increased in diseased hearts with left ventricular hypertrophy. CONCLUSION: This study demonstrates that MPO inhibition alleviates the relaxation defect in hypertrophic iPSC-CMs through MYBPC3 phosphorylation. These findings highlight cardiomyocyte MPO as a novel therapeutic target for improving myocardial relaxation associated with HCM, a treatment strategy which can be readily investigated in the clinical setting, given that MPO inhibitors are already available for clinical testing.


Subject(s)
Cardiomyopathy, Hypertrophic/drug therapy , Enzyme Inhibitors/pharmacology , Hypertrophy, Left Ventricular/drug therapy , Induced Pluripotent Stem Cells/drug effects , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Peroxidase/antagonists & inhibitors , Ventricular Function, Left/drug effects , Animals , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Cardiomyopathy, Hypertrophic/enzymology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/physiopathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Disease Models, Animal , Humans , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/physiopathology , Induced Pluripotent Stem Cells/enzymology , Induced Pluripotent Stem Cells/pathology , Male , Mice, Inbred C57BL , Mutation, Missense , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Peroxidase/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
7.
Cell Rep ; 37(3): 109864, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34686322

ABSTRACT

Increasing evidence suggests that neurodevelopmental alterations might contribute to increase the susceptibility to develop neurodegenerative diseases. We investigate the occurrence of developmental abnormalities in dopaminergic neurons in a model of Parkinson's disease (PD). We monitor the differentiation of human patient-specific neuroepithelial stem cells (NESCs) into dopaminergic neurons. Using high-throughput image analyses and single-cell RNA sequencing, we observe that the PD-associated LRRK2-G2019S mutation alters the initial phase of neuronal differentiation by accelerating cell-cycle exit with a concomitant increase in cell death. We identify the NESC-specific core regulatory circuit and a molecular mechanism underlying the observed phenotypes. The expression of NR2F1, a key transcription factor involved in neurogenesis, decreases in LRRK2-G2019S NESCs, neurons, and midbrain organoids compared to controls. We also observe accelerated dopaminergic differentiation in vivo in NR2F1-deficient mouse embryos. This suggests a pathogenic mechanism involving the LRRK2-G2019S mutation, where the dynamics of dopaminergic differentiation are modified via NR2F1.


Subject(s)
Brain/enzymology , COUP Transcription Factor I/metabolism , Dopaminergic Neurons/enzymology , Induced Pluripotent Stem Cells/enzymology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Neural Stem Cells/enzymology , Neurogenesis , Parkinson Disease/enzymology , Animals , Brain/pathology , COUP Transcription Factor I/genetics , Cell Cycle , Cell Line , Cell Proliferation , Cell Survival , Dopaminergic Neurons/pathology , Female , Humans , Induced Pluripotent Stem Cells/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Male , Mice, 129 Strain , Mice, Knockout , Mutation , Neural Stem Cells/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Phenotype , RNA-Seq , Signal Transduction , Single-Cell Analysis , Time Factors
8.
Elife ; 102021 09 30.
Article in English | MEDLINE | ID: mdl-34590578

ABSTRACT

Astrocytes are essential cells of the central nervous system, characterized by dynamic relationships with neurons that range from functional metabolic interactions and regulation of neuronal firing activities, to the release of neurotrophic and neuroprotective factors. In Parkinson's disease (PD), dopaminergic neurons are progressively lost during the course of the disease, but the effects of PD on astrocytes and astrocyte-to-neuron communication remain largely unknown. This study focuses on the effects of the PD-related mutation LRRK2 G2019S in astrocytes generated from patient-derived induced pluripotent stem cells. We report the alteration of extracellular vesicle (EV) biogenesis in astrocytes and identify the abnormal accumulation of key PD-related proteins within multivesicular bodies (MVBs). We found that dopaminergic neurons internalize astrocyte-secreted EVs and that LRRK2 G2019S EVs are abnormally enriched in neurites and fail to provide full neurotrophic support to dopaminergic neurons. Thus, dysfunctional astrocyte-to-neuron communication via altered EV biological properties may participate in the progression of PD.


Subject(s)
Astrocytes/enzymology , Cell Communication , Dopaminergic Neurons/enzymology , Exosomes/enzymology , Induced Pluripotent Stem Cells/enzymology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Neural Stem Cells/enzymology , Parkinson Disease/enzymology , Animals , Astrocytes/ultrastructure , Atrophy , Case-Control Studies , Cell Line , Dopaminergic Neurons/pathology , Endocytosis , Exosomes/genetics , Exosomes/ultrastructure , Humans , Induced Pluripotent Stem Cells/ultrastructure , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neural Stem Cells/ultrastructure , Organelle Biogenesis , Parkinson Disease/genetics , Parkinson Disease/pathology
9.
Sci Rep ; 11(1): 16580, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34400719

ABSTRACT

Membrane proteins constitute a substantial fraction of the human proteome, thus representing a vast source of therapeutic drug targets. Indeed, newly devised technologies now allow targeting "undruggable" regions of membrane proteins to modulate protein function in the cell. Despite the advances in technology, the rapid translation of basic science discoveries into potential drug candidates targeting transmembrane protein domains remains challenging. We address this issue by harmonizing single molecule-based and ensemble-based atomistic simulations of ligand-membrane interactions with patient-derived induced pluripotent stem cell (iPSC)-based experiments to gain insights into drug delivery, cellular efficacy, and safety of molecules directed at membrane proteins. In this study, we interrogated the pharmacological activation of the cardiac Ca2+ pump (Sarcoplasmic reticulum Ca2+-ATPase, SERCA2a) in human iPSC-derived cardiac cells as a proof-of-concept model. The combined computational-experimental approach serves as a platform to explain the differences in the cell-based activity of candidates with similar functional profiles, thus streamlining the identification of drug-like candidates that directly target SERCA2a activation in human cardiac cells. Systematic cell-based studies further showed that a direct SERCA2a activator does not induce cardiotoxic pro-arrhythmogenic events in human cardiac cells, demonstrating that pharmacological stimulation of SERCA2a activity is a safe therapeutic approach targeting the heart. Overall, this novel multiscale platform encompasses organ-specific drug potency, efficacy, and safety, and opens new avenues to accelerate the bench-to-patient research aimed at designing effective therapies directed at membrane protein domains.


Subject(s)
Membrane Proteins/drug effects , Molecular Targeted Therapy/methods , Sarcoplasmic Reticulum Calcium-Transporting ATPases/drug effects , Small Molecule Libraries/therapeutic use , Animals , Enzyme Activation/drug effects , Giant Cells/enzymology , Humans , Induced Pluripotent Stem Cells/enzymology , Microsomes/enzymology , Molecular Dynamics Simulation , Molecular Structure , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Phosphatidylcholines , Protein Domains/drug effects , Sarcoplasmic Reticulum/enzymology , Small Molecule Libraries/adverse effects , Small Molecule Libraries/pharmacology , Swine , Water
10.
Basic Res Cardiol ; 116(1): 34, 2021 05 20.
Article in English | MEDLINE | ID: mdl-34018053

ABSTRACT

Reducing infarct size (IS) by interfering with mechanisms for cardiomyocyte death remains an elusive goal. DMX-5804, a selective inhibitor of the stress-activated kinase MAP4K4, suppresses cell death in mouse myocardial infarction (MI), human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs), and 3D human engineered heart tissue, whose fidelity to human biology is hoped to strengthen the route to clinical success. Here, DMX-10001, a soluble, rapidly cleaved pro-drug of DMX-5804, was developed for i.v. testing in large-mammal MI. Following pharmacodynamic studies, a randomized, blinded efficacy study was performed in swine subjected to LAD balloon occlusion (60 min) and reperfusion (24 h). Thirty-six animals were enrolled; 12 were excluded by pre-defined criteria, death before infusion, or technical issues. DMX-10001 was begun 20 min before reperfusion (30 min, 60 mg/kg/h; 23.5 h, 17 mg/kg/h). At all times tested, beginning 30 min after the start of infusion, DMX-5804 concentrations exceeded > fivefold the levels that rescued hPSC-CMs and reduced IS in mice after oral dosing with DMX-5804 itself. No significant reduction occurred in IS or no-reflow corrected for the area at ischemic risk, even though DMX-10001 reduced IS, expressed in grams or % of LV mass, by 27%. In summary, a rapidly cleaved pro-drug of DMX-5804 failed to reduce IS in large-mammal MI, despite exceeding the concentrations for proven success in both mice and hPSC-CMs.


Subject(s)
Induced Pluripotent Stem Cells/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Myocardial Infarction/prevention & control , Myocytes, Cardiac/drug effects , Prodrugs/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Disease Models, Animal , Female , Hemodynamics/drug effects , Humans , Induced Pluripotent Stem Cells/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Prodrugs/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Protein Serine-Threonine Kinases/metabolism , Sus scrofa , Translational Research, Biomedical , Ventricular Function, Left/drug effects
11.
STAR Protoc ; 2(1): 100340, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33659904

ABSTRACT

Lysosomes are critical for maintaining protein homeostasis and cellular metabolism. Lysosomal dysfunction and disrupted protein trafficking contribute to cell death in neurodegenerative disorders, including Parkinson's disease and dementia. We describe three complementary protocols-the use of protein glycosylation, western blotting, immunofluorescence, and hydrolase activity measurement-to analyze the trafficking and activity of lysosomal proteins in patient-derived neurons differentiated from iPSCs. These methods should help to identify lysosomal phenotypes in patient-derived cultures and aid the discovery of therapeutics that augment lysosomal function. For complete details on the use and execution of this protocol, please refer to Cuddy et al. (2019).


Subject(s)
Cell Differentiation , Hydrolases/metabolism , Induced Pluripotent Stem Cells/enzymology , Lysosomes/enzymology , Models, Neurological , Parkinson Disease/enzymology , Humans , Induced Pluripotent Stem Cells/pathology , Lysosomes/pathology , Parkinson Disease/pathology , Protein Transport
12.
Cardiovasc Res ; 117(2): 472-483, 2021 01 21.
Article in English | MEDLINE | ID: mdl-32061134

ABSTRACT

AIMS: NOS1AP single-nucleotide polymorphisms (SNPs) correlate with QT prolongation and cardiac sudden death in patients affected by long QT syndrome type 1 (LQT1). NOS1AP targets NOS1 to intracellular effectors. We hypothesize that NOS1AP SNPs cause NOS1 dysfunction and this may converge with prolonged action-potential duration (APD) to facilitate arrhythmias. Here we test (i) the effects of NOS1 inhibition and their interaction with prolonged APD in a guinea pig cardiomyocyte (GP-CMs) LQT1 model; (ii) whether pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from LQT1 patients differing for NOS1AP variants and mutation penetrance display a phenotype compatible with NOS1 deficiency. METHODS AND RESULTS: In GP-CMs, NOS1 was inhibited by S-Methyl-L-thiocitrulline acetate (SMTC) or Vinyl-L-NIO hydrochloride (L-VNIO); LQT1 was mimicked by IKs blockade (JNJ303) and ß-adrenergic stimulation (isoproterenol). hiPSC-CMs were obtained from symptomatic (S) and asymptomatic (AS) KCNQ1-A341V carriers, harbouring the minor and major alleles of NOS1AP SNPs (rs16847548 and rs4657139), respectively. In GP-CMs, NOS1 inhibition prolonged APD, enhanced ICaL and INaL, slowed Ca2+ decay, and induced delayed afterdepolarizations. Under action-potential clamp, switching to shorter APD suppressed 'transient inward current' events induced by NOS1 inhibition and reduced cytosolic Ca2+. In S (vs. AS) hiPSC-CMs, APD was longer and ICaL larger; NOS1AP and NOS1 expression and co-localization were decreased. CONCLUSION: The minor NOS1AP alleles are associated with NOS1 loss of function. The latter likely contributes to APD prolongation in LQT1 and converges with it to perturb Ca2+ handling. This establishes a mechanistic link between NOS1AP SNPs and aggravation of the arrhythmia phenotype in prolonged repolarization syndromes.


Subject(s)
Action Potentials , Adaptor Proteins, Signal Transducing/genetics , Heart Rate , Induced Pluripotent Stem Cells/enzymology , KCNQ1 Potassium Channel/genetics , Mutation , Myocytes, Cardiac/enzymology , Nitric Oxide Synthase Type I/genetics , Polymorphism, Single Nucleotide , Romano-Ward Syndrome/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium Signaling , Cell Line , Genetic Predisposition to Disease , Guinea Pigs , Humans , KCNQ1 Potassium Channel/metabolism , Nitric Oxide Synthase Type I/metabolism , Phenotype , Romano-Ward Syndrome/diagnosis , Romano-Ward Syndrome/enzymology , Romano-Ward Syndrome/physiopathology , Time Factors
13.
Arch Toxicol ; 95(3): 907-922, 2021 03.
Article in English | MEDLINE | ID: mdl-33263786

ABSTRACT

Human intestinal organoids (HIOs) are a promising in vitro model consisting of different intestinal cell types with a 3D microarchitecture resembling native tissue. In the current study, we aimed to assess the expression of the most common intestinal CYP enzymes in a human induced pluripotent stem cell (hiPSC)-derived HIO model, and the suitability of that model to study chemical-induced changes in CYP expression and activity. We compared this model with the commonly used human colonic adenocarcinoma cell line Caco-2 and with a human primary intestinal epithelial cell (IEC)-based model, closely resembling in vivo tissue. We optimized an existing protocol to differentiate hiPSCs into HIOs and demonstrated that obtained HIOs contain a polarized epithelium with tight junctions consisting of enterocytes, goblet cells, enteroendocrine cells and Paneth cells. We extensively characterized the gene expression of CYPs and activity of CYP3A4/5, indicating relatively high gene expression levels of the most important intestinal CYP enzymes in HIOs compared to the other models. Furthermore, we showed that CYP1A1 and CYP1B1 were induced by ß-naphtoflavone in all three models, whereas CYP3A4 was induced by phenobarbital and rifampicin in HIOs, in the IEC-based model (although not statistically significant), but not in Caco-2 cells. Interestingly, CYP2B6 expression was not induced in any of the models by the well-known liver CYP2B6 inducer phenobarbital. In conclusion, our study indicates that hiPSC-based HIOs are a useful in vitro intestinal model to study biotransformation of chemicals in the intestine.


Subject(s)
Cytochrome P-450 Enzyme Inducers/pharmacology , Cytochrome P-450 Enzyme System/genetics , Induced Pluripotent Stem Cells/metabolism , Organoids/metabolism , Adult , Caco-2 Cells , Cell Line , Cells, Cultured , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Enzymologic , Humans , Induced Pluripotent Stem Cells/enzymology , Intestinal Mucosa/cytology , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism
14.
Sci Rep ; 10(1): 14302, 2020 08 31.
Article in English | MEDLINE | ID: mdl-32868812

ABSTRACT

Recent findings suggest a pathologic role of skeletal muscle in amyotrophic lateral sclerosis (ALS) onset and progression. However, the exact mechanism by which this occurs remains elusive due to limited human-based studies. To this end, phenotypic ALS skeletal muscle models were developed from induced pluripotent stem cells (iPSCs) derived from healthy individuals (WT) and ALS patients harboring mutations in the superoxide dismutase 1 (SOD1) gene. Although proliferative, SOD1 myoblasts demonstrated delayed and reduced fusion efficiency compared to WT. Additionally, SOD1 myotubes exhibited significantly reduced length and cross-section. Also, SOD1 myotubes had loosely arranged myosin heavy chain and reduced acetylcholine receptor expression per immunocytochemical analysis. Functional analysis indicated considerably reduced contractile force and synchrony in SOD1 myotubes. Mitochondrial assessment indicated reduced inner mitochondrial membrane potential (ΔΨm) and metabolic plasticity in the SOD1-iPSC derived myotubes. This work presents the first well-characterized in vitro iPSC-derived muscle model that demonstrates SOD1 toxicity effects on human muscle regeneration, contractility and metabolic function in ALS. Current findings align with previous ALS patient biopsy studies and suggest an active contribution of skeletal muscle in NMJ dysfunction. Further, the results validate this model as a human-relevant platform for ALS research and drug discovery studies.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Induced Pluripotent Stem Cells/metabolism , Muscle, Skeletal/pathology , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/etiology , Amyotrophic Lateral Sclerosis/genetics , Cell Lineage/genetics , Disease Progression , Humans , Induced Pluripotent Stem Cells/enzymology , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Mutation/genetics , Myoblasts/enzymology , Myoblasts/pathology
15.
Stem Cell Reports ; 15(6): 1233-1245, 2020 12 08.
Article in English | MEDLINE | ID: mdl-32976761

ABSTRACT

Methylation of histone 3 at lysine 9 (H3K9) constitutes a roadblock for cellular reprogramming. Interference with methyltransferases or activation of demethylases by the cofactor ascorbic acid (AA) facilitates the derivation of induced pluripotent stem cells (iPSCs), but possible interactions between specific methyltransferases and AA treatment remain insufficiently explored. We show that chemical inhibition of the methyltransferases EHMT1 and EHMT2 counteracts iPSC formation in an enhanced reprogramming system in the presence of AA, an effect that is dependent on EHMT1. EHMT inhibition during enhanced reprogramming is associated with rapid loss of H3K9 dimethylation, inefficient downregulation of somatic genes, and failed mesenchymal-to-epithelial transition. Furthermore, transient EHMT inhibition during reprogramming yields iPSCs that fail to efficiently give rise to viable mice upon blastocyst injection. Our observations establish novel functions of H3K9 methyltransferases and suggest that a functional balance between AA-stimulated enzymes and EHMTs supports efficient and less error-prone iPSC reprogramming to pluripotency.


Subject(s)
Cellular Reprogramming , Histone-Lysine N-Methyltransferase/metabolism , Induced Pluripotent Stem Cells/enzymology , Animals , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Induced Pluripotent Stem Cells/cytology , Methylation , Mice
16.
J Physiol Sci ; 70(1): 39, 2020 Sep 07.
Article in English | MEDLINE | ID: mdl-32895058

ABSTRACT

Signal regulators during early cardiogenetic differentiation for the cellular automaticity are largely unknown. Our investigations were designed to clarify the role of transcription factors and their modulators in P19-derived cardiomyocytes to the expression of cardiac pacemaker ion channels. Transcription factors Csx/Nkx2.5 and GATA4 but not MEF2C were markedly inhibited by p38 MAP kinase inhibition in a distinct manner; expression but not phosphorylation of GATA4 was reduced by inhibition of p38 MAP kinase actions. In the presence of an ERK1/2,5 inhibitor PD98059 or a JNK MAP kinase inhibitor SP600125, P19 cells successfully differentiated into cardiomyocytes displaying spontaneous beatings with expression of three types of pacemaker ion channels. We demonstrate that acquisition of cellular automaticity and the expression of pacemaker ion channels are regulated by the transcription factors, Csx/Nkx2.5 and GATA4, through intracellular signals including p38 MAP kinase in the process of P19-derived pluripotent cells differentiation into cardiomyocytes.


Subject(s)
Biological Clocks , Cell Differentiation , Heart Rate , Induced Pluripotent Stem Cells/enzymology , Ion Channels/metabolism , Myocytes, Cardiac/enzymology , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Action Potentials , Animals , Cell Line, Tumor , GATA4 Transcription Factor/metabolism , Homeobox Protein Nkx-2.5/metabolism , MEF2 Transcription Factors/metabolism , Mice , Phenotype , Phosphorylation , Signal Transduction , Time Factors , Transcription Factors/genetics
17.
FASEB J ; 34(7): 9141-9155, 2020 07.
Article in English | MEDLINE | ID: mdl-32421247

ABSTRACT

Human-induced pluripotent stem cells (hiPSCs) are invaluable sources for drug screening and toxicity tests because of their differentiation potential and proliferative capacity. Recently, the CRISPR-Cas9-mediated homologous recombination system has enabled reporter knock-ins at desired loci in hiPSCs, and here, we generated a hiPSC reporter line expressing mCherry-tagged cytochrome P450 1A1 (CYP1A1), which can be utilized to screen for the modulators of aryl hydrocarbon receptor (AHR) in live cells. CYP1A1-mCherry hiPSCs exhibited typical characteristics of pluripotent stem cells such as marker expression, differentiation potential, and normal karyotype. After differentiation into hepatocyte-like cells (HLCs), CYP1A1-mCherry fusion protein was expressed and localized at the endoplasmic reticulum, and induced by AHR agonists. We obtained 23 hits modulating CYP1A1 expression from high-content screening with 241 hepatotoxicity chemicals and nuclear receptor ligands, and identified three upregulating chemicals and two downregulating compounds. Responses of hiPSC-HLCs against an AHR agonist were more similar to human primary hepatocytes than of HepG2 hepatocellular carcinoma cells. This platform has the advantages of live-cell screening without sacrificing cells (unlike previously available CYP1A1 reporter cell lines), as well as an indefinite supply of cells, and can be utilized in a wide range of screening related to AHR- and CYP1A1-associated diseases in desired cell types.


Subject(s)
Cytochrome P-450 CYP1A1/chemistry , Fluorescence , Hepatocytes/cytology , Induced Pluripotent Stem Cells/cytology , Pharmaceutical Preparations/metabolism , Small Molecule Libraries/pharmacology , Cell Differentiation , Cytochrome P-450 CYP1A1/metabolism , Hep G2 Cells , Hepatocytes/drug effects , High-Throughput Screening Assays , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/enzymology , Signal Transduction
18.
Circ Res ; 126(10): e80-e96, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32134364

ABSTRACT

RATIONALE: Diabetes mellitus is a complex, multisystem disease, affecting large populations worldwide. Chronic CaMKII (Ca2+/calmodulin-dependent kinase II) activation may occur in diabetes mellitus and be arrhythmogenic. Diabetic hyperglycemia was shown to activate CaMKII by (1) O-linked attachment of N-acetylglucosamine (O-GlcNAc) at S280 leading to arrhythmia and (2) a reactive oxygen species (ROS)-mediated oxidation of CaMKII that can increase postinfarction mortality. OBJECTIVE: To test whether high extracellular glucose (Hi-Glu) promotes ventricular myocyte ROS generation and the role played by CaMKII. METHODS AND RESULTS: We tested how extracellular Hi-Glu influences ROS production in adult ventricular myocytes, using DCF (2',7'-dichlorodihydrofluorescein diacetate) and genetically targeted Grx-roGFP2 redox sensors. Hi-Glu (30 mmol/L) significantly increased the rate of ROS generation-an effect prevented in myocytes pretreated with CaMKII inhibitor KN-93 or from either global or cardiac-specific CaMKIIδ KO (knockout) mice. CaMKII KO or inhibition also prevented Hi-Glu-induced sarcoplasmic reticulum Ca2+ release events (Ca2+ sparks). Thus, CaMKII activation is required for Hi-Glu-induced ROS generation and sarcoplasmic reticulum Ca2+ leak in cardiomyocytes. To test the involvement of O-GlcNAc-CaMKII pathway, we inhibited GlcNAcylation removal by Thiamet G (ThmG), which mimicked the Hi-Glu-induced ROS production. Conversely, inhibition of GlcNAcylation (OSMI-1 [(αR)-α-[[(1,2-dihydro-2-oxo-6-quinolinyl)sulfonyl]amino]-N-(2-furanylmethyl)-2-methoxy-N-(2-thienylmethyl)-benzeneacetamide]) prevented ROS induction in response to either Hi-Glu or ThmG. Moreover, in a CRSPR-based knock-in mouse in which the functional GlcNAcylation site on CaMKIIδ was ablated (S280A), neither Hi-Glu nor ThmG induced myocyte ROS generation. So CaMKIIδ-S280 is required for the Hi-Glu-induced (and GlcNAc dependent) ROS production. To identify the ROS source(s), we used different inhibitors of NOX (NADPH oxidase) 2 (Gp91ds-tat peptide), NOX4 (GKT137831), mitochondrial ROS (MitoTempo), and NOS (NO synthase) pathway inhibitors (L-NAME, L-NIO, and L-NPA). Only NOX2 inhibition or KO prevented Hi-Glu/ThmG-induced ROS generation. CONCLUSIONS: Diabetic hyperglycemia induces acute cardiac myocyte ROS production by NOX2 that requires O-GlcNAcylation of CaMKIIδ at S280. This novel ROS induction may exacerbate pathological consequences of diabetic hyperglycemia.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Diabetic Cardiomyopathies/etiology , Glucose/toxicity , Hyperglycemia/complications , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Animals , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/deficiency , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cells, Cultured , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/physiopathology , Enzyme Activation , Glutaredoxins/genetics , Glutaredoxins/metabolism , Glycosylation , Humans , Hyperglycemia/enzymology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/enzymology , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/enzymology , NADPH Oxidase 2/deficiency , NADPH Oxidase 2/genetics , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/enzymology
19.
Am J Physiol Cell Physiol ; 318(1): C94-C102, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31618079

ABSTRACT

Matrix metalloproteinases (MMP) are important for cardiac remodeling. Recently, microRNA (miR)-451a has been found to inhibit the expression of both MMP-2 and MMP-9 in human malignancies, but its role in cardiomyocytes has not been explored. We hypothesized that miR-451a modulates MMP-2 and MMP-9 levels in human cardiomyocytes. The role of miR-451a on regulation of MMP-2 and MMP-9 was evaluated in two separate pathological models using Cor.4U human inducible pluripotent stem cell-derived cardiomyocytes (hiPS-CMs): 1) endothelin-1 (ET-1), and 2) 48-h hypoxia (1% O2). Both models were transfected with synthetic miR-451a mimics or scramble control. Expression of both mRNA and miR was determined by quantitative real-time polymerase chain reaction and protein activity by (MMP-2/9) activity assay. Bioinformatic analyses were performed using Targetscan 7.1 and STRING 10.5. hiPS-CMs stimulated by hypoxia increased both MMP-2 and MMP-9 expression levels compared with normoxia (P < 0.05), whereas ET-1 stimulation only increased the MMP-9 level compared with vehicle controls (P < 0.05). miR-451a mimics prevented the increase of MMP-2 and MMP-9 expression in both models. Protein activity of MMP-2 and MMP-9 was confirmed to be lower following treatment with miR-451a mimic compared with scramble-controls. Six of 28 predicted gene transcripts of miR-451a were linked to MMP-2 and MMP-9; Macrophage migration inhibitory factor (MIF) was the only predicted target of miR-451a that was increased by ET-1 and hypoxia and reduced following miR-451a mimic transfection. miR-451a prevent the increase of MMP-2 and MMP-9 in human cardiomyocytes during pathological stress. The modulation by miR-451a on MMP-2 and MMP-9 is caused by MIF.


Subject(s)
Cardiomegaly/enzymology , Induced Pluripotent Stem Cells/enzymology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , MicroRNAs/metabolism , Myocytes, Cardiac/enzymology , Cardiomegaly/genetics , Cardiomegaly/pathology , Cell Differentiation , Cell Hypoxia , Cell Line , Endothelin-1/toxicity , Enzyme Activation , Gene Expression Regulation, Enzymologic , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , MicroRNAs/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Signal Transduction
20.
Cell Stem Cell ; 25(5): 622-638.e13, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31588046

ABSTRACT

Post-transcriptional mechanisms have the potential to influence complex changes in gene expression, yet their role in cell fate transitions remains largely unexplored. Here, we show that suppression of the RNA helicase DDX6 endows human and mouse primed embryonic stem cells (ESCs) with a differentiation-resistant, "hyper-pluripotent" state, which readily reprograms to a naive state resembling the preimplantation embryo. We further demonstrate that DDX6 plays a key role in adult progenitors where it controls the balance between self-renewal and differentiation in a context-dependent manner. Mechanistically, DDX6 mediates the translational suppression of target mRNAs in P-bodies. Upon loss of DDX6 activity, P-bodies dissolve and release mRNAs encoding fate-instructive transcription and chromatin factors that re-enter the ribosome pool. Increased translation of these targets impacts cell fate by rewiring the enhancer, heterochromatin, and DNA methylation landscapes of undifferentiated cell types. Collectively, our data establish a link between P-body homeostasis, chromatin organization, and stem cell potency.


Subject(s)
Cell Differentiation/genetics , Cell Plasticity/genetics , DEAD-box RNA Helicases/metabolism , Induced Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , Ribonucleoproteins/metabolism , Animals , Cell Line , Chromatin Assembly and Disassembly/genetics , DEAD-box RNA Helicases/genetics , DNA Methylation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation/genetics , Gene Ontology , Homeostasis/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/enzymology , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mice, Inbred C57BL , Nanog Homeobox Protein/metabolism , Organoids/cytology , Organoids/diagnostic imaging , Organoids/metabolism , Protein Biosynthesis/genetics , Proteins/metabolism , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , RNA-Seq , Ribonucleoproteins/genetics , Ribosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...