Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nature ; 626(7997): 119-127, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38200310

ABSTRACT

The evolution of reproductive barriers is the first step in the formation of new species and can help us understand the diversification of life on Earth. These reproductive barriers often take the form of hybrid incompatibilities, in which alleles derived from two different species no longer interact properly in hybrids1-3. Theory predicts that hybrid incompatibilities may be more likely to arise at rapidly evolving genes4-6 and that incompatibilities involving multiple genes should be common7,8, but there has been sparse empirical data to evaluate these predictions. Here we describe a mitonuclear incompatibility involving three genes whose protein products are in physical contact within respiratory complex I of naturally hybridizing swordtail fish species. Individuals homozygous for mismatched protein combinations do not complete embryonic development or die as juveniles, whereas those heterozygous for the incompatibility have reduced complex I function and unbalanced representation of parental alleles in the mitochondrial proteome. We find that the effects of different genetic interactions on survival are non-additive, highlighting subtle complexity in the genetic architecture of hybrid incompatibilities. Finally, we document the evolutionary history of the genes involved, showing signals of accelerated evolution and evidence that an incompatibility has been transferred between species via hybridization.


Subject(s)
Cell Nucleus , Electron Transport Complex I , Fishes , Genes, Lethal , Genetic Speciation , Hybridization, Genetic , Mitochondrial Proteins , Animals , Alleles , Electron Transport Complex I/genetics , Fishes/classification , Fishes/embryology , Fishes/genetics , Fishes/growth & development , Homozygote , Genes, Lethal/genetics , Species Specificity , Embryonic Development/genetics , Mitochondrial Proteins/genetics , Cell Nucleus/genetics , Heterozygote , Evolution, Molecular
2.
medRxiv ; 2023 Nov 05.
Article in English | MEDLINE | ID: mdl-37961166

ABSTRACT

Patients with mitochondrial disorders present with clinically diverse symptoms, largely driven by heterogeneous mutations in mitochondrial-encoded and nuclear-encoded mitochondrial genes. These mutations ultimately lead to complex biochemical disorders with a myriad of clinical manifestations, often accumulating during childhood on into adulthood, contributing to life-altering and sometimes fatal events. It is therefore important to diagnose and characterize the associated disorders for each mitochondrial mutation as early as possible since medical management might be able to improve the quality and longevity of life in mitochondrial disease patients. Here we identify a novel mitochondrial variant in a mitochondrial transfer RNA for histidine (mt-tRNA-his) [m.12148T>C], that is associated with the development of ocular, aural, neurological, renal, and muscular dysfunctions. We provide a detailed account of a family harboring this mutation, as well as the molecular underpinnings contributing to cellular and mitochondrial dysfunction. In conclusion, this investigation provides clinical, biochemical, and morphological evidence of the pathogenicity of m.12148T>C. We highlight the importance of multiple tissue testing and in vitro disease modeling in diagnosing mitochondrial disease.

3.
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
4.
Hum Genomics ; 16(1): 55, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36357925

ABSTRACT

BACKGROUND: Cardiomyopathies are a leading cause of progressive heart failure and sudden cardiac death; however, their genetic aetiology remains poorly understood. We hypothesised that variants in noncoding regulatory regions and oligogenic inheritance mechanisms may help close the diagnostic gap. METHODS: We first analysed whole-genome sequencing data of 143 parent-offspring trios from Genomics England 100,000 Genomes Project. We used gene panel testing and a phenotype-based, variant prioritisation framework called Exomiser to identify candidate genes in trios. To assess the contribution of noncoding DNVs to cardiomyopathies, we intersected DNVs with open chromatin sequences from single-cell ATAC-seq data of cardiomyocytes. We also performed a case-control analysis in an exome-negative cohort, including 843 probands and 19,467 controls, to assess the association between noncoding variants in known cardiomyopathy genes and disease. RESULTS: In the trio analysis, a definite or probable genetic diagnosis was identified in 21 probands according to the American College of Medical Genetics guidelines. We identified novel DNVs in diagnostic-grade genes (RYR2, TNNT2, PTPN11, MYH7, LZR1, NKX2-5), and five cases harbouring a combination of prioritised variants, suggesting that oligogenic inheritance and genetic modifiers contribute to cardiomyopathies. Phenotype-based ranking of candidate genes identified in noncoding DNV analysis revealed JPH2 as the top candidate. Moreover, a case-control analysis revealed an enrichment of rare noncoding variants in regulatory elements of cardiomyopathy genes (p = .035, OR = 1.43, 95% Cl = 1.095-1.767) versus controls. Of the 25 variants associated with disease  (p< 0.5), 23 are novel and nine are predicted to disrupt transcription factor binding motifs. CONCLUSION: Our results highlight complex genetic mechanisms in cardiomyopathies and reveal novel genes for future investigations.


Subject(s)
Cardiomyopathies , Genetic Predisposition to Disease , Humans , Cardiomyopathies/genetics , Exome , Phenotype , Regulatory Sequences, Nucleic Acid
5.
Circulation ; 146(10): 770-787, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35938400

ABSTRACT

BACKGROUND: GATA4 (GATA-binding protein 4), a zinc finger-containing, DNA-binding transcription factor, is essential for normal cardiac development and homeostasis in mice and humans, and mutations in this gene have been reported in human heart defects. Defects in alternative splicing are associated with many heart diseases, yet relatively little is known about how cell type- or cell state-specific alternative splicing is achieved in the heart. Here, we show that GATA4 regulates cell type-specific splicing through direct interaction with RNA and the spliceosome in human induced pluripotent stem cell-derived cardiac progenitors. METHODS: We leveraged a combination of unbiased approaches including affinity purification of GATA4 and mass spectrometry, enhanced cross-linking with immunoprecipitation, electrophoretic mobility shift assays, in vitro splicing assays, and unbiased transcriptomic analysis to uncover GATA4's novel function as a splicing regulator in human induced pluripotent stem cell-derived cardiac progenitors. RESULTS: We found that GATA4 interacts with many members of the spliceosome complex in human induced pluripotent stem cell-derived cardiac progenitors. Enhanced cross-linking with immunoprecipitation demonstrated that GATA4 also directly binds to a large number of mRNAs through defined RNA motifs in a sequence-specific manner. In vitro splicing assays indicated that GATA4 regulates alternative splicing through direct RNA binding, resulting in functionally distinct protein products. Correspondingly, knockdown of GATA4 in human induced pluripotent stem cell-derived cardiac progenitors resulted in differential alternative splicing of genes involved in cytoskeleton organization and calcium ion import, with functional consequences associated with the protein isoforms. CONCLUSIONS: This study shows that in addition to its well described transcriptional function, GATA4 interacts with members of the spliceosome complex and regulates cell type-specific alternative splicing via sequence-specific interactions with RNA. Several genes that have splicing regulated by GATA4 have functional consequences and many are associated with dilated cardiomyopathy, suggesting a novel role for GATA4 in achieving the necessary cardiac proteome in normal and stress-responsive conditions.


Subject(s)
GATA4 Transcription Factor , Induced Pluripotent Stem Cells , Alternative Splicing , Animals , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Heart , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Myocytes, Cardiac/metabolism , RNA/genetics , RNA/metabolism
6.
Front Genet ; 13: 888025, 2022.
Article in English | MEDLINE | ID: mdl-35571054

ABSTRACT

There is considerable variability in the susceptibility and progression for COVID-19 and it appears to be strongly correlated with age, gender, ethnicity and pre-existing health conditions. However, to our knowledge, cohort studies of COVID-19 in clinically vulnerable groups are lacking. Host genetics has also emerged as a major risk factor for COVID-19, and variation in the ACE2 receptor, which facilitates entry of the SARS-CoV-2 virus into the cell, has become a major focus of attention. Thus, we interrogated an ethnically diverse cohort of National Health Service (NHS) patients in the United Kingdom (United Kingdom) to assess the association between variants in the ACE2 locus and COVID-19 risk. We analysed whole-genome sequencing (WGS) data of 1,837 cases who were tested positive for SARS-CoV-2, and 37,207 controls who were not tested, from the UK's 100,000 Genomes Project (100KGP) for the presence of ACE2 coding variants and extract expression quantitative trait loci (eQTLs). We identified a splice site variant (rs2285666) associated with increased ACE2 expression with an overrepresentation in SARS-CoV-2 positive patients relative to 100KGP controls (p = 0.015), and in hospitalised European patients relative to outpatients in intra-ethnic comparisons (p = 0.029). We also compared the prevalence of 288 eQTLs, of which 23 were enriched in SARS-CoV-2 positive patients. The eQTL rs12006793 had the largest effect size (d = 0.91), which decreases ACE2 expression and is more prevalent in controls, thus potentially reducing the risk of COVID-19. We identified three novel nonsynonymous variants predicted to alter ACE2 function, and showed that three variants (p.K26R, p. H378R, p. Y515N) alter receptor affinity for the viral Spike (S) protein. Variant p. N720D, more prevalent in the European population (p < 0.001), potentially increases viral entry by affecting the ACE2-TMPRSS2 complex. The spectrum of genetic variants in ACE2 may inform risk stratification of COVID-19 patients and could partially explain the differences in disease susceptibility and severity among different ethnic groups.

7.
Cell ; 185(5): 794-814.e30, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35182466

ABSTRACT

Congenital heart disease (CHD) is present in 1% of live births, yet identification of causal mutations remains challenging. We hypothesized that genetic determinants for CHDs may lie in the protein interactomes of transcription factors whose mutations cause CHDs. Defining the interactomes of two transcription factors haplo-insufficient in CHD, GATA4 and TBX5, within human cardiac progenitors, and integrating the results with nearly 9,000 exomes from proband-parent trios revealed an enrichment of de novo missense variants associated with CHD within the interactomes. Scoring variants of interactome members based on residue, gene, and proband features identified likely CHD-causing genes, including the epigenetic reader GLYR1. GLYR1 and GATA4 widely co-occupied and co-activated cardiac developmental genes, and the identified GLYR1 missense variant disrupted interaction with GATA4, impairing in vitro and in vivo function in mice. This integrative proteomic and genetic approach provides a framework for prioritizing and interrogating genetic variants in heart disease.


Subject(s)
GATA4 Transcription Factor/metabolism , Heart Defects, Congenital , Nuclear Proteins/metabolism , Oxidoreductases/metabolism , Transcription Factors , Animals , Heart Defects, Congenital/genetics , Mice , Mutation , Proteomics , T-Box Domain Proteins/genetics , Transcription Factors/genetics
8.
Nature ; 595(7867): 438-443, 2021 07.
Article in English | MEDLINE | ID: mdl-34163071

ABSTRACT

In diseased organs, stress-activated signalling cascades alter chromatin, thereby triggering maladaptive cell state transitions. Fibroblast activation is a common stress response in tissues that worsens lung, liver, kidney and heart disease, yet its mechanistic basis remains unclear1,2. Pharmacological inhibition of bromodomain and extra-terminal domain (BET) proteins alleviates cardiac dysfunction3-7, providing a tool to interrogate and modulate cardiac cell states as a potential therapeutic approach. Here we use single-cell epigenomic analyses of hearts dynamically exposed to BET inhibitors to reveal a reversible transcriptional switch that underlies the activation of fibroblasts. Resident cardiac fibroblasts demonstrated robust toggling between the quiescent and activated state in a manner directly correlating with BET inhibitor exposure and cardiac function. Single-cell chromatin accessibility revealed previously undescribed DNA elements, the accessibility of which dynamically correlated with cardiac performance. Among the most dynamic elements was an enhancer that regulated the transcription factor MEOX1, which was specifically expressed in activated fibroblasts, occupied putative regulatory elements of a broad fibrotic gene program and was required for TGFß-induced fibroblast activation. Selective CRISPR inhibition of the single most dynamic cis-element within the enhancer blocked TGFß-induced Meox1 activation. We identify MEOX1 as a central regulator of fibroblast activation associated with cardiac dysfunction and demonstrate its upregulation after activation of human lung, liver and kidney fibroblasts. The plasticity and specificity of BET-dependent regulation of MEOX1 in tissue fibroblasts provide previously unknown trans- and cis-targets for treating fibrotic disease.


Subject(s)
Enhancer Elements, Genetic , Fibroblasts/cytology , Heart Diseases/genetics , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Chromatin/metabolism , Epigenomics , Gene Expression Regulation , Humans , Mice , Proteins/antagonists & inhibitors , Single-Cell Analysis , Transcriptome , Transforming Growth Factor beta/metabolism
9.
Science ; 371(6530)2021 02 12.
Article in English | MEDLINE | ID: mdl-33303684

ABSTRACT

Mapping the gene-regulatory networks dysregulated in human disease would allow the design of network-correcting therapies that treat the core disease mechanism. However, small molecules are traditionally screened for their effects on one to several outputs at most, biasing discovery and limiting the likelihood of true disease-modifying drug candidates. Here, we developed a machine-learning approach to identify small molecules that broadly correct gene networks dysregulated in a human induced pluripotent stem cell (iPSC) disease model of a common form of heart disease involving the aortic valve (AV). Gene network correction by the most efficacious therapeutic candidate, XCT790, generalized to patient-derived primary AV cells and was sufficient to prevent and treat AV disease in vivo in a mouse model. This strategy, made feasible by human iPSC technology, network analysis, and machine learning, may represent an effective path for drug discovery.


Subject(s)
Aortic Valve Disease/drug therapy , Aortic Valve Stenosis/drug therapy , Aortic Valve/pathology , Calcinosis/drug therapy , Gene Regulatory Networks/drug effects , Machine Learning , Nitriles/pharmacology , Nitriles/therapeutic use , Thiazoles/pharmacology , Thiazoles/therapeutic use , Algorithms , Animals , Aortic Valve/drug effects , Aortic Valve/metabolism , Aortic Valve/physiopathology , Aortic Valve Disease/genetics , Aortic Valve Disease/physiopathology , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/physiopathology , Calcinosis/genetics , Calcinosis/physiopathology , Disease Models, Animal , Drug Discovery , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Haploinsufficiency , Humans , Induced Pluripotent Stem Cells , Mice, Inbred C57BL , RNA-Seq , Receptor, Notch1/genetics , Small Molecule Libraries
10.
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
11.
Nature ; 572(7767): 120-124, 2019 08.
Article in English | MEDLINE | ID: mdl-31341279

ABSTRACT

Organogenesis involves integration of diverse cell types; dysregulation of cell-type-specific gene networks results in birth defects, which affect 5% of live births. Congenital heart defects are the most common malformations, and result from disruption of discrete subsets of cardiac progenitor cells1, but the transcriptional changes in individual progenitors that lead to organ-level defects remain unknown. Here we used single-cell RNA sequencing to interrogate early cardiac progenitor cells as they become specified during normal and abnormal cardiogenesis, revealing how dysregulation of specific cellular subpopulations has catastrophic consequences. A network-based computational method for single-cell RNA-sequencing analysis that predicts lineage-specifying transcription factors2,3 identified Hand2 as a specifier of outflow tract cells but not right ventricular cells, despite the failure of right ventricular formation in Hand2-null mice4. Temporal single-cell-transcriptome analysis of Hand2-null embryos revealed failure of outflow tract myocardium specification, whereas right ventricular myocardium was specified but failed to properly differentiate and migrate. Loss of Hand2 also led to dysregulation of retinoic acid signalling and disruption of anterior-posterior patterning of cardiac progenitors. This work reveals transcriptional determinants that specify fate and differentiation in individual cardiac progenitor cells, and exposes mechanisms of disrupted cardiac development at single-cell resolution, providing a framework for investigating congenital heart defects.


Subject(s)
Heart Defects, Congenital/embryology , Heart Defects, Congenital/pathology , Heart/embryology , Single-Cell Analysis , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cell Movement , Cluster Analysis , Female , Heart Defects, Congenital/genetics , Male , Mice , Sequence Analysis, RNA , Tretinoin/metabolism
12.
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
13.
Nat Genet ; 50(2): 250-258, 2018 02.
Article in English | MEDLINE | ID: mdl-29358654

ABSTRACT

Transcription factors (TFs) direct developmental transitions by binding to target DNA sequences, influencing gene expression and establishing complex gene-regultory networks. To systematically determine the molecular components that enable or constrain TF activity, we investigated the genomic occupancy of FOXA2, GATA4 and OCT4 in several cell types. Despite their classification as pioneer factors, all three TFs exhibit cell-type-specific binding, even when supraphysiologically and ectopically expressed. However, FOXA2 and GATA4 can be distinguished by low enrichment at loci that are highly occupied by these factors in alternative cell types. We find that expression of additional cofactors increases enrichment at a subset of these sites. Finally, FOXA2 occupancy and changes to DNA accessibility can occur in G1-arrested cells, but subsequent loss of DNA methylation requires DNA replication.


Subject(s)
DNA/metabolism , Epigenesis, Genetic/physiology , Gene Regulatory Networks/physiology , Transcription Factors/metabolism , A549 Cells , Binding Sites/genetics , Cell Lineage/drug effects , Cell Lineage/genetics , Cells, Cultured , Computational Biology , DNA/genetics , Epistasis, Genetic/physiology , GATA4 Transcription Factor/metabolism , Gene Expression Regulation , Genes, Switch , HEK293 Cells , Hep G2 Cells , Hepatocyte Nuclear Factor 3-beta/metabolism , Humans , Octamer Transcription Factor-3/metabolism , Protein Binding
14.
Proc Natl Acad Sci U S A ; 114(19): 4969-4974, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28438991

ABSTRACT

The spinal cord consists of multiple neuronal cell types that are critical to motor control and arise from distinct progenitor domains in the developing neural tube. Excitatory V2a interneurons in particular are an integral component of central pattern generators that control respiration and locomotion; however, the lack of a robust source of human V2a interneurons limits the ability to molecularly profile these cells and examine their therapeutic potential to treat spinal cord injury (SCI). Here, we report the directed differentiation of CHX10+ V2a interneurons from human pluripotent stem cells (hPSCs). Signaling pathways (retinoic acid, sonic hedgehog, and Notch) that pattern the neural tube were sequentially perturbed to identify an optimized combination of small molecules that yielded ∼25% CHX10+ cells in four hPSC lines. Differentiated cultures expressed much higher levels of V2a phenotypic markers (CHX10 and SOX14) than other neural lineage markers. Over time, CHX10+ cells expressed neuronal markers [neurofilament, NeuN, and vesicular glutamate transporter 2 (VGlut2)], and cultures exhibited increased action potential frequency. Single-cell RNAseq analysis confirmed CHX10+ cells within the differentiated population, which consisted primarily of neurons with some glial and neural progenitor cells. At 2 wk after transplantation into the spinal cord of mice, hPSC-derived V2a cultures survived at the site of injection, coexpressed NeuN and VGlut2, extended neurites >5 mm, and formed putative synapses with host neurons. These results provide a description of V2a interneurons differentiated from hPSCs that may be used to model central nervous system development and serve as a potential cell therapy for SCI.


Subject(s)
Antigens, Differentiation/biosynthesis , Cell Differentiation , Human Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Neurons/cytology
15.
Stem Cell Reports ; 7(5): 983-997, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27773702

ABSTRACT

Following injury, adult skeletal muscle undergoes a well-coordinated sequence of molecular and physiological events to promote repair and regeneration. However, a thorough understanding of the in vivo epigenomic and transcriptional mechanisms that control these reparative events is lacking. To address this, we monitored the in vivo dynamics of three histone modifications and coding and noncoding RNA expression throughout the regenerative process in a mouse model of traumatic muscle injury. We first illustrate how both coding and noncoding RNAs in tissues and sorted satellite cells are modified and regulated during various stages after trauma. Next, we use chromatin immunoprecipitation followed by sequencing to evaluate the chromatin state of cis-regulatory elements (promoters and enhancers) and view how these elements evolve and influence various muscle repair and regeneration transcriptional programs. These results provide a comprehensive view of the central factors that regulate muscle regeneration and underscore the multiple levels through which both transcriptional and epigenetic patterns are regulated to enact appropriate repair and regeneration.


Subject(s)
Chromatin Assembly and Disassembly , Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Regeneration/genetics , Transcription, Genetic , Animals , Male , Mice , MicroRNAs/genetics , RNA, Messenger/genetics , Wound Healing/genetics
16.
Nat Cell Biol ; 18(1): 3-5, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26693916

ABSTRACT

The heart is a complex organ, consisting of multiple cell types that coordinately regulate blood flow. Reciprocal Notch pathway signalling in endocardial and myocardial cells is now shown to promote maturation of the ventricular chambers. These insights reveal mechanisms that, when disrupted, can lead to cardiomyopathies.


Subject(s)
Heart Ventricles/metabolism , Organogenesis/physiology , Receptors, Notch/metabolism , Humans
17.
Nat Biotechnol ; 33(11): 1182-92, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26501952

ABSTRACT

Research on human pluripotent stem cells has been hampered by the lack of a standardized, quantitative, scalable assay of pluripotency. We previously described an assay called ScoreCard that used gene expression signatures to quantify differentiation efficiency. Here we report an improved version of the assay based on qPCR that enables faster, more quantitative assessment of functional pluripotency. We provide an in-depth characterization of the revised signature panel (commercially available as the TaqMan hPSC Scorecard Assay) through embryoid body and directed differentiation experiments as well as a detailed comparison to the teratoma assay. We further show that the improved ScoreCard enables a wider range of applications, such as screening of small molecules, genetic perturbations and assessment of culture conditions. Our approach can be extended beyond stem cell applications to characterize and assess the utility of other cell types and lineages.


Subject(s)
Cell Differentiation/genetics , Computational Biology/methods , Embryoid Bodies/physiology , Pluripotent Stem Cells/physiology , Polymerase Chain Reaction/methods , Animals , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Humans , Mice , Neoplasms, Experimental/pathology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Teratoma/pathology
18.
Sci Rep ; 5: 13885, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26381351

ABSTRACT

Traumatic lower-limb musculoskeletal injuries are pervasive amongst athletes and the military and typically an individual returns to activity prior to fully healing, increasing a predisposition for additional injuries and chronic pain. Monitoring healing progression after a musculoskeletal injury typically involves different types of imaging but these approaches suffer from several disadvantages. Isolating and profiling transcripts from the injured site would abrogate these shortcomings and provide enumerative insights into the regenerative potential of an individual's muscle after injury. In this study, a traumatic injury was administered to a mouse model and healing progression was examined from 3 hours to 1 month using high-throughput RNA-Sequencing (RNA-Seq). Comprehensive dissection of the genome-wide datasets revealed the injured site to be a dynamic, heterogeneous environment composed of multiple cell types and thousands of genes undergoing significant expression changes in highly regulated networks. Four independent approaches were used to determine the set of genes, isoforms, and genetic pathways most characteristic of different time points post-injury and two novel approaches were developed to classify injured tissues at different time points. These results highlight the possibility to quantitatively track healing progression in situ via transcript profiling using high- throughput sequencing.


Subject(s)
Gene Expression Profiling , Lower Extremity , Muscle, Skeletal/injuries , Muscle, Skeletal/metabolism , Transcriptome , Wound Healing/genetics , Animals , Complement System Proteins/immunology , Complement System Proteins/metabolism , Computational Biology/methods , Fibroblasts/metabolism , Gene Expression Regulation , Gene Regulatory Networks , Male , Mice , Molecular Sequence Annotation , Muscle, Skeletal/pathology , Phenotype , Receptors, Notch/metabolism , Reproducibility of Results , Signal Transduction , Support Vector Machine , Wnt Proteins/metabolism
19.
Cell ; 162(2): 412-424, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26186193

ABSTRACT

Induced pluripotency is a promising avenue for disease modeling and therapy, but the molecular principles underlying this process, particularly in human cells, remain poorly understood due to donor-to-donor variability and intercellular heterogeneity. Here, we constructed and characterized a clonal, inducible human reprogramming system that provides a reliable source of cells at any stage of the process. This system enabled integrative transcriptional and epigenomic analysis across the human reprogramming timeline at high resolution. We observed distinct waves of gene network activation, including the ordered re-activation of broad developmental regulators followed by early embryonic patterning genes and culminating in the emergence of a signature reminiscent of pre-implantation stages. Moreover, complementary functional analyses allowed us to identify and validate novel regulators of the reprogramming process. Altogether, this study sheds light on the molecular underpinnings of induced pluripotency in human cells and provides a robust cell platform for further studies. PAPERCLIP.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Chromatin/metabolism , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Gene Expression Profiling , Histone Demethylases/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism
20.
Nat Genet ; 47(5): 469-78, 2015 May.
Article in English | MEDLINE | ID: mdl-25822089

ABSTRACT

DNA methylation is a key epigenetic modification involved in regulating gene expression and maintaining genomic integrity. Here we inactivated all three catalytically active DNA methyltransferases (DNMTs) in human embryonic stem cells (ESCs) using CRISPR/Cas9 genome editing to further investigate the roles and genomic targets of these enzymes. Disruption of DNMT3A or DNMT3B individually as well as of both enzymes in tandem results in viable, pluripotent cell lines with distinct effects on the DNA methylation landscape, as assessed by whole-genome bisulfite sequencing. Surprisingly, in contrast to findings in mouse, deletion of DNMT1 resulted in rapid cell death in human ESCs. To overcome this immediate lethality, we generated a doxycycline-responsive tTA-DNMT1* rescue line and readily obtained homozygous DNMT1-mutant lines. However, doxycycline-mediated repression of exogenous DNMT1* initiates rapid, global loss of DNA methylation, followed by extensive cell death. Our data provide a comprehensive characterization of DNMT-mutant ESCs, including single-base genome-wide maps of the targets of these enzymes.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Embryonic Stem Cells/enzymology , Animals , Apoptosis , Base Sequence , Catalytic Domain , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Coculture Techniques , CpG Islands , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methyltransferase 3A , Embryonic Stem Cells/physiology , Epigenesis, Genetic , Gene Expression , Gene Knockout Techniques , Humans , Mice , DNA Methyltransferase 3B
SELECTION OF CITATIONS
SEARCH DETAIL
...