Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Stem Cell Reports ; 18(1): 220-236, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36525964

ABSTRACT

Titin-truncating variants (TTNtv) are the single largest genetic cause of dilated cardiomyopathy (DCM). In this study we modeled disease phenotypes of A-band TTNtv-induced DCM in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) using genome editing and tissue engineering technologies. Transcriptomic, cellular, and micro-tissue studies revealed that A-band TTNtv hiPSC-CMs exhibit pathogenic proteinopathy, sarcomere defects, aberrant Na+ channel activities, and contractile dysfunction. These phenotypes establish a dual mechanism of poison peptide effect and haploinsufficiency that collectively contribute to DCM pathogenesis. However, TTNtv cellular defects did not interfere with the function of the core contractile machinery, the actin-myosin-troponin-Ca2+ complex, and preserved the therapeutic mechanism of sarcomere modulators. Treatment of TTNtv cardiac micro-tissues with investigational sarcomere modulators augmented contractility and resulted in sustained transcriptomic changes that promote reversal of DCM disease signatures. Together, our findings elucidate the underlying pathogenic mechanisms of A-band TTNtv-induced DCM and demonstrate the validity of sarcomere modulators as potential therapeutics.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Humans , Myocytes, Cardiac/pathology , Sarcomeres , Induced Pluripotent Stem Cells/pathology , Connectin/genetics , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Myocardial Contraction
2.
Sci Rep ; 9(1): 8833, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31222070

ABSTRACT

In response to environmental and nutrient stress, adipose tissues must establish a new homeostatic state. Here we show that cold exposure of obese mice triggers an adaptive tissue remodeling in visceral adipose tissue (VAT) that involves extracellular matrix deposition, angiogenesis, sympathetic innervation, and adipose tissue browning. Obese VAT is predominated by pro-inflammatory M1 macrophages; cold exposure induces an M1-to-M2 shift in macrophage composition and dramatic changes in macrophage gene expression in both M1 and M2 macrophages. Antibody-mediated CSF1R blocking prevented the cold-induced recruitment of adipose tissue M2 macrophages, suggesting the role of CSF1R signaling in the process. These cold-induced effects in obese VAT are phenocopied by an administration of the FGF21-mimetic antibody, consistent with its action to stimulate sympathetic nerves. Collectively, these studies illuminate adaptive visceral adipose tissue plasticity in obese mice in response to cold stress and antibody-based metabolic therapy.


Subject(s)
Adaptation, Physiological , Antibodies/pharmacology , Cold-Shock Response , Intra-Abdominal Fat/physiology , Animals , Cell Movement , Fibroblast Growth Factors/immunology , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Obese , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Stromal Cells/physiology
3.
Proc Natl Acad Sci U S A ; 114(23): 5800-5807, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28584128

ABSTRACT

T-cell development from hematopoietic progenitors depends on multiple transcription factors, mobilized and modulated by intrathymic Notch signaling. Key aspects of T-cell specification network architecture have been illuminated through recent reports defining roles of transcription factors PU.1, GATA-3, and E2A, their interactions with Notch signaling, and roles of Runx1, TCF-1, and Hes1, providing bases for a comprehensively updated model of the T-cell specification gene regulatory network presented herein. However, the role of lineage commitment factor Bcl11b has been unclear. We use self-organizing maps on 63 RNA-seq datasets from normal and perturbed T-cell development to identify functional targets of Bcl11b during commitment and relate them to other regulomes. We show that both activation and repression target genes can be bound by Bcl11b in vivo, and that Bcl11b effects overlap with E2A-dependent effects. The newly clarified role of Bcl11b distinguishes discrete components of commitment, resolving how innate lymphoid, myeloid, and dendritic, and B-cell fate alternatives are excluded by different mechanisms.


Subject(s)
Cell Differentiation/genetics , Gene Regulatory Networks , Repressor Proteins/physiology , T-Lymphocytes/cytology , Tumor Suppressor Proteins/physiology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Notch , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
4.
Nat Immunol ; 17(8): 956-65, 2016 08.
Article in English | MEDLINE | ID: mdl-27376470

ABSTRACT

During T cell development, multipotent progenitors relinquish competence for other fates and commit to the T cell lineage by turning on Bcl11b, which encodes a transcription factor. To clarify lineage commitment mechanisms, we followed developing T cells at the single-cell level using Bcl11b knock-in fluorescent reporter mice. Notch signaling and Notch-activated transcription factors collaborate to activate Bcl11b expression irrespectively of Notch-dependent proliferation. These inputs work via three distinct, asynchronous mechanisms: an early locus 'poising' function dependent on TCF-1 and GATA-3, a stochastic-permissivity function dependent on Notch signaling, and a separate amplitude-control function dependent on Runx1, a factor already present in multipotent progenitors. Despite their necessity for Bcl11b expression, these inputs act in a stage-specific manner, providing a multitiered mechanism for developmental gene regulation.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 1-alpha/metabolism , Lymphopoiesis/genetics , Receptors, Notch/metabolism , Repressor Proteins/metabolism , T-Lymphocytes/physiology , Tumor Suppressor Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Tracking , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , GATA3 Transcription Factor/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Repressor Proteins/genetics , Signal Transduction , Single-Cell Analysis , Tumor Suppressor Proteins/genetics
5.
Immunol Rev ; 271(1): 72-97, 2016 May.
Article in English | MEDLINE | ID: mdl-27088908

ABSTRACT

The pathway to generate T cells from hematopoietic stem cells guides progenitors through a succession of fate choices while balancing differentiation progression against proliferation, stage to stage. Many elements of the regulatory system that controls this process are known, but the requirement for multiple, functionally distinct transcription factors needs clarification in terms of gene network architecture. Here, we compare the features of the T-cell specification system with the rule sets underlying two other influential types of gene network models: first, the combinatorial, hierarchical regulatory systems that generate the orderly, synchronized increases in complexity in most invertebrate embryos; second, the dueling 'master regulator' systems that are commonly used to explain bistability in microbial systems and in many fate choices in terminal differentiation. The T-cell specification process shares certain features with each of these prevalent models but differs from both of them in central respects. The T-cell system is highly combinatorial but also highly dose-sensitive in its use of crucial regulatory factors. The roles of these factors are not always T-lineage-specific, but they balance and modulate each other's activities long before any mutually exclusive silencing occurs. T-cell specification may provide a new hybrid model for gene networks in vertebrate developmental systems.


Subject(s)
Cell Differentiation , Hematopoiesis , Hematopoietic Stem Cells/physiology , Immune System/embryology , T-Lymphocytes/physiology , Animals , Cell Lineage , Gene Expression Regulation, Developmental , Gene Regulatory Networks/immunology , Humans , Immune System/growth & development , Models, Biological
6.
J Immunol ; 193(7): 3470-91, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25172496

ABSTRACT

GATA-3 expression is crucial for T cell development and peaks during commitment to the T cell lineage, midway through the CD4(-)CD8(-) (double-negative [DN]) stages 1-3. We used RNA interference and conditional deletion to reduce GATA-3 protein acutely at specific points during T cell differentiation in vitro. Even moderate GATA-3 reduction killed DN1 cells, delayed progression to the DN2 stage, skewed DN2 gene regulation, and blocked appearance of the DN3 phenotype. Although a Bcl-2 transgene rescued DN1 survival and improved DN2 cell generation, it did not restore DN3 differentiation. Gene expression analyses (quantitative PCR, RNA sequencing) showed that GATA-3-deficient DN2 cells quickly upregulated genes, including Spi1 (PU.1) and Bcl11a, and downregulated genes, including Cpa3, Ets1, Zfpm1, Bcl11b, Il9r, and Il17rb with gene-specific kinetics and dose dependencies. These targets could mediate two distinct roles played by GATA-3 in lineage commitment, as revealed by removing wild-type or GATA-3-deficient early T lineage cells from environmental Notch signals. GATA-3 worked as a potent repressor of B cell potential even at low expression levels, so that only full deletion of GATA-3 enabled pro-T cells to reveal B cell potential. The ability of GATA-3 to block B cell development did not require T lineage commitment factor Bcl11b. In prethymic multipotent precursors, however, titration of GATA-3 activity using tamoxifen-inducible GATA-3 showed that GATA-3 inhibits B and myeloid developmental alternatives at different threshold doses. Furthermore, differential impacts of a GATA-3 obligate repressor construct imply that B and myeloid development are inhibited through distinct transcriptional mechanisms. Thus, the pattern of GATA-3 expression sequentially produces B lineage exclusion, T lineage progression, and myeloid-lineage exclusion for commitment.


Subject(s)
GATA3 Transcription Factor/immunology , Precursor Cells, T-Lymphoid/immunology , Signal Transduction/immunology , Up-Regulation/immunology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cell Line , GATA3 Transcription Factor/genetics , Mice , Myeloid Cells/cytology , Myeloid Cells/immunology , Precursor Cells, T-Lymphoid/cytology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , Receptors, Notch/genetics , Receptors, Notch/immunology , Repressor Proteins/genetics , Repressor Proteins/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , Tamoxifen/pharmacology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology , Up-Regulation/drug effects , Up-Regulation/genetics
7.
Blood ; 122(6): 902-11, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23741008

ABSTRACT

Bcl11b is a T-cell specific gene in hematopoiesis that begins expression during T-lineage commitment and is required for this process. Aberrant expression of BCL11B or proto-oncogene translocation to the vicinity of BCL11B can be a contributing factor in human T-ALL. To identify the mechanism that controls its distinctive T-lineage expression, we corrected the identified Bcl11b transcription start site and mapped a cell-type-specific differentially methylated region bracketing the Bcl11b promoter. We identified a 1.9-kb region 850 kb downstream of Bcl11b, "Major Peak," distinguished by its dynamic histone marking pattern in development that mirrors the pattern at the Bcl11b promoter. Looping interactions between promoter-proximal elements including the differentially methylated region and downstream elements in the Major Peak are required to recapitulate the T-cell specific expression of Bcl11b in stable reporter assays. Functional dissection of the Major Peak sequence showed distinct subregions, in which TCF-1 sites and a conserved element were required for T-lineage-specific activation and silencing in non-T cells. A bacterial artificial chromosome encompassing the full Bcl11b gene still required the addition of the Major Peak to exhibit T-cell specific expression. Thus, promoter-proximal and Major Peak sequences are cis-regulatory elements that interact over 850 kb to control expression of Bcl11b in hematopoietic cells.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Repressor Proteins/genetics , T-Lymphocytes/cytology , Tumor Suppressor Proteins/genetics , Animals , Cell Lineage , CpG Islands , DNA Methylation , Gene Silencing , Genes, Reporter , Hematopoietic Stem Cells , Histones/metabolism , Mice , Promoter Regions, Genetic , Proto-Oncogene Mas , Repressor Proteins/metabolism , T-Lymphocytes/immunology , Transcription Factors/genetics , Tumor Suppressor Proteins/metabolism
8.
J Immunol ; 190(7): 3276-88, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23440410

ABSTRACT

NOD mice exhibit major defects in the earliest stages of T cell development in the thymus. Genome-wide genetic and transcriptome analyses were used to investigate the origins and consequences of an early T cell developmental checkpoint breakthrough in Rag1-deficient NOD mice. Quantitative trait locus analysis mapped the presence of checkpoint breakthrough cells to several known NOD diabetes susceptibility regions, particularly insulin-dependent diabetes susceptibility genes (Idd)9/11 on chromosome 4, suggesting common genetic origins for T cell defects affecting this trait and autoimmunity. Genome-wide RNA deep-sequencing of NOD and B6 Rag1-deficient thymocytes revealed the effects of genetic background prior to breakthrough, as well as the cellular consequences of the breakthrough. Transcriptome comparison between the two strains showed enrichment in differentially expressed signal transduction genes, prominently tyrosine kinase and actin-binding genes, in accord with their divergent sensitivities to activating signals. Emerging NOD breakthrough cells aberrantly expressed both stem cell-associated proto-oncogenes, such as Lmo2, Hhex, Lyl1, and Kit, which are normally repressed at the commitment checkpoint, and post-ß-selection checkpoint genes, including Cd2 and Cd5. Coexpression of genes characteristic of multipotent progenitors and more mature T cells persists in the expanding population of thymocytes and in the thymic leukemias that emerge with age in these mice. These results show that Rag1-deficient NOD thymocytes have T cell defects that can collapse regulatory boundaries at two early T cell checkpoints, which may predispose them to both leukemia and autoimmunity.


Subject(s)
Cell Transformation, Neoplastic/genetics , Homeodomain Proteins/genetics , Precursor Cells, T-Lymphoid/metabolism , Actins/metabolism , Age Factors , Animals , Cell Transformation, Neoplastic/immunology , Chromosome Mapping , Chromosomes, Mammalian , Cluster Analysis , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Lymphoma/genetics , Lymphoma/immunology , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Precursor Cells, T-Lymphoid/immunology , Quantitative Trait Loci , Signal Transduction , Stem Cells/metabolism , Thymocytes/immunology , Thymocytes/metabolism , Transcription, Genetic
9.
Cell ; 149(2): 467-82, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22500808

ABSTRACT

T cell development comprises a stepwise process of commitment from a multipotent precursor. To define molecular mechanisms controlling this progression, we probed five stages spanning the commitment process using RNA-seq and ChIP-seq to track genome-wide shifts in transcription, cohorts of active transcription factor genes, histone modifications at diverse classes of cis-regulatory elements, and binding repertoire of GATA-3 and PU.1, transcription factors with complementary roles in T cell development. The results highlight potential promoter-distal cis-regulatory elements in play and reveal both activation sites and diverse mechanisms of repression that silence genes used in alternative lineages. Histone marking is dynamic and reversible, and though permissive marks anticipate, repressive marks often lag behind changes in transcription. In vivo binding of PU.1 and GATA-3 relative to epigenetic marking reveals distinctive factor-specific rules for recruitment of these crucial transcription factors to different subsets of their potential sites, dependent on dose and developmental context.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , T-Lymphocytes/cytology , Animals , GATA3 Transcription Factor/metabolism , Gene Expression Regulation , Genome-Wide Association Study , Histone Code , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Receptors, Notch/metabolism , Regulatory Elements, Transcriptional , Signal Transduction , T-Lymphocytes/metabolism , Trans-Activators/metabolism , Transcription, Genetic
10.
Curr Top Microbiol Immunol ; 356: 117-43, 2012.
Article in English | MEDLINE | ID: mdl-21833836

ABSTRACT

T-cell development endows cells with a flexible range of effector differentiation options, superimposed on a stable core of lineage-specific gene expression that is maintained while access to alternative hematopoietic lineages is permanently renounced. This combination of features could be explained by environmentally responsive transcription factor mobilization overlaying an epigenetically stabilized base gene expression state. For example, "poising" of promoters could offer preferential access to T-cell genes, while repressive histone modifications and DNA methylation of non-T regulatory genes could be responsible for keeping non-T developmental options closed. Here, we critically review the evidence for the actual deployment of epigenetic marking to support the stable aspects of T-cell identity. Much of epigenetic marking is dynamically maintained or subject to rapid modification by local action of transcription factors. Repressive histone marks are used in gene-specific ways that do not fit a simple, developmental lineage-exclusion hierarchy. We argue that epigenetic analysis may achieve its greatest impact for illuminating regulatory biology when it is used to locate cis-regulatory elements by catching them in the act of mediating regulatory change.


Subject(s)
Epigenesis, Genetic , T-Lymphocytes/cytology , Animals , Cell Differentiation , Cell Lineage , Histones/genetics , Histones/metabolism , Humans , T-Lymphocytes/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL