Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Commun Med (Lond) ; 4(1): 38, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38499690

ABSTRACT

BACKGROUND: Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a familial cardiac disease associated with ventricular arrhythmias and an increased risk of sudden cardiac death. Currently, there are no approved treatments that address the underlying genetic cause of this disease, representing a significant unmet need. Mutations in Plakophilin-2 (PKP2), encoding a desmosomal protein, account for approximately 40% of ARVC cases and result in reduced gene expression. METHODS: Our goal is to examine the feasibility and the efficacy of adeno-associated virus 9 (AAV9)-mediated restoration of PKP2 expression in a cardiac specific knock-out mouse model of Pkp2. RESULTS: We show that a single dose of AAV9:PKP2 gene delivery prevents disease development before the onset of cardiomyopathy and attenuates disease progression after overt cardiomyopathy. Restoration of PKP2 expression leads to a significant extension of lifespan by restoring cellular structures of desmosomes and gap junctions, preventing or halting decline in left ventricular ejection fraction, preventing or reversing dilation of the right ventricle, ameliorating ventricular arrhythmia event frequency and severity, and preventing adverse fibrotic remodeling. RNA sequencing analyses show that restoration of PKP2 expression leads to highly coordinated and durable correction of PKP2-associated transcriptional networks beyond desmosomes, revealing a broad spectrum of biological perturbances behind ARVC disease etiology. CONCLUSIONS: We identify fundamental mechanisms of PKP2-associated ARVC beyond disruption of desmosome function. The observed PKP2 dose-function relationship indicates that cardiac-selective AAV9:PKP2 gene therapy may be a promising therapeutic approach to treat ARVC patients with PKP2 mutations.


Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a heart disease that leads to abnormal heartbeats and a higher risk of sudden cardiac death. ARVC is often caused by changes in a gene called PKP2, that then makes less PKP2 protein. PKP2 protein is important for the normal structure and function of the heart. Human ARVC characteristics can be mimicked in a mouse model missing this gene. Given no therapeutic option, our goal was to test if adding a working copy of PKP2 gene in the heart of this mouse model, using a technique called gene therapy that can deliver genes to cells, could improve heart function. Here, we show that a single dose of PKP2 gene therapy can improve heart function and heartbeats as well as extend lifespan in mice. PKP2 gene therapy may be a promising approach to treat ARVC patients with PKP2 mutations.

2.
Stem Cell Reports ; 18(11): 2138-2153, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37863045

ABSTRACT

Congenital heart disease often arises from perturbations of transcription factors (TFs) that guide cardiac development. ISLET1 (ISL1) is a TF that influences early cardiac cell fate, as well as differentiation of other cell types including motor neuron progenitors (MNPs) and pancreatic islet cells. While lineage specificity of ISL1 function is likely achieved through combinatorial interactions, its essential cardiac interacting partners are unknown. By assaying ISL1 genomic occupancy in human induced pluripotent stem cell-derived cardiac progenitors (CPs) or MNPs and leveraging the deep learning approach BPNet, we identified motifs of other TFs that predicted ISL1 occupancy in each lineage, with NKX2.5 and GATA motifs being most closely associated to ISL1 in CPs. Experimentally, nearly two-thirds of ISL1-bound loci were co-occupied by NKX2.5 and/or GATA4. Removal of NKX2.5 from CPs led to widespread ISL1 redistribution, and overexpression of NKX2.5 in MNPs led to ISL1 occupancy of CP-specific loci. These results reveal how ISL1 guides lineage choices through a combinatorial code that dictates genomic occupancy and transcription.


Subject(s)
Induced Pluripotent Stem Cells , Transcription Factors , Humans , Transcription Factors/metabolism , Myocytes, Cardiac , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation/genetics , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism
3.
Circulation ; 148(14): 1099-1112, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37602409

ABSTRACT

BACKGROUND: Cardiac reprogramming is a technique to directly convert nonmyocytes into myocardial cells using genes or small molecules. This intervention provides functional benefit to the rodent heart when delivered at the time of myocardial infarction or activated transgenically up to 4 weeks after myocardial infarction. Yet, several hurdles have prevented the advancement of cardiac reprogramming for clinical use. METHODS: Through a combination of screening and rational design, we identified a cardiac reprogramming cocktail that can be encoded in a single adeno-associated virus. We also created a novel adeno-associated virus capsid that can transduce cardiac fibroblasts more efficiently than available parental serotypes by mutating posttranslationally modified capsid residues. Because a constitutive promoter was needed to drive high expression of these cell fate-altering reprogramming factors, we included binding sites to a cardiomyocyte-restricted microRNA within the 3' untranslated region of the expression cassette that limits expression to nonmyocytes. After optimizing this expression cassette to reprogram human cardiac fibroblasts into induced cardiomyocyte-like cells in vitro, we also tested the ability of this capsid/cassette combination to confer functional benefit in acute mouse myocardial infarction and chronic rat myocardial infarction models. RESULTS: We demonstrated sustained, dose-dependent improvement in cardiac function when treating a rat model 2 weeks after myocardial infarction, showing that cardiac reprogramming, when delivered in a single, clinically relevant adeno-associated virus vector, can support functional improvement in the postremodeled heart. This benefit was not observed with GFP (green fluorescent protein) or a hepatocyte reprogramming cocktail and was achieved even in the presence of immunosuppression, supporting myocyte formation as the underlying mechanism. CONCLUSIONS: Collectively, these results advance the application of cardiac reprogramming gene therapy as a viable therapeutic approach to treat chronic heart failure resulting from ischemic injury.


Subject(s)
MicroRNAs , Myocardial Infarction , Rats , Mice , Humans , Animals , Dependovirus/genetics , Myocytes, Cardiac/metabolism , Myocardial Infarction/therapy , Myocardial Infarction/drug therapy , MicroRNAs/genetics , MicroRNAs/metabolism , Genetic Therapy/methods , Green Fluorescent Proteins/genetics , Cellular Reprogramming , Fibroblasts/metabolism
4.
Circulation ; 145(17): 1339-1355, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35061545

ABSTRACT

BACKGROUND: The regenerative capacity of the heart after myocardial infarction is limited. Our previous study showed that ectopic introduction of 4 cell cycle factors (4F; CDK1 [cyclin-dependent kinase 1], CDK4 [cyclin-dependent kinase 4], CCNB [cyclin B1], and CCND [cyclin D1]) promotes cardiomyocyte proliferation in 15% to 20% of infected cardiomyocytes in vitro and in vivo and improves cardiac function after myocardial infarction in mice. METHODS: Using temporal single-cell RNA sequencing, we aimed to identify the necessary reprogramming stages during the forced cardiomyocyte proliferation with 4F on a single cell basis. Using rat and pig models of ischemic heart failure, we aimed to start the first preclinical testing to introduce 4F gene therapy as a candidate for the treatment of ischemia-induced heart failure. RESULTS: Temporal bulk and single-cell RNA sequencing and further biochemical validations of mature human induced pluripotent stem cell-derived cardiomyocytes treated with either LacZ or 4F adenoviruses revealed full cell cycle reprogramming in 15% of the cardiomyocyte population at 48 hours after infection with 4F, which was associated mainly with sarcomere disassembly and metabolic reprogramming (n=3/time point/group). Transient overexpression of 4F, specifically in cardiomyocytes, was achieved using a polycistronic nonintegrating lentivirus (NIL) encoding 4F; each is driven by a TNNT2 (cardiac troponin T isoform 2) promoter (TNNT2-4Fpolycistronic-NIL). TNNT2-4Fpolycistronic-NIL or control virus was injected intramyocardially 1 week after myocardial infarction in rats (n=10/group) or pigs (n=6-7/group). Four weeks after injection, TNNT2-4Fpolycistronic-NIL-treated animals showed significant improvement in left ventricular ejection fraction and scar size compared with the control virus-treated animals. At 4 months after treatment, rats that received TNNT2-4Fpolycistronic-NIL still showed a sustained improvement in cardiac function and no obvious development of cardiac arrhythmias or systemic tumorigenesis (n=10/group). CONCLUSIONS: This study provides mechanistic insights into the process of forced cardiomyocyte proliferation and advances the clinical feasibility of this approach by minimizing the oncogenic potential of the cell cycle factors owing to the use of a novel transient and cardiomyocyte-specific viral construct.


Subject(s)
Heart Failure , Induced Pluripotent Stem Cells , Myocardial Infarction , Animals , Cell Cycle , Heart Failure/complications , Heart Failure/genetics , Heart Failure/therapy , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Myocardial Infarction/complications , Myocardial Infarction/genetics , Myocardial Infarction/therapy , Myocytes, Cardiac/metabolism , Rats , Stroke Volume , Swine , Ventricular Function, Left
5.
Circ Res ; 125(6): 628-642, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31310161

ABSTRACT

RATIONALE: Preclinical testing of cardiotoxicity and efficacy of novel heart failure therapies faces a major limitation: the lack of an in situ culture system that emulates the complexity of human heart tissue and maintains viability and functionality for a prolonged time. OBJECTIVE: To develop a reliable, easily reproducible, medium-throughput method to culture pig and human heart slices under physiological conditions for a prolonged period of time. METHODS AND RESULTS: Here, we describe a novel, medium-throughput biomimetic culture system that maintains viability and functionality of human and pig heart slices (300 µm thickness) for 6 days in culture. We optimized the medium and culture conditions with continuous electrical stimulation at 1.2 Hz and oxygenation of the medium. Functional viability of these slices over 6 days was confirmed by assessing their calcium homeostasis, twitch force generation, and response to ß-adrenergic stimulation. Temporal transcriptome analysis using RNAseq at day 2, 6, and 10 in culture confirmed overall maintenance of normal gene expression for up to 6 days, while over 500 transcripts were differentially regulated after 10 days. Electron microscopy demonstrated intact mitochondria and Z-disc ultra-structures after 6 days in culture under our optimized conditions. This biomimetic culture system was successful in keeping human heart slices completely viable and functionally and structurally intact for 6 days in culture. We also used this system to demonstrate the effects of a novel gene therapy approach in human heart slices. Furthermore, this culture system enabled the assessment of contraction and relaxation kinetics on isolated single myofibrils from heart slices after culture. CONCLUSIONS: We have developed and optimized a reliable medium-throughput culture system for pig and human heart slices as a platform for testing the efficacy of novel heart failure therapeutics and reliable testing of cardiotoxicity in a 3-dimensional heart model.


Subject(s)
Biomimetics/methods , Heart Ventricles/ultrastructure , Ventricular Function/physiology , Adult , Animals , Female , Heart/physiology , Heart Ventricles/cytology , Humans , Male , Metabolomics/methods , Middle Aged , Myocardium/cytology , Myocardium/ultrastructure , Organ Culture Techniques/methods , Swine , Transcriptome/physiology
6.
Cell Stem Cell ; 25(1): 87-102.e9, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31271750

ABSTRACT

Ectopic expression of combinations of transcription factors (TFs) can drive direct lineage conversion, thereby reprogramming a somatic cell's identity. To determine the molecular mechanisms by which Gata4, Mef2c, and Tbx5 (GMT) induce conversion from a cardiac fibroblast toward an induced cardiomyocyte, we performed comprehensive transcriptomic, DNA-occupancy, and epigenomic interrogation throughout the reprogramming process. Integration of these datasets identified new TFs involved in cardiac reprogramming and revealed context-specific roles for GMT, including the ability of Mef2c and Tbx5 to independently promote chromatin remodeling at previously inaccessible sites. We also find evidence for cooperative facilitation and refinement of each TF's binding profile in a combinatorial setting. A reporter assay employing newly defined regulatory elements confirmed that binding of a single TF can be sufficient for gene activation, suggesting that co-binding events do not necessarily reflect synergy. These results shed light on fundamental mechanisms by which combinations of TFs direct lineage conversion.


Subject(s)
GATA4 Transcription Factor/metabolism , MEF2 Transcription Factors/metabolism , Myocytes, Cardiac/physiology , T-Box Domain Proteins/metabolism , Animals , Cell Differentiation , Cell Lineage , Cells, Cultured , Cellular Reprogramming , Chromatin Assembly and Disassembly , Epigenesis, Genetic , GATA4 Transcription Factor/genetics , MEF2 Transcription Factors/genetics , Machine Learning , Mice , Protein Binding , T-Box Domain Proteins/genetics , Transcriptional Activation
7.
Science ; 364(6443): 865-870, 2019 05 31.
Article in English | MEDLINE | ID: mdl-31147515

ABSTRACT

Complex genetic mechanisms are thought to underlie many human diseases, yet experimental proof of this model has been elusive. Here, we show that a human cardiac anomaly can be caused by a combination of rare, inherited heterozygous mutations. Whole-exome sequencing of a nuclear family revealed that three offspring with childhood-onset cardiomyopathy had inherited three missense single-nucleotide variants in the MKL2, MYH7, and NKX2-5 genes. The MYH7 and MKL2 variants were inherited from the affected, asymptomatic father and the rare NKX2-5 variant (minor allele frequency, 0.0012) from the unaffected mother. We used CRISPR-Cas9 to generate mice encoding the orthologous variants and found that compound heterozygosity for all three variants recapitulated the human disease phenotype. Analysis of murine hearts and human induced pluripotent stem cell-derived cardiomyocytes provided histologic and molecular evidence for the NKX2-5 variant's contribution as a genetic modifier.


Subject(s)
Cardiomyopathies/genetics , Heterozygote , Homeobox Protein Nkx-2.5/genetics , Multifactorial Inheritance , Thyroid Nuclear Factor 1/genetics , Animals , CRISPR-Associated Protein 9 , Cardiac Myosins/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Exome , Gene Frequency , Humans , Induced Pluripotent Stem Cells , Mice , Mice, Mutant Strains , Mutation, Missense , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Paternal Inheritance/genetics , Transcription Factors/genetics
8.
Development ; 144(5): 866-875, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28246214

ABSTRACT

miR-1 is a small noncoding RNA molecule that modulates gene expression in heart and skeletal muscle. Loss of Drosophila miR-1 produces defects in somatic muscle and embryonic heart development, which have been partly attributed to miR-1 directly targeting Delta to decrease Notch signaling. Here, we show that overexpression of miR-1 in the fly wing can paradoxically increase Notch activity independently of its effects on Delta. Analyses of potential miR-1 targets revealed that miR-1 directly regulates the 3'UTR of the E3 ubiquitin ligase Nedd4 Analysis of embryonic and adult fly heart revealed that the Nedd4 protein regulates heart development in Drosophila Larval fly hearts overexpressing miR-1 have profound defects in actin filament organization that are partially rescued by concurrent overexpression of Nedd4. These results indicate that miR-1 and Nedd4 act together in the formation and actin-dependent patterning of the fly heart. Importantly, we have found that the biochemical and genetic relationship between miR-1 and the mammalian ortholog Nedd4-like (Nedd4l) is evolutionarily conserved in the mammalian heart, potentially indicating a role for Nedd4L in mammalian postnatal maturation. Thus, miR-1-mediated regulation of Nedd4/Nedd4L expression may serve to broadly modulate the trafficking or degradation of Nedd4/Nedd4L substrates in the heart.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , MicroRNAs/metabolism , Ubiquitin-Protein Ligases/metabolism , 3' Untranslated Regions , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Body Patterning , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Green Fluorescent Proteins/metabolism , Heart/physiology , Nedd4 Ubiquitin Protein Ligases , Phenotype , Phosphorylation , Protein Transport , Receptors, Notch/metabolism , Signal Transduction , Ubiquitination , Wings, Animal/metabolism
9.
Cell Stem Cell ; 20(2): 151-152, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28157494

ABSTRACT

The identity of the cells and molecular events driving deleterious calcification of heart muscle remains elusive. In this issue of Cell Stem Cell, Pillai et al. (2017) report that cardiac fibroblasts respond to injury by adopting an osteogenic cell fate and creating damaging calcific deposits, which can be prevented by inhibiting the activated mineralization process.


Subject(s)
Fibroblasts , Osteogenesis , Bone and Bones , Calcinosis , Cell Differentiation , Humans
10.
Circulation ; 135(10): 978-995, 2017 Mar 07.
Article in English | MEDLINE | ID: mdl-27834668

ABSTRACT

BACKGROUND: Reprogramming of cardiac fibroblasts into induced cardiomyocyte-like cells in situ represents a promising strategy for cardiac regeneration. A combination of 3 cardiac transcription factors, Gata4, Mef2c, and Tbx5 (GMT), can convert fibroblasts into induced cardiomyocyte-like cells, albeit with low efficiency in vitro. METHODS: We screened 5500 compounds in primary cardiac fibroblasts to identify the pathways that can be modulated to enhance cardiomyocyte reprogramming. RESULTS: We found that a combination of the transforming growth factor-ß inhibitor SB431542 and the WNT inhibitor XAV939 increased reprogramming efficiency 8-fold when added to GMT-overexpressing cardiac fibroblasts. The small molecules also enhanced the speed and quality of cell conversion; we observed beating cells as early as 1 week after reprogramming compared with 6 to 8 weeks with GMT alone. In vivo, mice exposed to GMT, SB431542, and XAV939 for 2 weeks after myocardial infarction showed significantly improved reprogramming and cardiac function compared with those exposed to only GMT. Human cardiac reprogramming was similarly enhanced on transforming growth factor-ß and WNT inhibition and was achieved most efficiently with GMT plus myocardin. CONCLUSIONS: Transforming growth factor-ß and WNT inhibitors jointly enhance GMT-induced direct cardiac reprogramming from cardiac fibroblasts in vitro and in vivo and provide a more robust platform for cardiac regeneration.


Subject(s)
Benzamides/pharmacology , Cellular Reprogramming/drug effects , Dioxoles/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Transcription Factors/metabolism , Animals , Benzamides/therapeutic use , Cells, Cultured , Dioxoles/therapeutic use , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Heart/diagnostic imaging , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Magnetic Resonance Imaging , Mice , Myocardial Infarction/drug therapy , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism
11.
Curr Protoc Hum Genet ; 87: 21.2.1-21.2.21, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26439714

ABSTRACT

The advent of induced pluripotent stem (iPS) cell technology has revolutionized biomedicine and basic research by yielding cells with embryonic stem (ES) cell-like properties. The use of iPS-derived cells for cell-based therapies and modeling of human disease holds great potential. While the initial description of iPS cells involved overexpression of four transcription factors via viral vectors that integrated within genomic DNA, advances in recent years by our group and others have led to safer and higher quality iPS cells with greater efficiency. Here, we describe commonly practiced methods for non-integrating induced pluripotent stem cell generation using nucleofection of episomal reprogramming plasmids. These methods are adapted from recent studies that demonstrate increased hiPS cell reprogramming efficacy with the application of three powerful episomal hiPS cell reprogramming factor vectors and the inclusion of an accessory vector expressing EBNA1.


Subject(s)
Cellular Reprogramming Techniques , Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Plasmids/genetics , Cell Culture Techniques , Cell Separation , Fibroblasts/cytology , Fibroblasts/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Transfection
12.
Cold Spring Harb Perspect Biol ; 7(7): a008144, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26134312

ABSTRACT

The field of miRNA biology is relatively young, but its impact on our understanding of the regulation of a wide array of cell functions is far-reaching. The importance of miRNAs in development has become nearly ubiquitous, with miRNAs contributing to development of most cells and organs. Although miRNAs are clearly interwoven into known regulatory networks that control cell development, the specific modalities by which they intersect are often quite distinct and cleverly achieved. The frequently emerging theme of feed-back and feed-forward loops to either counterbalance or reinforce the gene programs that they influence is a common thread. Many of these examples of miRNAs as developmental regulators are presently found in organs with different miRNAs and targets, whereas novel, unexpected themes emerge in the context of mouse development as we learn more about this rapidly developing area of biology.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/physiology , Models, Genetic , Animals , Bone Development/genetics , Cell Differentiation/genetics , Gastrulation/genetics , Hematopoiesis/genetics , Mice , MicroRNAs/chemistry , MicroRNAs/metabolism , Muscle Development/genetics , Nervous System/embryology , Signal Transduction
13.
J Mol Cell Cardiol ; 84: 13-23, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25871831

ABSTRACT

Valvular and vascular calcification are common causes of cardiovascular morbidity and mortality. Developing effective treatments requires understanding the molecular underpinnings of these processes. Shear stress is thought to play a role in inhibiting calcification. Furthermore, NOTCH1 regulates vascular and valvular endothelium, and human mutations in NOTCH1 can cause calcific aortic valve disease. Here, we determined the genome-wide impact of altering shear stress and NOTCH signaling on human aortic valve endothelium. mRNA-sequencing of primary human aortic valve endothelial cells (HAVECs) with or without knockdown of NOTCH1, in the presence or absence of shear stress, revealed NOTCH1-dependency of the atherosclerosis-related gene connexin 40 (GJA5), and numerous repressors of endochondral ossification. Among these, matrix gla protein (MGP) is highly expressed in aortic valve and vasculature, and inhibits soft tissue calcification by sequestering bone morphogenetic proteins (BMPs). Altering NOTCH1 levels affected MGP mRNA and protein in HAVECs. Furthermore, shear stress activated NOTCH signaling and MGP in a NOTCH1-dependent manner. NOTCH1 positively regulated endothelial MGP in vivo through specific binding motifs upstream of MGP. Our studies suggest that shear stress activates NOTCH1 in primary human aortic valve endothelial cells leading to downregulation of osteoblast-like gene networks that play a role in tissue calcification.


Subject(s)
Aortic Valve Stenosis/genetics , Aortic Valve/pathology , Calcinosis/genetics , Calcium-Binding Proteins/metabolism , Endothelium, Vascular/metabolism , Extracellular Matrix Proteins/metabolism , Gene Regulatory Networks , Receptor, Notch1/metabolism , Aortic Valve Stenosis/pathology , Calcinosis/pathology , Chromatin Immunoprecipitation , Cluster Analysis , DNA/metabolism , Endothelial Cells/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Genome, Human , Humans , Protein Binding , Rheology , Sequence Analysis, RNA , Signal Transduction/genetics , Stress, Mechanical , Matrix Gla Protein
14.
J Biol Chem ; 289(20): 14263-71, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24719334

ABSTRACT

MicroRNA (miRNA) maturation is regulated by interaction of particular miRNA precursors with specific RNA-binding proteins. Following their biogenesis, mature miRNAs are incorporated into the RNA-induced silencing complex (RISC) where they interact with mRNAs to negatively regulate protein production. However, little is known about how mature miRNAs are regulated at the level of their activity. To address this, we screened for proteins differentially bound to the mature form of the miR-1 or miR-133 miRNA families. These muscle-enriched, co-transcribed miRNA pairs cooperate to suppress smooth muscle gene expression in the heart. However, they also have opposing roles, with the miR-1 family, composed of miR-1 and miR-206, promoting myogenic differentiation, whereas miR-133 maintains the progenitor state. Here, we describe a physical interaction between TDP-43, an RNA-binding protein that forms aggregates in the neuromuscular disease, amyotrophic lateral sclerosis, and the miR-1, but not miR-133, family. Deficiency of the TDP-43 Drosophila ortholog enhanced dmiR-1 activity in vivo. In mammalian cells, TDP-43 limited the activity of both miR-1 and miR-206, but not the miR-133 family, by disrupting their RISC association. Consistent with TDP-43 dampening miR-1/206 activity, protein levels of the miR-1/206 targets, IGF-1 and HDAC4, were elevated in TDP-43 transgenic mouse muscle. This occurred without corresponding Igf-1 or Hdac4 mRNA increases and despite higher miR-1 and miR-206 expression. Our findings reveal that TDP-43 negatively regulates the activity of the miR-1 family of miRNAs by limiting their bioavailability for RISC loading and suggest a processing-independent mechanism for differential regulation of miRNA activity.


Subject(s)
DNA-Binding Proteins/metabolism , MicroRNAs/metabolism , RNA-Induced Silencing Complex/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Argonaute Proteins/metabolism , Drosophila melanogaster/genetics , Humans , Male , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Protein Binding
15.
Elife ; 2: e01323, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24252873

ABSTRACT

microRNA-1 (miR-1) is an evolutionarily conserved, striated muscle-enriched miRNA. Most mammalian genomes contain two copies of miR-1, and in mice, deletion of a single locus, miR-1-2, causes incompletely penetrant lethality and subtle cardiac defects. Here, we report that deletion of miR-1-1 resulted in a phenotype similar to that of the miR-1-2 mutant. Compound miR-1 knockout mice died uniformly before weaning due to severe cardiac dysfunction. miR-1-null cardiomyocytes had abnormal sarcomere organization and decreased phosphorylation of the regulatory myosin light chain-2 (MLC2), a critical cytoskeletal regulator. The smooth muscle-restricted inhibitor of MLC2 phosphorylation, Telokin, was ectopically expressed in the myocardium, along with other smooth muscle genes. miR-1 repressed Telokin expression through direct targeting and by repressing its transcriptional regulator, Myocardin. Our results reveal that miR-1 is required for postnatal cardiac function and reinforces the striated muscle phenotype by regulating both transcriptional and effector nodes of the smooth muscle gene expression network. DOI: http://dx.doi.org/10.7554/eLife.01323.001.


Subject(s)
Gene Expression , MicroRNAs/physiology , Muscle, Smooth/metabolism , Myocardium/metabolism , Sarcomeres , Animals , Mice , Mice, Knockout , MicroRNAs/genetics , Phosphorylation
16.
Stem Cells Transl Med ; 2(3): 161-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23430692

ABSTRACT

Several mutations in αB-crystallin (CryAB), a heat shock protein with chaperone-like activities, are causally linked to skeletal and cardiac myopathies in humans. To better understand the underlying pathogenic mechanisms, we had previously generated transgenic (TG) mice expressing R120GCryAB, which recapitulated distinguishing features of the myopathic disorder (e.g., protein aggregates, hypertrophic cardiomyopathy). To determine whether induced pluripotent stem cell (iPSC)-derived cardiomyocytes, a new experimental approach for human disease modeling, would be relevant to aggregation-prone disorders, we decided to exploit the existing transgenic mouse model to derive iPSCs from tail tip fibroblasts. Several iPSC lines were generated from TG and non-TG mice and validated for pluripotency. TG iPSC-derived cardiomyocytes contained perinuclear aggregates positive for CryAB staining, whereas CryAB protein accumulated in both detergent-soluble and insoluble fractions. iPSC-derived cardiomyocytes identified by cardiac troponin T staining were significantly larger when expressing R120GCryAB at a high level in comparison with TG low expressor or non-TG cells. Expression of fetal genes such as atrial natriuretic factor, B-type natriuretic peptide, and α-skeletal α-actin, assessed by quantitative reverse transcription-polymerase chain reaction, were increased in TG cardiomyocytes compared with non-TG, indicating the activation of the hypertrophic genetic program in vitro. Our study demonstrates for the first time that differentiation of R120G iPSCs into cardiomyocytes causes protein aggregation and cellular hypertrophy, recapitulating in vitro key pathognomonic hallmarks found in both animal models and patients. Our findings pave the way for further studies exploiting this cell model system for mechanistic and therapeutic investigations.


Subject(s)
Cardiomyopathy, Hypertrophic/metabolism , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , alpha-Crystallins/metabolism , beta-Crystallins/metabolism , Actins/genetics , Actins/metabolism , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Cell Differentiation , Cell Line , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/pathology , Mice , Mice, Transgenic , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Protein Multimerization , Reverse Transcriptase Polymerase Chain Reaction , Troponin T/metabolism , alpha-Crystallins/genetics , beta-Crystallins/genetics
17.
Stem Cells ; 31(1): 92-103, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23079999

ABSTRACT

Recent evidence suggests human embryonic stem cell (hESC) and induced pluripotent stem (iPS) cell lines have differences in their epigenetic marks and transcriptomes, yet the impact of these differences on subsequent terminally differentiated cells is less well understood. Comparison of purified, homogeneous populations of somatic cells derived from multiple independent human iPS and ES lines will be required to address this critical question. Here, we report a differentiation protocol based on embryonic development that consistently yields large numbers of endothelial cells (ECs) derived from multiple hESCs or iPS cells. Mesoderm differentiation of embryoid bodies was maximized, and defined growth factors were used to generate KDR(+) EC progenitors. Magnetic purification of a KDR(+) progenitor subpopulation resulted in an expanding, homogeneous pool of ECs that expressed EC markers and had functional properties of ECs. Comparison of the transcriptomes revealed limited gene expression variability between multiple lines of human iPS-derived ECs or between lines of ES- and iPS-derived ECs. These results demonstrate a method to generate large numbers of pure human EC progenitors and differentiated ECs from pluripotent stem cells and suggest individual lineages derived from human iPS cells may have significantly less variance than their pluripotent founders.


Subject(s)
Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation/genetics , Cell Line , Cell Lineage , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genetic Variation , Humans , Mesoderm/embryology , Mesoderm/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Transcriptome/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
Circ Res ; 111(11): 1421-33, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-22955733

ABSTRACT

RATIONALE: Formation and remodeling of the vasculature during development and disease involve a highly conserved and precisely regulated network of attractants and repellants. Various signaling pathways control the behavior of endothelial cells, but their posttranscriptional dose titration by microRNAs is poorly understood. OBJECTIVE: To identify microRNAs that regulate angiogenesis. METHODS AND RESULTS: We show that the highly conserved microRNA family encoding miR-10 regulates the behavior of endothelial cells during angiogenesis by positively titrating proangiogenic signaling. Knockdown of miR-10 led to premature truncation of intersegmental vessel growth in the trunk of zebrafish larvae, whereas overexpression of miR-10 promoted angiogenic behavior in zebrafish and cultured human umbilical venous endothelial cells. We found that miR-10 functions, in part, by directly regulating the level of fms-related tyrosine kinase 1 (FLT1), a cell-surface protein that sequesters vascular endothelial growth factor, and its soluble splice variant sFLT1. The increase in FLT1/sFLT1 protein levels upon miR-10 knockdown in zebrafish and in human umbilical venous endothelial cells inhibited the angiogenic behavior of endothelial cells largely by antagonizing vascular endothelial growth factor receptor 2 signaling. CONCLUSIONS: Our study provides insights into how FLT1 and vascular endothelial growth factor receptor 2 signaling is titrated in a microRNA-mediated manner and establishes miR-10 as a potential new target for the selective modulation of angiogenesis.


Subject(s)
Endothelial Cells/metabolism , MicroRNAs/genetics , Neovascularization, Physiologic/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Animals, Genetically Modified , Base Sequence , Cell Proliferation/drug effects , Cells, Cultured , Female , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunoblotting , Larva/genetics , Larva/metabolism , Male , Mice , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zebrafish
19.
Dev Biol ; 351(1): 62-9, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21185281

ABSTRACT

Cardiogenesis involves the contributions of multiple progenitor pools, including mesoderm-derived cardiac progenitors known as the first and second heart fields. Disruption of genetic pathways regulating individual subsets of cardiac progenitors likely underlies many forms of human cardiac malformations. Hand2 is a member of the basic helix loop helix (bHLH) family of transcription factors and is expressed in numerous cell lineages that contribute to the developing heart. However, the early embryonic lethality of Hand2-null mice has precluded lineage-specific study of its function in myocardial progenitors. Here, we generated and used a floxed allele of Hand2 to ablate its expression in specific cardiac cell populations at defined developmental points. We found that Hand2 expression within the mesoderm-derived second heart field progenitors was required for their survival and deletion in this domain recapitulated the complete Hand2-null phenotype. Loss of Hand2 at later stages of development and in restricted domains of the second heart field revealed a spectrum of cardiac anomalies resembling forms of human congenital heart disease. Molecular analyses of Hand2 mutant cells revealed several genes by which Hand2 may influence expansion of the cardiac progenitors. These findings demonstrate that Hand2 is essential for survival of second heart field progenitors and that the graded loss of Hand2 function in this cardiac progenitor pool can cause a spectrum of congenital heart malformation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Heart/embryology , Stem Cells/physiology , Alleles , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Survival , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Pharynx/embryology
20.
Proc Natl Acad Sci U S A ; 107(48): 20750-5, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21071677

ABSTRACT

Cardiac and skeletal muscle development and maintenance require complex interactions between DNA-binding proteins and chromatin remodeling factors. We previously reported that Smyd1, a muscle-restricted histone methyltransferase, is essential for cardiogenesis and functions with a network of cardiac regulatory proteins. Here we show that the muscle-specific transcription factor skNAC is the major binding partner for Smyd1 in the developing heart. Targeted deletion of skNAC in mice resulted in partial embryonic lethality by embryonic day 12.5, with ventricular hypoplasia and decreased cardiomyocyte proliferation that were similar but less severe than in Smyd1 mutants. Expression of Irx4, a ventricle-specific transcription factor down-regulated in hearts lacking Smyd1, also depended on the presence of skNAC. Viable skNAC(-/-) adult mice had reduced postnatal skeletal muscle growth and impaired regenerative capacity after cardiotoxin-induced injury. Satellite cells isolated from skNAC(-/-) mice had impaired survival compared with wild-type littermate satellite cells. Our results indicate that skNAC plays a critical role in ventricular cardiomyocyte expansion and regulates postnatal skeletal muscle growth and regeneration in mice.


Subject(s)
DNA-Binding Proteins/metabolism , Heart/growth & development , Molecular Chaperones/metabolism , Muscle Development/physiology , Muscle Proteins/metabolism , Muscle, Skeletal/growth & development , Regeneration/physiology , Transcription Factors/metabolism , Animals , Animals, Newborn , Body Patterning , Cell Proliferation , DNA-Binding Proteins/genetics , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Gene Expression Regulation, Developmental , Gene Targeting , Heart Ventricles/abnormalities , Heart Ventricles/embryology , Heart Ventricles/pathology , Mice , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Proteins/genetics , Muscle, Skeletal/embryology , Muscle, Skeletal/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Organ Specificity/genetics , Organogenesis/genetics , Protein Binding , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...