Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Nat Immunol ; 20(6): 687-700, 2019 06.
Article in English | MEDLINE | ID: mdl-31061528

ABSTRACT

Most tissue-resident macrophage populations develop during embryogenesis, self-renew in the steady state and expand during type 2 immunity. Whether shared mechanisms regulate the proliferation of macrophages in homeostasis and disease is unclear. Here we found that the transcription factor Bhlhe40 was required in a cell-intrinsic manner for the self-renewal and maintenance of large peritoneal macrophages (LPMs), but not that of other tissue-resident macrophages. Bhlhe40 was necessary for the proliferation, but not the polarization, of LPMs in response to the cytokine IL-4. During infection with the helminth Heligmosomoides polygyrus bakeri, Bhlhe40 was required for cell cycling of LPMs. Bhlhe40 repressed the expression of genes encoding the transcription factors c-Maf and Mafb and directly promoted expression of transcripts encoding cell cycle-related proteins to enable the proliferation of LPMs. In LPMs, Bhlhe40 bound to genomic sites co-bound by the macrophage lineage-determining factor PU.1 and to unique sites, including Maf and loci encoding cell-cycle-related proteins. Our findings demonstrate a tissue-specific control mechanism that regulates the proliferation of resident macrophages in homeostasis and type 2 immunity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Homeodomain Proteins/genetics , Homeostasis/genetics , Homeostasis/immunology , Immunity/genetics , Macrophages/immunology , Macrophages/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers , Cell Cycle/genetics , Cell Cycle/immunology , Cell Proliferation , Gene Expression Profiling , Gene Expression Regulation , Gene Knockout Techniques , Helicobacter Infections/genetics , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Homeodomain Proteins/metabolism , Immunophenotyping , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Transgenic , Monocytes/immunology , Monocytes/metabolism , Organ Specificity/genetics , Organ Specificity/immunology , Transcriptome
2.
Mol Cell ; 2023 Aug 25.
Article in English | MEDLINE | ID: mdl-37657444

ABSTRACT

N6-methyladenosine (m6A) RNA modification plays important roles in the governance of gene expression and is temporally regulated in different cell states. In contrast to global m6A profiling in bulk sequencing, single-cell technologies for analyzing m6A heterogeneity are not extensively established. Here, we developed single-nucleus m6A-CUT&Tag (sn-m6A-CT) for simultaneous profiling of m6A methylomes and transcriptomes within a single nucleus using mouse embryonic stem cells (mESCs). m6A-CT is capable of enriching m6A-marked RNA molecules in situ, without isolating RNAs from cells. We adapted m6A-CT to the droplet-based single-cell omics platform and demonstrated high-throughput performance in analyzing nuclei isolated from thousands of cells from various cell types. We show that sn-m6A-CT profiling is sufficient to determine cell identity and allows the generation of cell-type-specific m6A methylome landscapes from heterogeneous populations. These indicate that sn-m6A-CT provides additional dimensions to multimodal datasets and insights into epitranscriptomic landscape in defining cell fate identity and states.

3.
Nat Immunol ; 18(4): 442-455, 2017 04.
Article in English | MEDLINE | ID: mdl-28250425

ABSTRACT

Innate-like B-1a cells provide a first line of defense against pathogens, yet little is known about their transcriptional control. Here we identified an essential role for the transcription factor Bhlhe41, with a lesser contribution by Bhlhe40, in controlling B-1a cell differentiation. Bhlhe41-/-Bhlhe40-/- B-1a cells were present at much lower abundance than were their wild-type counterparts. Mutant B-1a cells exhibited an abnormal cell-surface phenotype and altered B cell receptor (BCR) repertoire exemplified by loss of the phosphatidylcholine-specific VH12Vκ4 BCR. Expression of a pre-rearranged VH12Vκ4 BCR failed to 'rescue' the mutant phenotype and revealed enhanced proliferation accompanied by increased cell death. Bhlhe41 directly repressed the expression of cell-cycle regulators and inhibitors of BCR signaling while enabling pro-survival cytokine signaling. Thus, Bhlhe41 controls the development, BCR repertoire and self-renewal of B-1a cells.


Subject(s)
B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Self Renewal , Receptors, Antigen, B-Cell/metabolism , Animals , B-Lymphocyte Subsets/immunology , Basic Helix-Loop-Helix Transcription Factors/genetics , Binding Sites , Biomarkers , Cell Differentiation/genetics , Cell Self Renewal/genetics , Gene Expression Regulation , Genes, Immunoglobulin , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Nucleotide Motifs , Organ Specificity/genetics , Organ Specificity/immunology , Phenotype , Position-Specific Scoring Matrices , Promoter Regions, Genetic , Protein Binding , Repressor Proteins/metabolism , Signal Transduction
4.
Immunity ; 51(3): 491-507.e7, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31533057

ABSTRACT

Tissue-resident memory CD8+ T (Trm) cells share core residency gene programs with tumor-infiltrating lymphocytes (TILs). However, the transcriptional, metabolic, and epigenetic regulation of Trm cell and TIL development and function is largely undefined. Here, we found that the transcription factor Bhlhe40 was specifically required for Trm cell and TIL development and polyfunctionality. Local PD-1 signaling inhibited TIL Bhlhe40 expression, and Bhlhe40 was critical for TIL reinvigoration following anti-PD-L1 blockade. Mechanistically, Bhlhe40 sustained Trm cell and TIL mitochondrial fitness and a functional epigenetic state. Building on these findings, we identified an epigenetic and metabolic regimen that promoted Trm cell and TIL gene signatures associated with tissue residency and polyfunctionality. This regimen empowered the anti-tumor activity of CD8+ T cells and possessed therapeutic potential even at an advanced tumor stage in mouse models. Our results provide mechanistic insights into the local regulation of Trm cell and TIL function.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , CD8-Positive T-Lymphocytes/immunology , Homeodomain Proteins/immunology , Mitochondria/immunology , Animals , Epigenesis, Genetic/immunology , Gene Expression Regulation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology
5.
EMBO Rep ; 25(2): 832-852, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38191874

ABSTRACT

BRD4, a bromodomain and extraterminal (BET) protein, is deregulated in multiple cancers and has emerged as a promising drug target. However, the function of the two main BRD4 isoforms (BRD4-L and BRD4-S) has not been analysed in parallel in most cancers. This complicates determining therapeutic efficacy of pan-BET inhibitors. In this study, using functional and transcriptomic analysis, we show that BRD-L and BRD4-S isoforms play distinct roles in fusion negative embryonal rhabdomyosarcoma. BRD4-L has an oncogenic role and inhibits myogenic differentiation, at least in part, by activating myostatin expression. Depletion of BRD4-L in vivo impairs tumour progression but does not impact metastasis. On the other hand, depletion of BRD4-S has no significant impact on tumour growth, but strikingly promotes metastasis in vivo. Interestingly, BRD4-S loss results in the enrichment of BRD4-L and RNA Polymerase II at integrin gene promoters resulting in their activation. In fusion positive alveolar rhabdomyosarcoma, BRD4-L is unrestricted in its oncogenic role, with no evident involvement of BRD4-S. Our work unveils isoform-specific functions of BRD4 in rhabdomyosarcoma.


Subject(s)
Rhabdomyosarcoma , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Protein Isoforms/genetics , Rhabdomyosarcoma/genetics , Bromodomain Containing Proteins
6.
J Immunol ; 212(11): 1766-1781, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38683120

ABSTRACT

Better understanding of the host responses to Mycobacterium tuberculosis infections is required to prevent tuberculosis and develop new therapeutic interventions. The host transcription factor BHLHE40 is essential for controlling M. tuberculosis infection, in part by repressing Il10 expression, where excess IL-10 contributes to the early susceptibility of Bhlhe40-/- mice to M. tuberculosis infection. Deletion of Bhlhe40 in lung macrophages and dendritic cells is sufficient to increase the susceptibility of mice to M. tuberculosis infection, but how BHLHE40 impacts macrophage and dendritic cell responses to M. tuberculosis is unknown. In this study, we report that BHLHE40 is required in myeloid cells exposed to GM-CSF, an abundant cytokine in the lung, to promote the expression of genes associated with a proinflammatory state and better control of M. tuberculosis infection. Loss of Bhlhe40 expression in murine bone marrow-derived myeloid cells cultured in the presence of GM-CSF results in lower levels of proinflammatory associated signaling molecules IL-1ß, IL-6, IL-12, TNF-α, inducible NO synthase, IL-2, KC, and RANTES, as well as higher levels of the anti-inflammatory-associated molecules MCP-1 and IL-10 following exposure to heat-killed M. tuberculosis. Deletion of Il10 in Bhlhe40-/- myeloid cells restored some, but not all, proinflammatory signals, demonstrating that BHLHE40 promotes proinflammatory responses via both IL-10-dependent and -independent mechanisms. In addition, we show that macrophages and neutrophils within the lungs of M. tuberculosis-infected Bhlhe40-/- mice exhibit defects in inducible NO synthase production compared with infected wild-type mice, supporting that BHLHE40 promotes proinflammatory responses in innate immune cells, which may contribute to the essential role for BHLHE40 during M. tuberculosis infection in vivo.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Interleukin-10 , Mice, Knockout , Myeloid Cells , Animals , Mice , Interleukin-10/immunology , Interleukin-10/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Myeloid Cells/immunology , Mycobacterium tuberculosis/immunology , Macrophages/immunology , Homeodomain Proteins/genetics , Mice, Inbred C57BL , Granulocyte-Macrophage Colony-Stimulating Factor , Dendritic Cells/immunology , Lung/immunology , Tuberculosis/immunology , Cell Polarity , Cells, Cultured
7.
Nucleic Acids Res ; 52(12): 7063-7080, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38808662

ABSTRACT

Cohesin plays a crucial role in the organization of topologically-associated domains (TADs), which influence gene expression and DNA replication timing. Whether epigenetic regulators may affect TADs via cohesin to mediate DNA replication remains elusive. Here, we discover that the histone demethylase PHF2 associates with RAD21, a core subunit of cohesin, to regulate DNA replication in mouse neural stem cells (NSC). PHF2 loss impairs DNA replication due to the activation of dormant replication origins in NSC. Notably, the PHF2/RAD21 co-bound genomic regions are characterized by CTCF enrichment and epigenomic features that resemble efficient, active replication origins, and can act as boundaries to separate adjacent domains. Accordingly, PHF2 loss weakens TADs and chromatin loops at the co-bound loci due to reduced RAD21 occupancy. The observed topological and DNA replication defects in PHF2 KO NSC support a cohesin-dependent mechanism. Furthermore, we demonstrate that the PHF2/RAD21 complex exerts little effect on gene regulation, and that PHF2's histone-demethylase activity is dispensable for normal DNA replication and proliferation of NSC. We propose that PHF2 may serve as a topological accessory to cohesin for cohesin localization to TADs and chromatin loops, where cohesin represses dormant replication origins directly or indirectly, to sustain DNA replication in NSC.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Cohesins , DNA Replication , DNA-Binding Proteins , Neural Stem Cells , Animals , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Mice , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Chromatin/metabolism , Replication Origin , Histone Demethylases/metabolism , Histone Demethylases/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Genome/genetics , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Mice, Knockout
9.
J Pathol ; 259(3): 342-356, 2023 03.
Article in English | MEDLINE | ID: mdl-36573560

ABSTRACT

The relatively quiet mutational landscape of rhabdomyosarcoma (RMS) suggests that epigenetic deregulation could be central to oncogenesis and tumour aggressiveness. Histone variants have long been recognised as important epigenetic regulators of gene expression. However, the role of histone variants in RMS has not been studied hitherto. In this study, we show that histone variant H3.3 is overexpressed in alveolar RMS (ARMS), an aggressive subtype of RMS. Functionally, knockdown of H3F3A, which encodes for H3.3, significantly impairs the ability of ARMS cells to undertake migration and invasion and reduces Rho activation. In addition, a striking reduction in metastatic tumour burden and improved survival is apparent in vivo. Using RNA-sequencing and ChIP-sequencing analyses, we identified melanoma cell adhesion molecule (MCAM/CD146) as a direct downstream target of H3.3. Loss of H3.3 resulted in a reduction in the presence of active marks and an increase in the occupancy of H1 at the MCAM promoter. Cell migration and invasion were rescued in H3F3A-depleted cells through MCAM overexpression. Moreover, we identified G9a, a lysine methyltransferase encoded by EHMT2, as an upstream regulator of H3F3A. Therefore, this study identifies a novel H3.3 dependent axis involved in ARMS metastasis. These findings establish the potential of MCAM as a therapeutic target for high-risk ARMS patients. © 2022 The Pathological Society of Great Britain and Ireland.


Subject(s)
Histones , Rhabdomyosarcoma, Alveolar , Humans , Cell Line, Tumor , Histocompatibility Antigens/genetics , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Histones/metabolism , Promoter Regions, Genetic , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/pathology
10.
Med Teach ; : 1-7, 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38295769

ABSTRACT

PURPOSE: Generative AI will become an integral part of education in future. The potential of this technology in different disciplines should be identified to promote effective adoption. This study evaluated the performance of ChatGPT in tutorial and case-based learning questions in physiology and biochemistry for medical undergraduates. Our study mainly focused on the performance of GPT-3.5 version while a subgroup was comparatively assessed on GPT-3.5 and GPT-4 performances. MATERIALS AND METHODS: Answers were generated in GPT-3.5 for 44 modified essay questions (MEQs) in physiology and 43 MEQs in biochemistry. Each answer was graded by two independent examiners. Subsequently, a subset of 15 questions from each subject were selected to represent different score categories of the GPT-3.5 answers; responses were generated in GPT-4, and graded. RESULTS: The mean score for physiology answers was 74.7 (SD 25.96). GPT-3.5 demonstrated a statistically significant (p = .009) superior performance in lower-order questions of Bloom's taxonomy in comparison to higher-order questions. Deficiencies in the application of physiological principles in clinical context were noted as a drawback. Scores in biochemistry were relatively lower with a mean score of 59.3 (SD 26.9) for GPT-3.5. There was no statistically significant difference in the scores for higher and lower-order questions of Bloom's taxonomy. The deficiencies highlighted were lack of in-depth explanations and precision. The subset of questions where the GPT-4 and GPT-3.5 were compared demonstrated a better overall performance in GPT-4 responses in both subjects. This difference between the GPT-3.5 and GPT-4 performance was statistically significant in biochemistry but not in physiology. CONCLUSIONS: The differences in performance across the two versions, GPT-3.5 and GPT-4 across the disciplines are noteworthy. Educators and students should understand the strengths and limitations of this technology in different fields to effectively integrate this technology into teaching and learning.

11.
EMBO J ; 38(19): e101233, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31414712

ABSTRACT

Tissues in multicellular organisms are populated by resident macrophages, which perform both generic and tissue-specific functions. The latter are induced by signals from the microenvironment and rely on unique tissue-specific molecular programs requiring the combinatorial action of tissue-specific and broadly expressed transcriptional regulators. Here, we identify the transcription factors Bhlhe40 and Bhlhe41 as novel regulators of alveolar macrophages (AMs)-a population that provides the first line of immune defense and executes homeostatic functions in lung alveoli. In the absence of these factors, AMs exhibited decreased proliferation that resulted in a severe disadvantage of knockout AMs in a competitive setting. Gene expression analyses revealed a broad cell-intrinsic footprint of Bhlhe40/Bhlhe41 deficiency manifested by a downregulation of AM signature genes and induction of signature genes of other macrophage lineages. Genome-wide characterization of Bhlhe40 DNA binding suggested that these transcription factors directly repress the expression of lineage-inappropriate genes in AMs. Taken together, these results identify Bhlhe40 and Bhlhe41 as key regulators of AM self-renewal and guardians of their identity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Profiling/methods , Homeodomain Proteins/genetics , Macrophages, Alveolar/cytology , Acetylation , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Proliferation , Cell Self Renewal , Cell Survival , Down-Regulation , Gene Knockdown Techniques , Histones/metabolism , Homeodomain Proteins/metabolism , Macrophages, Alveolar/metabolism , Mice , Organ Specificity , Phenotype , Sequence Analysis, RNA
12.
J Pathol ; 256(3): 349-362, 2022 03.
Article in English | MEDLINE | ID: mdl-34897678

ABSTRACT

Alveolar rhabdomyosarcoma (ARMS) is an aggressive pediatric cancer with poor prognosis. Cancer stem cells (CSCs) are seeds for tumor relapse and metastasis. However, pathways that maintain stemness genes are not fully understood. Here, we report that the enzyme euchromatic histone lysine methyltransferase 1 (EHMT1) is expressed in primary and relapse ARMS tumors. EHMT1 suppression impaired motility and induced differentiation in ARMS cell lines and reduced tumor progression in a mouse xenograft model in vivo. RNA sequencing of EHMT1-depleted cells revealed downregulation of ALDH1A1 that is associated with CSCs. Consistent with this, inhibition of ALDH1A1 expression and activity mimicked EHMT1 depletion phenotypes and reduced tumorsphere formation. Mechanistically, we demonstrate that EHMT1 does not bind to the ALDH1A1 promoter but activates it by stabilizing C/EBPß, a known regulator of ALDH1A1 expression. Our findings identify a role for EHMT1 in maintenance of stemness by regulating ALDH1A1 expression and suggest that targeting ALDH+ cells is a promising strategy in ARMS. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Neoplastic Stem Cells/enzymology , Retinal Dehydrogenase/metabolism , Rhabdomyosarcoma, Alveolar/enzymology , Aldehyde Dehydrogenase 1 Family/genetics , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/genetics , Humans , Mice, Nude , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Phenotype , Retinal Dehydrogenase/genetics , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/pathology , Signal Transduction , Tumor Burden
13.
Subcell Biochem ; 100: 523-555, 2022.
Article in English | MEDLINE | ID: mdl-36301505

ABSTRACT

Metabolic deregulation is a key factor in cancer progression. Epigenetic changes and metabolic rewiring are intertwined in cancer. Deregulated epigenetic modifiers cause metabolic aberrations by targeting the expression of metabolic enzymes. Conversely, metabolites and cofactors affect the expression and activity of epigenetic regulators. Small molecules are promising therapeutic approaches to target the epigenetic-metabolomic crosstalk in cancer. Here, we focus on the interplay between metabolic rewiring and epigenetic landscape in the context of tumourigenesis and highlight recent advances in the use of small-molecule drug targets for therapy.


Subject(s)
Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Epigenomics , Epigenesis, Genetic , Metabolic Networks and Pathways
14.
Blood ; 135(8): 568-581, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31880771

ABSTRACT

Gastrointestinal (GI) tract involvement is the major cause of morbidity and mortality in acute graft-versus-host disease (GVHD), and pathological damage is largely attributable to inflammatory cytokine production. Recently, granulocyte-macrophage colony stimulating factor (GM-CSF) has been identified as a cytokine that mediates inflammation in the GI tract, but the transcriptional program that governs GM-CSF production and the mechanism by which GM-CSF links adaptive to innate immunity within this tissue site have not been defined. In the current study, we identified Bhlhe40 as a key transcriptional regulator that governs GM-CSF production by CD4+ T cells and mediates pathological damage in the GI tract during GVHD. In addition, we observed that GM-CSF was not regulated by either interleukin 6 (IL-6) or IL-23, which are both potent inducers of GVHD-induced colonic pathology, indicating that GM-CSF constitutes a nonredundant inflammatory pathway in the GI tract. Mechanistically, GM-CSF had no adverse effect on regulatory T-cell reconstitution, but linked adaptive to innate immunity by enhancing the activation of donor-derived dendritic cells in the colon and subsequent accumulation of these cells in the mLNs. In addition, GM-CSF promoted indirect alloantigen presentation, resulting in the accumulation of donor-derived T cells with a proinflammatory cytokine phenotype in the colon. Thus, Bhlhe40+ GM-CSF+ CD4+ T cells constitute a colitogenic T-cell population that promotes indirect alloantigen presentation and pathological damage within the GI tract, positioning GM-CSF as a key regulator of GVHD in the colon and a potential therapeutic target for amelioration of this disease.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/pathology , Graft vs Host Disease/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Homeodomain Proteins/immunology , Isoantigens/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Colon/immunology , Colon/pathology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Graft vs Host Disease/immunology , Mice, Inbred C57BL
15.
J Immunol ; 204(4): 923-932, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31900338

ABSTRACT

The transcription factor BHLHE40 is an emerging regulator of the immune system. Recent studies suggest that BHLHE40 regulates type 2 immunity, but this has not been demonstrated in vivo. We found that BHLHE40 is required in T cells for a protective TH2 cell response in mice infected with the helminth Heligmosomoides polygyrus bakeri H. polygyrus elicited changes in gene and cytokine expression by lamina propria CD4+ T cells, many of which were BHLHE40 dependent, including production of the common ß (CSF2RB) chain family cytokines GM-CSF and IL-5. In contrast to deficiency in GM-CSF or IL-5 alone, loss of both GM-CSF and IL-5 signaling impaired protection against H. polygyrus Overall, we show that BHLHE40 regulates the TH2 cell transcriptional program during helminth infection to support normal expression of Csf2, Il5, and other genes required for protection and reveal unexpected redundancy of common ß chain-dependent cytokines previously thought to possess substantially divergent functions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Homeodomain Proteins/metabolism , Interleukin-5/metabolism , Nematospiroides dubius/immunology , Strongylida Infections/immunology , Th2 Cells/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/metabolism , Disease Models, Animal , Female , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Homeodomain Proteins/genetics , Immunity, Cellular/drug effects , Immunity, Cellular/genetics , Interleukin-5/antagonists & inhibitors , Interleukin-5/genetics , Interleukin-5/immunology , Mice , Mice, Knockout , Mucous Membrane/cytology , Mucous Membrane/immunology , Mucous Membrane/metabolism , Strongylida Infections/parasitology , Th2 Cells/drug effects , Transcription, Genetic/immunology
16.
FASEB J ; 33(1): 219-230, 2019 01.
Article in English | MEDLINE | ID: mdl-29995440

ABSTRACT

Aurora kinases are critical mitotic serine/threonine kinases and are often implicated in tumorigenesis. Recent studies of the interphase functions for aurora kinase (Aurk)A have considerably expanded our understanding of its role beyond mitosis. To identify the unknown targets of AurkA, we used peptide array-based screening and found E2F4 to be a novel substrate. Phosphorylation of E2F4 by AurkA at Ser75 regulates its DNA binding and subcellular localization. Because E2F4 plays an important role in skeletal muscle differentiation, we attempted to gain insight into E2F4 phosphorylation in this context. We observed that a block in E2F4 phosphorylation retained it better within the nucleus and inhibited muscle differentiation. RNA sequencing analysis revealed a perturbation of the gene network involved in the process of muscle differentiation and mitochondrial biogenesis. Collectively, our findings establish a novel role of AurkA in the process of skeletal muscle differentiation.-Dhanasekaran, K., Bose, A., Rao, V. J., Boopathi, R., Shankar, S. R., Rao, V. K., Swaminathan, A., Vasudevan, M., Taneja, R., Kundu, T. K. Unravelling the role of aurora A beyond centrosomes and spindle assembly: implications in muscle differentiation.


Subject(s)
Aurora Kinase A/metabolism , Cell Differentiation , Centrosome/metabolism , E2F4 Transcription Factor/metabolism , Muscle, Skeletal/cytology , Myoblasts/cytology , Spindle Apparatus/metabolism , Animals , Aurora Kinase A/genetics , Cell Cycle , Cells, Cultured , E2F4 Transcription Factor/genetics , HEK293 Cells , Humans , Mice , Mitosis , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Phosphorylation
17.
FASEB J ; 33(11): 12853-12872, 2019 11.
Article in English | MEDLINE | ID: mdl-31518158

ABSTRACT

We show that both supplemental and ambient magnetic fields modulate myogenesis. A lone 10 min exposure of myoblasts to 1.5 mT amplitude supplemental pulsed magnetic fields (PEMFs) accentuated in vitro myogenesis by stimulating transient receptor potential (TRP)-C1-mediated calcium entry and downstream nuclear factor of activated T cells (NFAT)-transcriptional and P300/CBP-associated factor (PCAF)-epigenetic cascades, whereas depriving myoblasts of ambient magnetic fields slowed myogenesis, reduced TRPC1 expression, and silenced NFAT-transcriptional and PCAF-epigenetic cascades. The expression levels of peroxisome proliferator-activated receptor γ coactivator 1α, the master regulator of mitochondriogenesis, was also enhanced by brief PEMF exposure. Accordingly, mitochondriogenesis and respiratory capacity were both enhanced with PEMF exposure, paralleling TRPC1 expression and pharmacological sensitivity. Clustered regularly interspaced short palindromic repeats-Cas9 knockdown of TRPC1 precluded proliferative and mitochondrial responses to supplemental PEMFs, whereas small interfering RNA gene silencing of TRPM7 did not, coinciding with data that magnetoreception did not coincide with the expression or function of other TRP channels. The aminoglycoside antibiotics antagonized and down-regulated TRPC1 expression and, when applied concomitantly with PEMF exposure, attenuated PEMF-stimulated calcium entry, mitochondrial respiration, proliferation, differentiation, and epigenetic directive in myoblasts, elucidating why the developmental potential of magnetic fields may have previously escaped detection. Mitochondrial-based survival adaptations were also activated upon PEMF stimulation. Magnetism thus deploys an authentic myogenic directive that relies on an interplay between mitochondria and TRPC1 to reach fruition.-Yap, J. L. Y., Tai, Y. K., Fröhlich, J., Fong, C. H. H., Yin, J. N., Foo, Z. L., Ramanan, S., Beyer, C., Toh, S. J., Casarosa, M., Bharathy, N., Kala, M. P., Egli, M., Taneja, R., Lee, C. N., Franco-Obregón, A. Ambient and supplemental magnetic fields promote myogenesis via a TRPC1-mitochondrial axis: evidence of a magnetic mitohormetic mechanism.


Subject(s)
Magnetic Fields , Mitochondria, Muscle/metabolism , Muscle Development , Myoblasts, Skeletal/metabolism , Signal Transduction , TRPC Cation Channels/metabolism , Animals , Cell Line , Mice , Mitochondria, Muscle/genetics , Myoblasts, Skeletal/cytology , TRPC Cation Channels/genetics
18.
Nucleic Acids Res ; 44(17): 8129-43, 2016 09 30.
Article in English | MEDLINE | ID: mdl-27229136

ABSTRACT

Differentiation of skeletal muscle cells, like most other cell types, requires a permanent exit from the cell cycle. The epigenetic programming underlying these distinct cellular states is not fully understood. In this study, we provide evidence that the lysine methyltransferase G9a functions as a central axis to regulate proliferation and differentiation of skeletal muscle cells. Transcriptome analysis of G9a knockdown cells revealed deregulation of many cell cycle regulatory genes. We demonstrate that G9a enhances cellular proliferation by two distinct mechanisms. G9a blocks cell cycle exit via methylation-dependent transcriptional repression of the MyoD target genes p21(Cip/Waf1) and Rb1. In addition, it activates E2F1-target genes in a methyltransferase activity-independent manner. We show that G9a is present in the E2F1/PCAF complex, and enhances PCAF occupancy and histone acetylation marks at E2F1-target promoters. Interestingly, G9a preferentially associates with E2F1 at the G1/S phase and with MyoD at the G2/M phase. Our results provide evidence that G9a functions both as a co-activator and a co-repressor to enhance cellular proliferation and inhibit myogenic differentiation.


Subject(s)
Cell Cycle , Cell Differentiation , Histone-Lysine N-Methyltransferase/metabolism , Muscle Development , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , E2F1 Transcription Factor/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Lysine/metabolism , Methylation , Mice, Inbred C57BL , Muscle Development/genetics , MyoD Protein/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Promoter Regions, Genetic , p300-CBP Transcription Factors/metabolism
19.
J Pathol ; 240(3): 269-281, 2016 11.
Article in English | MEDLINE | ID: mdl-27453350

ABSTRACT

Alveolar rhabdomyosarcoma (ARMS) is an aggressive paediatric cancer of skeletal muscle with poor prognosis. A PAX3-FOXO1 fusion protein acts as a driver of malignancy in ARMS by disrupting tightly coupled but mutually exclusive pathways of proliferation and differentiation. While PAX3-FOXO1 is an attractive therapeutic target, no current treatments are designed to block its oncogenic activity. The present work shows that the histone acetyltransferase P/CAF (KAT2B) is overexpressed in primary tumours from ARMS patients. Interestingly, in fusion-positive ARMS cell lines, P/CAF acetylates and stabilizes PAX3-FOXO1 rather than MyoD, a master regulator of muscle differentiation. Silencing P/CAF, or pharmacological inhibition of its acetyltransferase activity, down-regulates PAX3-FOXO1 levels concomitant with reduced proliferation and tumour burden in xenograft mouse models. Our studies identify a P/CAF-PAX3-FOXO1 signalling node that promotes oncogenesis and may contribute to MyoD dysfunction in ARMS. This work exemplifies the therapeutic potential of targeting chromatin-modifying enzymes to inhibit fusion oncoproteins that are a frequent event in sarcomas. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/metabolism , Paired Box Transcription Factors/metabolism , Protein Processing, Post-Translational , Rhabdomyosarcoma, Alveolar/genetics , p300-CBP Transcription Factors/metabolism , Animals , Carcinogenesis/pathology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Epigenomics , Gene Silencing , Heterografts , Mice , Mice, Nude , Muscles/pathology , MyoD Protein/genetics , MyoD Protein/metabolism , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/genetics , Paired Box Transcription Factors/genetics , Rhabdomyosarcoma, Alveolar/pathology , Signal Transduction , p300-CBP Transcription Factors/genetics
20.
J Cell Sci ; 127(Pt 3): 599-608, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24357723

ABSTRACT

Sharp-1 is a basic helix-loop-helix (bHLH) transcriptional repressor that is involved in a number of cellular processes. Our previous studies have demonstrated that Sharp-1 is a negative regulator of skeletal myogenesis and it blocks differentiation of muscle precursor cells by modulating the activity of MyoD. In order to understand its role in pre- and post-natal myogenesis, we assessed skeletal muscle development and freeze-injury-induced regeneration in Sharp-1-deficient mice. We show that embryonic skeletal muscle development is not impaired in the absence of Sharp-1; however, post-natally, the regenerative capacity is compromised. Although the initial phases of injury-induced regeneration proceed normally in Sharp-1(-/-) mice, during late stages, the mutant muscle exhibits necrotic fibers, calcium deposits and fibrosis. TGF-ß expression, as well as levels of phosphorylated Smad2 and Smad3, are sustained in the mutant tissue and treatment with decorin, which blocks TGF-ß signaling, improves the histopathology of Sharp-1(-/-) injured muscles. In vitro, Sharp-1 associates with Smad3, and its overexpression inhibits TGF-ß- and Smad3-mediated expression of extracellular matrix genes in myofibroblasts. These results demonstrate that Sharp-1 regulates muscle regenerative capacity, at least in part, by modulation of TGF-ß signaling.


Subject(s)
Muscle, Skeletal/metabolism , Regeneration/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation , Embryo, Mammalian , Embryonic Development , Gene Expression Regulation, Developmental , Mice , Muscle, Skeletal/growth & development , Myofibroblasts/metabolism , Signal Transduction , Smad3 Protein , Transcription Factors/biosynthesis , Transforming Growth Factor beta/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL