Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
Front Cell Dev Biol ; 10: 920683, 2022.
Article in English | MEDLINE | ID: mdl-36060812

ABSTRACT

Osteoclasts are bone-resorbing cells that undergo extensive changes in morphology throughout their differentiation. Altered osteoclast differentiation and activity lead to changes in pathological bone resorption. The mammalian target of rapamycin (mTOR) is a kinase, and aberrant mTOR complex 1 (mTORC1) signaling is associated with altered bone homeostasis. The activation of mTORC1 is biphasically regulated during osteoclastogenesis; however, the mechanism behind mTORC1-mediated regulation of osteoclastogenesis and bone resorption is incompletely understood. Here, we found that MYC coordinates the dynamic regulation of mTORC1 activation during osteoclastogenesis. MYC-deficiency blocked the early activation of mTORC1 and also reversed the decreased activity of mTORC1 at the late stage of osteoclastogenesis. The suppression of mTORC1 activity by rapamycin in mature osteoclasts enhances bone resorption activity despite the indispensable role of high mTORC1 activation in osteoclast formation in both mouse and human cells. Mechanistically, MYC induces Growth arrest and DNA damage-inducible protein (GADD34) expression and suppresses mTORC1 activity at the late phase of osteoclastogenesis. Taken together, our findings identify a MYC-GADD34 axis as an upstream regulator of dynamic mTORC1 activation in osteoclastogenesis and highlight the interplay between MYC and mTORC1 pathways in determining osteoclast activity.

3.
PLoS One ; 12(7): e0179762, 2017.
Article in English | MEDLINE | ID: mdl-28708839

ABSTRACT

During rheumatoid arthritis (RA), Tumor Necrosis Factor (TNF) activates fibroblast-like synoviocytes (FLS) inducing in a temporal order a constellation of genes, which perpetuate synovial inflammation. Although the molecular mechanisms regulating TNF-induced transcription are well characterized, little is known about the impact of mRNA stability on gene expression and the impact of TNF on decay rates of mRNA transcripts in FLS. To address these issues we performed RNA sequencing and genome-wide analysis of the mRNA stabilome in RA FLS. We found that TNF induces a biphasic gene expression program: initially, the inducible transcriptome consists primarily of unstable transcripts but progressively switches and becomes dominated by very stable transcripts. This temporal switch is due to: a) TNF-induced prolonged stabilization of previously unstable transcripts that enables progressive transcript accumulation over days and b) sustained expression and late induction of very stable transcripts. TNF-induced mRNA stabilization in RA FLS occurs during the late phase of TNF response, is MAPK-dependent, and involves several genes with pathogenic potential such as IL6, CXCL1, CXCL3, CXCL8/IL8, CCL2, and PTGS2. These results provide the first insights into genome-wide regulation of mRNA stability in RA FLS and highlight the potential contribution of dynamic regulation of the mRNA stabilome by TNF to chronic synovitis.


Subject(s)
RNA Stability/drug effects , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cells, Cultured , Chemokines/genetics , Chemokines/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/genetics , Cytokines/metabolism , Fibroblasts/cytology , Gene Expression Regulation/drug effects , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , RNA/chemistry , RNA/isolation & purification , RNA/metabolism , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Synoviocytes/cytology , Synoviocytes/drug effects , Synoviocytes/metabolism
4.
Immunity ; 47(1): 66-79.e5, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28723554

ABSTRACT

Hypoxia augments inflammatory responses and osteoclastogenesis by incompletely understood mechanisms. We identified COMMD1 as a cell-intrinsic negative regulator of osteoclastogenesis that is suppressed by hypoxia. In human macrophages, COMMD1 restrained induction of NF-κB signaling and a transcription factor E2F1-dependent metabolic pathway by the cytokine RANKL. Downregulation of COMMD1 protein expression by hypoxia augmented RANKL-induced expression of inflammatory and E2F1 target genes and downstream osteoclastogenesis. E2F1 targets included glycolysis and metabolic genes including CKB that enabled cells to meet metabolic demands in challenging environments, as well as inflammatory cytokine-driven target genes. Expression quantitative trait locus analysis linked increased COMMD1 expression with decreased bone erosion in rheumatoid arthritis. Myeloid deletion of Commd1 resulted in increased osteoclastogenesis in arthritis and inflammatory osteolysis models. These results identify COMMD1 and an E2F-metabolic pathway as key regulators of osteoclastogenic responses under pathological inflammatory conditions and provide a mechanism by which hypoxia augments inflammation and bone destruction.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Arthritis, Rheumatoid/immunology , Macrophages/immunology , Osteogenesis/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Cells, Cultured , Disease Models, Animal , E2F1 Transcription Factor/metabolism , Female , Humans , Hypoxia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , NF-kappa B/metabolism , RNA, Small Interfering/genetics , Signal Transduction
5.
J Clin Invest ; 127(7): 2555-2568, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28530645

ABSTRACT

Osteoporosis is a metabolic bone disorder associated with compromised bone strength and an increased risk of fracture. Inhibition of the differentiation of bone-resorbing osteoclasts is an effective strategy for the treatment of osteoporosis. Prior work by our laboratory and others has shown that MYC promotes osteoclastogenesis in vitro, but the underlying mechanisms are not well understood. In addition, the in vivo importance of osteoclast-expressed MYC in physiological and pathological bone loss is not known. Here, we have demonstrated that deletion of Myc in osteoclasts increases bone mass and protects mice from ovariectomy-induced (OVX-induced) osteoporosis. Transcriptomic analysis revealed that MYC drives metabolic reprogramming during osteoclast differentiation and functions as a metabolic switch to an oxidative state. We identified a role for MYC action in the transcriptional induction of estrogen receptor-related receptor α (ERRα), a nuclear receptor that cooperates with the transcription factor nuclear factor of activated T cells, c1 (NFATc1) to drive osteoclastogenesis. Accordingly, pharmacological inhibition of ERRα attenuated OVX-induced bone loss in mice. Our findings highlight a MYC/ERRα pathway that contributes to physiological and pathological bone loss by integrating the MYC/ERRα axis to drive metabolic reprogramming during osteoclast differentiation.


Subject(s)
Cell Differentiation , Osteoclasts/metabolism , Osteoporosis/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Estrogen/metabolism , Signal Transduction , Animals , Female , Humans , Male , Mice , Mice, Knockout , Osteoclasts/pathology , Osteoporosis/genetics , Osteoporosis/pathology , Osteoporosis/therapy , Proto-Oncogene Proteins c-myc/genetics , Receptors, Estrogen/genetics , Transcriptome , ERRalpha Estrogen-Related Receptor
6.
Nat Commun ; 7: 11275, 2016 04 27.
Article in English | MEDLINE | ID: mdl-27117588

ABSTRACT

Spermatogonial stem and progenitor cells (SSCs) generate adult male gametes. During in vitro expansion, these unipotent murine cells spontaneously convert to multipotent adult spermatogonial-derived stem cells (MASCs). Here we investigate this conversion process through integrative transcriptomic and epigenomic analyses. We find in SSCs that promoters essential to maintenance and differentiation of embryonic stem cells (ESCs) are enriched with histone H3-lysine4 and -lysine 27 trimethylations. These bivalent modifications are maintained at most somatic promoters after conversion, bestowing MASCs an ESC-like promoter chromatin. At enhancers, the core pluripotency circuitry is activated partially in SSCs and completely in MASCs, concomitant with loss of germ cell-specific gene expression and initiation of embryonic-like programs. Furthermore, SSCs in vitro maintain the epigenomic characteristics of germ cells in vivo. Our observations suggest that SSCs encode innate plasticity through the epigenome and that both conversion of promoter chromatin states and activation of cell type-specific enhancers are prominent features of reprogramming.


Subject(s)
Cell Differentiation/genetics , Cell Plasticity/genetics , Embryonic Stem Cells/metabolism , Epigenomics/methods , Multipotent Stem Cells/metabolism , Spermatogonia/metabolism , Animals , Cells, Cultured , Gene Expression Profiling/methods , Histones/metabolism , Lysine/metabolism , Male , Methylation , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Spermatogenesis/genetics , Spermatogonia/cytology
7.
Science ; 350(6266): 1391-6, 2015 Dec 11.
Article in English | MEDLINE | ID: mdl-26541605

ABSTRACT

More than half of human colorectal cancers (CRCs) carry either KRAS or BRAF mutations and are often refractory to approved targeted therapies. We found that cultured human CRC cells harboring KRAS or BRAF mutations are selectively killed when exposed to high levels of vitamin C. This effect is due to increased uptake of the oxidized form of vitamin C, dehydroascorbate (DHA), via the GLUT1 glucose transporter. Increased DHA uptake causes oxidative stress as intracellular DHA is reduced to vitamin C, depleting glutathione. Thus, reactive oxygen species accumulate and inactivate glyceraldehyde 3-phosphate dehydrogenase (GAPDH). Inhibition of GAPDH in highly glycolytic KRAS or BRAF mutant cells leads to an energetic crisis and cell death not seen in KRAS and BRAF wild-type cells. High-dose vitamin C impairs tumor growth in Apc/Kras(G12D) mutant mice. These results provide a mechanistic rationale for exploring the therapeutic use of vitamin C for CRCs with KRAS or BRAF mutations.


Subject(s)
Ascorbic Acid/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Adenomatous Polyposis Coli Protein/genetics , Animals , Ascorbic Acid/administration & dosage , Ascorbic Acid/pharmacology , Cell Line, Tumor , Dehydroascorbic Acid/metabolism , Female , Glucose Transporter Type 1/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Glycolysis/drug effects , Humans , Mice , Mice, Mutant Strains , Mice, Nude , Proto-Oncogene Proteins p21(ras)/genetics , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
8.
Nat Immunol ; 16(8): 838-849, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26147685

ABSTRACT

Interferon-γ (IFN-γ) primes macrophages for enhanced microbial killing and inflammatory activation by Toll-like receptors (TLRs), but little is known about the regulation of cell metabolism or mRNA translation during this priming. We found that IFN-γ regulated the metabolism and mRNA translation of human macrophages by targeting the kinases mTORC1 and MNK, both of which converge on the selective regulator of translation initiation eIF4E. Physiological downregulation of mTORC1 by IFN-γ was associated with autophagy and translational suppression of repressors of inflammation such as HES1. Genome-wide ribosome profiling in TLR2-stimulated macrophages showed that IFN-γ selectively modulated the macrophage translatome to promote inflammation, further reprogram metabolic pathways and modulate protein synthesis. These results show that IFN-γ-mediated metabolic reprogramming and translational regulation are key components of classical inflammatory macrophage activation.


Subject(s)
Interferon-gamma/immunology , Macrophage Activation/immunology , Macrophages/immunology , Protein Biosynthesis/immunology , RNA, Messenger/immunology , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cells, Cultured , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/immunology , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Profiling , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Homeodomain Proteins/metabolism , Humans , Interferon-gamma/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophage Activation/drug effects , Macrophage Activation/genetics , Macrophages/drug effects , Macrophages/metabolism , Mechanistic Target of Rapamycin Complex 1 , MicroRNAs/genetics , Microscopy, Fluorescence , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Multiprotein Complexes/metabolism , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Transcription Factor HES-1
9.
Arthritis Res Ther ; 17: 167, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26126608

ABSTRACT

Studying the factors that control gene expression is of substantial importance for rheumatic diseases with poorly understood etiopathogenesis. In the past, gene expression microarrays have been used to measure transcript abundance on a genome-wide scale in a particular cell, tissue or organ. Microarray analysis has led to gene signatures that differentiate rheumatic diseases, and stages of a disease, as well as response to treatments. Nowadays, however, with the advent of next-generation sequencing methods, massive parallel sequencing of RNA tends to be the technology of choice for gene expression profiling, due to several advantages over microarrays, as well as for the detection of non-coding transcripts and alternative splicing events. In this review, we describe how RNA sequencing enables unbiased interrogation of the abundance and complexity of the transcriptome, and present a typical experimental workflow and bioinformatics tools that are often used for RNA sequencing analysis. We also discuss different uses of this next-generation sequencing technology to evaluate rheumatic disease patients and investigate the pathogenesis of rheumatic diseases such as rheumatoid arthritis, systemic lupus erythematosus, juvenile idiopathic arthritis and Sjögren's syndrome.


Subject(s)
Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , Rheumatic Diseases/genetics , Sequence Analysis, RNA/methods , Humans
10.
PLoS Genet ; 11(4): e1005174, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25894978

ABSTRACT

While the importance of gene enhancers in transcriptional regulation is well established, the mechanisms and the protein factors that determine enhancers activity have only recently begun to be unravelled. Recent studies have shown that progesterone receptor (PR) binds regions that display typical features of gene enhancers. Here, we show by ChIP-seq experiments that the chromatin remodeler CHD8 mostly binds promoters under proliferation conditions. However, upon progestin stimulation, CHD8 re-localizes to PR enhancers also enriched in p300 and H3K4me1. Consistently, CHD8 depletion severely impairs progestin-dependent gene regulation. CHD8 binding is PR-dependent but independent of the pioneering factor FOXA1. The SWI/SNF chromatin-remodelling complex is required for PR-dependent gene activation. Interestingly, we show that CHD8 interacts with the SWI/SNF complex and that depletion of BRG1 and BRM, the ATPases of SWI/SNF complex, impairs CHD8 recruitment. We also show that CHD8 is not required for H3K27 acetylation, but contributes to increase accessibility of the enhancer to DNaseI. Furthermore, CHD8 was required for RNAPII recruiting to the enhancers and for transcription of enhancer-derived RNAs (eRNAs). Taken together our data demonstrate that CHD8 is involved in late stages of PR enhancers activation.


Subject(s)
DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Receptors, Progesterone/genetics , Transcription Factors/genetics , Transcription, Genetic , Acetylation , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Promoter Regions, Genetic , Protein Binding , RNA Polymerase II/genetics , Receptors, Progesterone/metabolism , Transcription Factors/metabolism
11.
PLoS Genet ; 10(9): e1004566, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25188243

ABSTRACT

Overexpression of the histone methyltransferase MMSET in t(4;14)+ multiple myeloma patients is believed to be the driving factor in the pathogenesis of this subtype of myeloma. MMSET catalyzes dimethylation of lysine 36 on histone H3 (H3K36me2), and its overexpression causes a global increase in H3K36me2, redistributing this mark in a broad, elevated level across the genome. Here, we demonstrate that an increased level of MMSET also induces a global reduction of lysine 27 trimethylation on histone H3 (H3K27me3). Despite the net decrease in H3K27 methylation, specific genomic loci exhibit enhanced recruitment of the EZH2 histone methyltransferase and become hypermethylated on this residue. These effects likely contribute to the myeloma phenotype since MMSET-overexpressing cells displayed increased sensitivity to EZH2 inhibition. Furthermore, we demonstrate that such MMSET-mediated epigenetic changes require a number of functional domains within the protein, including PHD domains that mediate MMSET recruitment to chromatin. In vivo, targeting of MMSET by an inducible shRNA reversed histone methylation changes and led to regression of established tumors in athymic mice. Together, our work elucidates previously unrecognized interplay between MMSET and EZH2 in myeloma oncogenesis and identifies domains to be considered when designing inhibitors of MMSET function.


Subject(s)
DNA Methylation/genetics , Epigenesis, Genetic/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Multiple Myeloma/genetics , Polycomb Repressive Complex 2/metabolism , Protein Binding/genetics , Animals , Cell Line , Cell Transformation, Neoplastic/genetics , Chromatin/genetics , Female , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Humans , Lysine/genetics , Mice , Mice, Inbred C57BL , Multiple Myeloma/metabolism , Polycomb Repressive Complex 2/genetics , RNA, Small Interfering/genetics
12.
J Immunol ; 193(5): 2373-83, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25057003

ABSTRACT

Mesenchymal stromal cells have emerged as powerful modulators of the immune system. In this study, we explored how the human macrophage response to TNF is regulated by human synovial fibroblasts, the representative stromal cell type in the synovial lining of joints that become activated during inflammatory arthritis. We found that synovial fibroblasts strongly suppressed TNF-mediated induction of an IFN-ß autocrine loop and downstream expression of IFN-stimulated genes (ISGs), including chemokines CXCL9 and CXCL10 that are characteristic of classical macrophage activation. TNF induced the production of soluble synovial fibroblast factors that suppressed the macrophage production of IFN-ß, and cooperated with TNF to limit the responsiveness of macrophages to IFN-ß by suppressing activation of Jak-STAT signaling. Genome-wide transcriptome analysis showed that cocultured synovial fibroblasts modulate the expression of approximately one third of TNF-regulated genes in macrophages, including genes in pathways important for macrophage survival and polarization toward an alternatively activated phenotype. Pathway analysis revealed that gene expression programs regulated by synovial fibroblasts in our coculture system were also regulated in rheumatoid arthritis synovial macrophages, suggesting that these fibroblast-mediated changes may contribute to rheumatoid arthritis pathogenesis. This work furthers our understanding of the interplay between innate immune and stromal cells during an inflammatory response, one that is particularly relevant to inflammatory arthritis. Our findings also identify modulation of macrophage phenotype as a new function for synovial fibroblasts that may prove to be a contributing factor in arthritis pathogenesis.


Subject(s)
Arthritis, Rheumatoid/immunology , Fibroblasts/immunology , Macrophages/immunology , Signal Transduction/immunology , Synovial Membrane/immunology , Tumor Necrosis Factor-alpha/immunology , Arthritis, Rheumatoid/pathology , Autocrine Communication/immunology , Cells, Cultured , Chemokine CXCL10/immunology , Chemokine CXCL9/immunology , Coculture Techniques , Female , Fibroblasts/pathology , Genome-Wide Association Study , Humans , Interferon-beta/immunology , Janus Kinases/immunology , Macrophages/pathology , Male , STAT Transcription Factors/immunology , Synovial Membrane/pathology , Transcription, Genetic/immunology
13.
Immunity ; 39(3): 454-69, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-24012417

ABSTRACT

Synergistic activation of inflammatory cytokine genes by interferon-γ (IFN-γ) and Toll-like receptor (TLR) signaling is important for innate immunity and inflammatory disease pathogenesis. Enhancement of TLR signaling, a previously proposed mechanism, is insufficient to explain strong synergistic activation of cytokine production in human macrophages. Rather, we found that IFN-γ induced sustained occupancy of transcription factors STAT1, IRF-1, and associated histone acetylation at promoters and enhancers at the TNF, IL6, and IL12B loci. This priming of chromatin did not activate transcription but greatly increased and prolonged recruitment of TLR4-induced transcription factors and RNA polymerase II to gene promoters and enhancers. Priming sensitized cytokine transcription to suppression by Jak inhibitors. Genome-wide analysis revealed pervasive priming of regulatory elements by IFN-γ and linked coordinate priming of promoters and enhancers with synergistic induction of transcription. Our results provide a synergy mechanism whereby IFN-γ creates a primed chromatin environment to augment TLR-induced gene transcription.


Subject(s)
Chromatin Assembly and Disassembly , Cytokines/metabolism , Interferon-gamma/metabolism , Toll-Like Receptors/metabolism , Acetylation , Cells, Cultured , Enzyme Activation , Histones/metabolism , Humans , Interferon Regulatory Factor-1/metabolism , Interleukin-12 Subunit p40/metabolism , Interleukin-6/metabolism , Janus Kinases/antagonists & inhibitors , Macrophages/metabolism , Promoter Regions, Genetic , Protein Binding , RNA Polymerase II/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction , Transcription, Genetic , Tumor Necrosis Factors/metabolism
14.
Cell Rep ; 4(3): 578-88, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23911289

ABSTRACT

The BCL6 transcriptional repressor is required for the development of germinal center (GC) B cells and diffuse large B cell lymphomas (DLBCLs). Although BCL6 can recruit multiple corepressors, its transcriptional repression mechanism of action in normal and malignant B cells is unknown. We find that in B cells, BCL6 mostly functions through two independent mechanisms that are collectively essential to GC formation and DLBCL, both mediated through its N-terminal BTB domain. These are (1) the formation of a unique ternary BCOR-SMRT complex at promoters, with each corepressor binding to symmetrical sites on BCL6 homodimers linked to specific epigenetic chromatin features, and (2) the "toggling" of active enhancers to a poised but not erased conformation through SMRT-dependent H3K27 deacetylation, which is mediated by HDAC3 and opposed by p300 histone acetyltransferase. Dynamic toggling of enhancers provides a basis for B cells to undergo rapid transcriptional and phenotypic changes in response to signaling or environmental cues.


Subject(s)
B-Lymphocytes/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Animals , B-Lymphocytes/metabolism , Cell Line, Tumor , Heterografts , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Mice , Models, Molecular , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-6 , Signal Transduction
15.
Genome Res ; 23(8): 1295-306, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23554462

ABSTRACT

Genome-wide binding assays can determine where individual transcription factors bind in the genome. However, these factors rarely bind chromatin alone, but instead frequently bind to cis-regulatory elements (CREs) together with other factors thus forming protein complexes. Currently there are no integrative analytical approaches that can predict which complexes are formed on chromatin. Here, we describe a computational methodology to systematically capture protein complexes and infer their impact on gene expression. We applied our method to three human cell types, identified thousands of CREs, inferred known and undescribed complexes recruited to these CREs, and determined the role of the complexes as activators or repressors. Importantly, we found that the predicted complexes have a higher number of physical interactions between their members than expected by chance. Our work provides a mechanism for developing hypotheses about gene regulation via binding partners, and deciphering the interplay between combinatorial binding and gene expression.


Subject(s)
Chromatin/metabolism , Transcription Factors/metabolism , Chromatin Immunoprecipitation , Cluster Analysis , Computational Biology , Gene Expression Regulation , Genome, Human , Humans , Models, Genetic , Protein Binding , Regulatory Sequences, Nucleic Acid , Sequence Analysis, DNA
16.
Neoplasia ; 15(4): 373-83, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23555183

ABSTRACT

Although prostate cancer (PCa) is the second leading cause of cancer death among men worldwide, not all men diagnosed with PCa will die from the disease. A critical challenge, therefore, is to distinguish indolent PCa from more advanced forms to guide appropriate treatment decisions. We used Enhanced Reduced Representation Bisulfite Sequencing, a genome-wide high-coverage single-base resolution DNA methylation method to profile seven localized PCa samples, seven matched benign prostate tissues, and six aggressive castration-resistant prostate cancer (CRPC) samples. We integrated these data with RNA-seq and whole-genome DNA-seq data to comprehensively characterize the PCa methylome, detect changes associated with disease progression, and identify novel candidate prognostic biomarkers. Our analyses revealed the correlation of cytosine guanine dinucleotide island (CGI)-specific hypermethylation with disease severity and association of certain breakpoints (deletion, tandem duplications, and interchromosomal translocations) with DNA methylation. Furthermore, integrative analysis of methylation and single-nucleotide polymorphisms (SNPs) uncovered widespread allele-specific methylation (ASM) for the first time in PCa. We found that most DNA methylation changes occurred in the context of ASM, suggesting that variations in tumor epigenetic landscape of individuals are partly mediated by genetic differences, which may affect PCa disease progression. We further selected a panel of 13 CGIs demonstrating increased DNA methylation with disease progression and validated this panel in an independent cohort of 20 benign prostate tissues, 16 PCa, and 8 aggressive CRPCs. These results warrant clinical evaluation in larger cohorts to help distinguish indolent PCa from advanced disease.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Epigenesis, Genetic , Prostatic Neoplasms/genetics , Adenocarcinoma/pathology , Aged , Chromosome Breakpoints , CpG Islands , DNA Methylation , Humans , Male , Middle Aged , Molecular Diagnostic Techniques , Mutation , Neoplasm Grading , Polymorphism, Single Nucleotide , Prognosis , Prostatic Neoplasms/pathology , Sequence Analysis, DNA
17.
Nature ; 495(7439): 98-102, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23389444

ABSTRACT

Adult stem cells reside in specialized niches where they receive environmental cues to maintain tissue homeostasis. In mammals, the stem cell niche within hair follicles is home to epithelial hair follicle stem cells and melanocyte stem cells, which sustain cyclical bouts of hair regeneration and pigmentation. To generate pigmented hairs, synchrony is achieved such that upon initiation of a new hair cycle, stem cells of each type activate lineage commitment. Dissecting the inter-stem-cell crosstalk governing this intricate coordination has been difficult, because mutations affecting one lineage often affect the other. Here we identify transcription factor NFIB as an unanticipated coordinator of stem cell behaviour. Hair follicle stem-cell-specific conditional targeting of Nfib in mice uncouples stem cell synchrony. Remarkably, this happens not by perturbing hair cycle and follicle architecture, but rather by promoting melanocyte stem cell proliferation and differentiation. The early production of melanin is restricted to melanocyte stem cells at the niche base. Melanocyte stem cells more distant from the dermal papilla are unscathed, thereby preventing hair greying typical of melanocyte stem cell differentiation mutants. Furthermore, we pinpoint KIT-ligand as a dermal papilla signal promoting melanocyte stem cell differentiation. Additionally, through chromatin-immunoprecipitation with high-throughput-sequencing and transcriptional profiling, we identify endothelin 2 (Edn2) as an NFIB target aberrantly activated in NFIB-deficient hair follicle stem cells. Ectopically induced Edn2 recapitulates NFIB-deficient phenotypes in wild-type mice. Conversely, endothelin receptor antagonists and/or KIT blocking antibodies prevent precocious melanocyte stem cell differentiation in the NFIB-deficient niche. Our findings reveal how melanocyte and hair follicle stem cell behaviours maintain reliance upon cooperative factors within the niche, and how this can be uncoupled in injury, stress and disease states.


Subject(s)
Hair Follicle/cytology , Melanocytes/cytology , NFI Transcription Factors/metabolism , Stem Cell Niche , Stem Cells/cytology , Stem Cells/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Chromatin Immunoprecipitation , Endothelin-2/genetics , Endothelin-2/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Hair/cytology , Hair/growth & development , Hair Color , Hair Follicle/metabolism , Melanocytes/metabolism , Mice , NFI Transcription Factors/deficiency , NFI Transcription Factors/genetics , Sequence Analysis , Stem Cell Factor/metabolism
18.
J Proteome Res ; 12(4): 1591-603, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23418717

ABSTRACT

A naturally occurring benzofuran derivative, Ebenfuran III (Eb III), was investigated for its antiproliferative effects using the DU-145 prostate cell line. Eb III was isolated from Onobrychis ebenoides of the Leguminosae family, a plant endemic in Central and Southern Greece. We have previously reported that Eb III exerts significant cytotoxic effects on certain cancer cell lines. This effect is thought to occur via the isoprenyl moiety at the C-5 position of the molecule. The study aim was to gain a deeper understanding of the pharmacological effect of Eb III on DU-145 cell death at the translational level using a relative quantitative and temporal proteomics approach. Proteins extracted from the cell pellets were subjected to solution phase trypsin proteolysis followed by iTRAQ-labeling. The labeled tryptic peptide extracts were then fractionated using strong cation exchange chromatography and the fractions were analyzed by nanoflow reverse phase ultraperformance liquid chromatography-nanoelectrospray ionization-tandem mass spectrometry analysis using a hybrid QqTOF platform. Using this approach, we compared the expression levels of 1360 proteins analyzed at ≤ 1% global protein false discovery rate (FDR), commonly present in untreated (control, vehicle only) and Eb III-treated cells at the different exposure time points. Through the iterative use of Ingenuity Pathway Analysis with hierarchical clustering of protein expression patterns, followed by bibliographic research, the temporal regulation of the Calpain-1, ERK2, PAR-4, RAB-7, and Bap31 proteins were identified as potential nodes of multipathway convergence to Eb III induced DU-145 cell death. These proteins were further verified with Western blot analysis. This gel-free, quantitative 2DLC-MS/MS proteomics method effectively captured novel modulated proteins in the DU-145 cell line as a response to Eb III treatment. This approach also provided greater insight to the multifocal and combinatorial signaling pathways implicated in Eb III-induced cell death.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Benzofurans/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Proteins/metabolism , Resorcinols/pharmacology , Apoptosis Regulatory Proteins/metabolism , Calpain/metabolism , Cell Death/drug effects , Cell Line, Tumor , Chromatography, Reverse-Phase/methods , Cluster Analysis , Humans , Male , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Prostatic Neoplasms/pathology , Proteins/analysis , Tandem Mass Spectrometry/methods , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
19.
BMC Bioinformatics ; 12: 277, 2011 Jul 07.
Article in English | MEDLINE | ID: mdl-21736739

ABSTRACT

BACKGROUND: Chromatin immunoprecipitation followed by next generation sequencing (ChIP-seq), enables unbiased and genome-wide mapping of protein-DNA interactions and epigenetic marks. The first step in ChIP-seq data analysis involves the identification of peaks (i.e., genomic locations with high density of mapped sequence reads). The next step consists of interpreting the biological meaning of the peaks through their association with known genes, pathways, regulatory elements, and integration with other experiments. Although several programs have been published for the analysis of ChIP-seq data, they often focus on the peak detection step and are usually not well suited for thorough, integrative analysis of the detected peaks. RESULTS: To address the peak interpretation challenge, we have developed ChIPseeqer, an integrative, comprehensive, fast and user-friendly computational framework for in-depth analysis of ChIP-seq datasets. The novelty of our approach is the capability to combine several computational tools in order to create easily customized workflows that can be adapted to the user's needs and objectives. In this paper, we describe the main components of the ChIPseeqer framework, and also demonstrate the utility and diversity of the analyses offered, by analyzing a published ChIP-seq dataset. CONCLUSIONS: ChIPseeqer facilitates ChIP-seq data analysis by offering a flexible and powerful set of computational tools that can be used in combination with one another. The framework is freely available as a user-friendly GUI application, but all programs are also executable from the command line, thus providing flexibility and automatability for advanced users.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromosome Mapping , High-Throughput Nucleotide Sequencing/methods , Software , Enhancer Elements, Genetic , Histone Code , Humans , Jurkat Cells , Proto-Oncogene Protein c-ets-1/metabolism , Workflow
20.
BMC Bioinformatics ; 12: 308, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21798033

ABSTRACT

BACKGROUND: The steps of a high-throughput proteomics experiment include the separation, differential expression and mass spectrometry-based identification of proteins. However, the last and more challenging step is inferring the biological role of the identified proteins through their association with interaction networks, biological pathways, analysis of the effect of post-translational modifications, and other protein-related information. RESULTS: In this paper, we present an integrative visualization methodology that allows combining experimentally produced proteomic features with protein meta-features, typically coming from meta-analysis tools and databases, in synthetic Proteomic Feature Maps. Using three proteomics analysis scenarios, we show that the proposed visualization approach is effective in filtering, navigating and interacting with the proteomics data in order to address visually challenging biological questions. The novelty of our approach lies in the ease of integration of any user-defined proteomic features in easy-to-comprehend visual representations that resemble the familiar 2D-gel images, and can be adapted to the user's needs. The main capabilities of the developed VIP software, which implements the presented visualization methodology, are also highlighted and discussed. CONCLUSIONS: By using this visualization and the associated VIP software, researchers can explore a complex heterogeneous proteomics dataset from different perspectives in order to address visually important biological queries and formulate new hypotheses for further investigation. VIP is freely available at http://pelopas.uop.gr/~egian/VIP/index.html.


Subject(s)
Proteomics/methods , Software , Mass Spectrometry/methods , Phosphorylation , Protein Processing, Post-Translational , Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...