Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 30(11): 2408-2431, 2023 11.
Article in English | MEDLINE | ID: mdl-37838774

ABSTRACT

Receptor clustering is the most critical step to activate extrinsic apoptosis by death receptors belonging to the TNF superfamily. Although clinically unsuccessful, using agonist antibodies, the death receptors-5 remains extensively studied from a cancer therapeutics perspective. However, despite its regulatory role and elevated function in ovarian and other solid tumors, another tumor-enriched death receptor called Fas (CD95) remained undervalued in cancer immunotherapy until recently, when its role in off-target tumor killing by CAR-T therapies was imperative. By comprehensively analyzing structure studies in the context of the binding epitope of FasL and various preclinical Fas agonist antibodies, we characterize a highly significant patch of positively charged residue epitope (PPCR) in its cysteine-rich domain 2 of Fas. PPCR engagement is indispensable for superior Fas agonist signaling and CAR-T bystander function in ovarian tumor models. A single-point mutation in FasL or Fas that interferes with the PPCR engagement inhibited apoptotic signaling in tumor cells and T cells. Furthermore, considering that clinical and immunological features of the autoimmune lymphoproliferative syndrome (ALPS) are directly attributed to homozygous mutations in FasL, we reveal differential mechanistic details of FasL/Fas clustering at the PPCR interface compared to described ALPS mutations. As Fas-mediated bystander killing remains vital to the success of CAR-T therapies in tumors, our findings highlight the therapeutic analytical design for potentially effective Fas-targeting strategies using death agonism to improve cancer immunotherapy in ovarian and other solid tumors.


Subject(s)
Ovarian Neoplasms , Receptors, Chimeric Antigen , Humans , Female , Epitopes , fas Receptor/genetics , fas Receptor/metabolism , Fas Ligand Protein , T-Lymphocytes , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Apoptosis , Antibodies/pharmacology
2.
Methods ; 217: 43-48, 2023 09.
Article in English | MEDLINE | ID: mdl-37423473

ABSTRACT

Transcriptomic profiling is a mainstay of translational cancer research and is often used to identify cancer subtypes, stratify responders vs. non-responders patients, predict survival, and identify potential targets for therapeutic intervention. Analysis of gene expression data gathered by RNA sequencing (RNA-seq) and microarray is generally the first step in identifying and characterizing cancer-associated molecular determinants. The methodological advancements and reduced costs associated with transcriptomic profiling have increased the number of publicly available gene expression profiles for cancer subtypes. Data integration from multiple datasets is routinely done to increase the number of samples, improve statistical power, and provide better insight into the heterogeneity of the biological determinant. However, utilizing raw data from multiple platforms, species, and sources introduces systematic variations due to noise, batch effects, and biases. As such, the integrated data is mathematically adjusted through normalization, which allows direct comparison of expression measures among studies while minimizing technical and systemic variations. This study applied meta-analysis to multiple independent Affymetrix microarray and Illumina RNA-seq datasets available through the Gene Expression Omnibus (GEO) and The Cancer Gene Atlas (TCGA). We have previously identified a tripartite motif containing 37 (TRIM37), a breast cancer oncogene, that drives tumorigenesis and metastasis in triple-negative breast cancer. In this article, we adapted and assessed the validity of Stouffer's z-score normalization method to interrogate TRIM37 expression across different cancer types using multiple large-scale datasets.


Subject(s)
Breast Neoplasms , Gene Expression Profiling , Humans , Female , Gene Expression Profiling/methods , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Transcriptome , RNA , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/metabolism
3.
Nat Commun ; 14(1): 2122, 2023 04 14.
Article in English | MEDLINE | ID: mdl-37055433

ABSTRACT

Targeting DNA methyltransferase 1 (DNMT1) has immunomodulatory and anti-neoplastic activity, especially when paired with cancer immunotherapies. Here we explore the immunoregulatory functions of DNMT1 in the tumor vasculature of female mice. Dnmt1 deletion in endothelial cells (ECs) impairs tumor growth while priming expression of cytokine-driven cell adhesion molecules and chemokines important for CD8+ T-cell trafficking across the vasculature; consequently, the efficacy of immune checkpoint blockade (ICB) is enhanced. We find that the proangiogenic factor FGF2 promotes ERK-mediated DNMT1 phosphorylation and nuclear translocation to repress transcription of the chemokines Cxcl9/Cxcl10 in ECs. Targeting Dnmt1 in ECs reduces proliferation but augments Th1 chemokine production and extravasation of CD8+ T-cells, suggesting DNMT1 programs immunologically anergic tumor vasculature. Our study is in good accord with preclinical observations that pharmacologically disrupting DNMT1 enhances the activity of ICB but suggests an epigenetic pathway presumed to be targeted in cancer cells is also operative in the tumor vasculature.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Female , Mice , Animals , Cytokines/metabolism , Endothelial Cells/metabolism , Virus Internalization , Neoplasms/therapy , Neoplasms/metabolism , Chemokine CXCL10/metabolism
4.
Cell Rep ; 37(5): 109953, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731630

ABSTRACT

Receptor clustering is the first and critical step to activate apoptosis by death receptor-5 (DR5). The recent discovery of the autoinhibitory DR5 ectodomain has challenged the long-standing view of its mechanistic activation by the natural ligand Apo2L. Because the autoinhibitory residues have remained unknown, here we characterize a crucial patch of positively charged residues (PPCR) in the highly variable domain of DR5. The PPCR electrostatically separates DR5 receptors to autoinhibit their clustering in the absence of ligand and antibody binding. Mutational substitution and antibody-mediated PPCR interference resulted in increased apoptotic cytotoxic function. A dually specific antibody that enables sustained tampering with PPCR function exceptionally enhanced DR5 clustering and apoptotic activation and distinctively improved the survival of animals bearing aggressive metastatic and recurrent tumors, whereas clinically tested DR5 antibodies without PPCR blockade function were largely ineffective. Our study provides mechanistic insights into DR5 activation and a therapeutic analytical design for potential clinical success.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Apoptosis/drug effects , Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , A549 Cells , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antibody Specificity , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/metabolism , Epitopes , Humans , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplasms/immunology , Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
5.
EMBO Mol Med ; 13(3): e12716, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33587338

ABSTRACT

Lack of effective immune infiltration represents a significant barrier to immunotherapy in solid tumors. Thus, solid tumor-enriched death receptor-5 (DR5) activating antibodies, which generates tumor debulking by extrinsic apoptotic cytotoxicity, remains a crucial alternate therapeutic strategy. Over past few decades, many DR5 antibodies moved to clinical trials after successfully controlling tumors in immunodeficient tumor xenografts. However, DR5 antibodies failed to significantly improve survival in phase-II trials, leading in efforts to generate second generation of DR5 agonists to supersize apoptotic cytotoxicity in tumors. Here we have discovered that clinical DR5 antibodies activate an unexpected immunosuppressive PD-L1 stabilization pathway, which potentially had contributed to their limited success in clinics. The DR5 agonist stimulated caspase-8 signaling not only activates ROCK1 but also undermines proteasome function, both of which contributes to increased PD-L1 stability on tumor cell surface. Targeting DR5-ROCK1-PD-L1 axis markedly increases immune effector T-cell function, promotes tumor regression, and improves overall survival in animal models. These insights have identified a potential clinically viable combinatorial strategy to revive solid cancer immunotherapy using death receptor agonism.


Subject(s)
B7-H1 Antigen , Triple Negative Breast Neoplasms , Animals , Antibodies, Monoclonal , Humans , Immune Evasion , Immunotherapy , rho-Associated Kinases
6.
J Neurodev Disord ; 12(1): 29, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33172406

ABSTRACT

BACKGROUND: Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in the X-linked methyl-CpG binding protein 2 (MeCP2) gene. While MeCP2 mutations are lethal in most males, females survive birth but show severe neurological defects. Because X-chromosome inactivation (XCI) is a random process, approximately 50% of the cells silence the wild-type (WT) copy of the MeCP2 gene. Thus, reactivating the silent WT copy of MeCP2 could provide therapeutic intervention for RTT. METHODS: Toward this goal, we screened ~ 28,000 small-molecule compounds from several libraries using a MeCP2-luciferase reporter cell line and cortical neurons from a MeCP2-EGFP mouse model. We used gain/increase of luminescence or fluorescence as a readout of MeCP2 reactivation and tested the efficacy of these drugs under different drug regimens, conditions, and cellular contexts. RESULTS: We identified inhibitors of the JAK/STAT pathway as XCI-reactivating agents, both by in vitro and ex vivo assays. In particular, we show that AG-490, a Janus Kinase 2 (JAK2) kinase inhibitor, and Jaki, a pan JAK/STAT inhibitor, are capable of reactivating MeCP2 from the inactive X chromosome, in different cellular contexts. CONCLUSIONS: Our results suggest that inhibition of the JAK/STAT pathway is a new potential pathway to reinstate MeCP2 gene expression as an efficient RTT treatment.


Subject(s)
Methyl-CpG-Binding Protein 2 , Rett Syndrome , Animals , Chromosomes , Female , Male , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mutation , Rett Syndrome/drug therapy , Rett Syndrome/genetics , X Chromosome Inactivation
7.
Cancer Res ; 80(21): 4791-4804, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32855208

ABSTRACT

The majority of clinical deaths in patients with triple-negative breast cancer (TNBC) are due to chemoresistance and aggressive metastases, with high prevalence in younger women of African ethnicity. Although tumorigenic drivers are numerous and varied, the drivers of metastatic transition remain largely unknown. Here, we uncovered a molecular dependence of TNBC tumors on the TRIM37 network, which enables tumor cells to resist chemotherapeutic as well as metastatic stress. TRIM37-directed histone H2A monoubiquitination enforces changes in DNA repair that rendered TP53-mutant TNBC cells resistant to chemotherapy. Chemotherapeutic drugs triggered a positive feedback loop via ATM/E2F1/STAT signaling, amplifying the TRIM37 network in chemoresistant cancer cells. High expression of TRIM37 induced transcriptomic changes characteristic of a metastatic phenotype, and inhibition of TRIM37 substantially reduced the in vivo propensity of TNBC cells. Selective delivery of TRIM37-specific antisense oligonucleotides using antifolate receptor 1-conjugated nanoparticles in combination with chemotherapy suppressed lung metastasis in spontaneous metastatic murine models. Collectively, these findings establish TRIM37 as a clinically relevant target with opportunities for therapeutic intervention. SIGNIFICANCE: TRIM37 drives aggressive TNBC biology by promoting resistance to chemotherapy and inducing a prometastatic transcriptional program; inhibition of TRIM37 increases chemotherapy efficacy and reduces metastasis risk in patients with TNBC.


Subject(s)
Drug Resistance, Neoplasm/physiology , Tripartite Motif Proteins/metabolism , Triple Negative Breast Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism , Animals , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Mice , Xenograft Model Antitumor Assays
8.
FASEB J ; 34(6): 7687-7702, 2020 06.
Article in English | MEDLINE | ID: mdl-32277852

ABSTRACT

miR-206, miR-1a-1, and miR-1a-2 are induced during differentiation of skeletal myoblasts and promote myogenesis in vitro. miR-206 is required for skeletal muscle regeneration in vivo. Although this miRNA family is hypothesized to play an essential role in differentiation, a triple knock-out (tKO) of the three genes has not been done to test this hypothesis. We report that tKO C2C12 myoblasts generated using CRISPR/Cas9 method differentiate despite the expected derepression of the miRNA targets. Surprisingly, their mitochondrial function is diminished. tKO mice demonstrate partial embryonic lethality, most likely due to the role of miR-1a in cardiac muscle differentiation. Two tKO mice survive and grow normally to adulthood with smaller myofiber diameter, diminished physical performance, and an increase in PAX7 positive satellite cells. Thus, unlike other miRNAs important in other differentiation pathways, the miR-206 family is not absolutely essential for myogenesis and is instead a modulator of optimal differentiation of skeletal myoblasts.


Subject(s)
MicroRNAs/genetics , Mitochondria/genetics , Muscle Development/genetics , Muscle, Skeletal/physiology , Myoblasts, Skeletal/physiology , Animals , CRISPR-Cas Systems/genetics , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , HEK293 Cells , Humans , Mice , Mice, Knockout , Muscular Diseases/genetics
9.
J Vis Exp ; (147)2019 05 22.
Article in English | MEDLINE | ID: mdl-31180354

ABSTRACT

X chromosome inactivation (XCI) is the random silencing of one X chromosome in females to achieve gene dosage balance between the sexes. As a result, all females are heterozygous for X-linked gene expression. One of the key regulators of XCI is Xist, which is essential for the initiation and maintenance of XCI. Previous studies have identified 13 trans acting X chromosome inactivation factors (XCIFs) using a large-scale, loss-of-function genetic screen. Inhibition of XCIFs, such as ACVR1 and PDPK1, using short-hairpin RNA or small molecule inhibitors, reactivates X chromosome-linked genes in cultured cells. But the feasibility and tolerability of reactivating the inactive X chromosome in vivo remains to be determined. Towards this goal, a XistΔ:Mecp2/Xist:Mecp2-Gfp mouse model has been generated with non-random XCI due to deletion of Xist on one X chromosome. Using this model, the extent of inactive X reactivation was quantitated in the mouse brain following treatment with XCIF inhibitors. Recently published results show, for the first time, that pharmacological inhibition of XCIFs reactivates Mecp2 from the inactive X chromosome in cortical neurons of the living mouse brain.


Subject(s)
Methyl-CpG-Binding Protein 2/genetics , Models, Biological , X Chromosome Inactivation/genetics , Animals , Female , Mice , Mice, Knockout , Neurons/metabolism , RNA, Long Noncoding/genetics , Sequence Deletion , Small Molecule Libraries/pharmacology , X Chromosome/genetics , X Chromosome Inactivation/drug effects
10.
Mol Cell Oncol ; 6(3): 1571984, 2019.
Article in English | MEDLINE | ID: mdl-31131305

ABSTRACT

The structural basis of blocking human epidermal growth factor receptor-2 (HER2) dimerization remains of great interest to generate effective anti-cancer therapies. Despite clinically feasible outcome in mammary tumors, a fine consensus between efficacy and safety remains a critical challenge beyond breast cancer. Here we extrapolate on the balancing act using recently reported clinical findings in salivary ductal carcinomas.

11.
Methods Mol Biol ; 1870: 41-50, 2019.
Article in English | MEDLINE | ID: mdl-30539545

ABSTRACT

X-inactive specific transcript (Xist) is a long noncoding RNA that is essential for initiating and maintaining epigenetic silencing of one copy of the X chromosome in mammalian females. But the mechanism by which Xist localizes and spreads on the X chromosome and facilitates transcriptional silencing remains largely unknown. This limited understanding, at least in part, is due to the technical difficulties in the visualization and functional characterization of Xist. Development of a successful method for Xist tracking is a key to better understanding of the X chromosome silencing, as well as to gain insight into the regulatory role of other long noncoding RNAs. Here, we describe an alternative method for visualization of Xist lncRNA in cells using a CRISPR/Cas9-based approach. This strategy is relatively simple approach to track Xist at different stages of cell differentiation, providing mechanistic insights into the initiation, maintenance, and establishment of X inactivation.


Subject(s)
CRISPR-Cas Systems , Molecular Imaging , RNA, Long Noncoding/genetics , Animals , Cell Line , Cloning, Molecular , Female , Fluorescent Antibody Technique , Gene Editing , Genes, Reporter , Histones/metabolism , Mice , Molecular Imaging/methods , RNA, Guide, Kinetoplastida , RNA, Long Noncoding/metabolism , Transfection
12.
Cancer Cell ; 34(2): 331-345.e11, 2018 08 13.
Article in English | MEDLINE | ID: mdl-30107179

ABSTRACT

Therapeutic antibodies targeting ovarian cancer (OvCa)-enriched receptors have largely been disappointing due to limited tumor-specific antibody-dependent cellular cytotoxicity. Here we report a symbiotic approach that is highly selective and superior compared with investigational clinical antibodies. This bispecific-anchored cytotoxicity activator antibody is rationally designed to instigate "cis" and "trans" cytotoxicity by combining specificities against folate receptor alpha-1 (FOLR1) and death receptor 5 (DR5). Whereas the in vivo agonist DR5 signaling requires FcγRIIB interaction, the FOLR1 anchor functions as a primary clustering point to retain and maintain a high level of tumor-specific apoptosis. The presented proof of concept study strategically makes use of a tumor cell-enriched anchor receptor for agonist death receptor targeting to potentially generate a clinically viable strategy for OvCa.


Subject(s)
Antibodies, Bispecific/therapeutic use , Folate Receptor 1/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Animals , Cell Line, Tumor , Female , Humans , Male , Mice , Mice, Inbred C57BL , Ovarian Neoplasms/pathology , Receptors, IgG/physiology
13.
Proc Natl Acad Sci U S A ; 115(31): 7991-7996, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30012595

ABSTRACT

Rett syndrome (RTT) is a genetic disorder resulting from a loss-of-function mutation in one copy of the X-linked gene methyl-CpG-binding protein 2 (MECP2). Typical RTT patients are females and, due to random X chromosome inactivation (XCI), ∼50% of cells express mutant MECP2 and the other ∼50% express wild-type MECP2. Cells expressing mutant MECP2 retain a wild-type copy of MECP2 on the inactive X chromosome (Xi), the reactivation of which represents a potential therapeutic approach for RTT. Previous studies have demonstrated reactivation of Xi-linked MECP2 in cultured cells by biological or pharmacological inhibition of factors that promote XCI (called "XCI factors" or "XCIFs"). Whether XCIF inhibitors in living animals can reactivate Xi-linked MECP2 in cerebral cortical neurons, the cell type most therapeutically relevant to RTT, remains to be determined. Here, we show that pharmacological inhibitors targeting XCIFs in the PI3K/AKT and bone morphogenetic protein signaling pathways reactivate Xi-linked MECP2 in cultured mouse fibroblasts and human induced pluripotent stem cell-derived postmitotic RTT neurons. Notably, reactivation of Xi-linked MECP2 corrects characteristic defects of human RTT neurons including reduced soma size and branch points. Most importantly, we show that intracerebroventricular injection of the XCIF inhibitors reactivates Xi-linked Mecp2 in cerebral cortical neurons of adult living mice. In support of these pharmacological results, we also demonstrate genetic reactivation of Xi-linked Mecp2 in cerebral cortical neurons of living mice bearing a homozygous XCIF deletion. Collectively, our results further establish the feasibility of pharmacological reactivation of Xi-linked MECP2 as a therapeutic approach for RTT.


Subject(s)
Cerebral Cortex/metabolism , Methyl-CpG-Binding Protein 2 , Mutation , Neurons/metabolism , Rett Syndrome/metabolism , Animals , Cell Line , Cerebral Cortex/pathology , Humans , Methyl-CpG-Binding Protein 2/biosynthesis , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Knockout , Neurons/pathology , Rett Syndrome/drug therapy , Rett Syndrome/genetics , Rett Syndrome/pathology
14.
J Clin Invest ; 128(1): 294-308, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29202482

ABSTRACT

Oncogenomic studies indicate that copy number variation (CNV) alters genes involved in tumor progression; however, identification of specific driver genes affected by CNV has been difficult, as these rearrangements are often contained in large chromosomal intervals among several bystander genes. Here, we addressed this problem and identified a CNV-targeted oncogene by performing comparative oncogenomics of human and zebrafish melanomas. We determined that the gene encoding growth differentiation factor 6 (GDF6), which is the ligand for the BMP family, is recurrently amplified and transcriptionally upregulated in melanoma. GDF6-induced BMP signaling maintained a trunk neural crest gene signature in melanomas. Additionally, GDF6 repressed the melanocyte differentiation gene MITF and the proapoptotic factor SOX9, thereby preventing differentiation, inhibiting cell death, and promoting tumor growth. GDF6 was specifically expressed in melanomas but not melanocytes. Moreover, GDF6 expression levels in melanomas were inversely correlated with patient survival. Our study has identified a fundamental role for GDF6 and BMP signaling in governing an embryonic cell gene signature to promote melanoma progression, thus providing potential opportunities for targeted therapy to treat GDF6-positive cancers.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Growth Differentiation Factor 6/metabolism , Melanoma/metabolism , Neoplasm Proteins/metabolism , Signal Transduction , Animals , Bone Morphogenetic Proteins/genetics , Cell Line, Tumor , Female , Growth Differentiation Factor 6/genetics , HEK293 Cells , Humans , Ligands , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Neoplasm Proteins/genetics
15.
Methods Mol Biol ; 1507: 235-244, 2017.
Article in English | MEDLINE | ID: mdl-27832544

ABSTRACT

In mammals, gene expression is largely controlled at the transcriptional level. In response to environmental or intrinsic signaling, gene expression is often fine-tuned by epigenetic modifications, including DNA methylation and histone modifications. One such histone modification is ubiquitination that predominately occurs in mono-ubiquitinated forms on histone H2A and H2B. We recently identified and characterized a novel E3 ligase called TRIM37 that ubiquitinates H2A. This study highlights the consequence of aberrant histone ubiquitination at the promoters of tumor suppressor genes in breast cancer. Regulatory mechanism by which TRIM37 and other auxiliary proteins are involved in the initiation and progression of breast cancer is of utmost importance toward generating effective therapeutics. Here, we describe a detailed step-by-step process of carrying out in vitro ubiquitination assay using purified histone proteins or reconstituted nucleosomes and affinity-purified recombinant E3 ligase like TRIM37. These experimental procedures are largely based on our studies in mammalian cells and will be a useful tool to identify substrate for E3 ubiquitin ligase as well as characterizing new E3 ligases.


Subject(s)
Epigenesis, Genetic , Histones/metabolism , Ubiquitination , Animals , COS Cells , Chlorocebus aethiops , Chromatography, Affinity , Enzyme Assays , Histones/chemistry , Histones/isolation & purification , Nucleosomes/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/isolation & purification , Ubiquitin-Protein Ligases/metabolism
17.
Nature ; 516(7529): 116-20, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25470042

ABSTRACT

The TRIM37 (also known as MUL) gene is located in the 17q23 chromosomal region, which is amplified in up to ∼ 40% of breast cancers. TRIM37 contains a RING finger domain, a hallmark of E3 ubiquitin ligases, but its protein substrate(s) is unknown. Here we report that TRIM37 mono-ubiquitinates histone H2A, a chromatin modification associated with transcriptional repression. We find that in human breast cancer cell lines containing amplified 17q23, TRIM37 is upregulated and, reciprocally, the major H2A ubiquitin ligase RNF2 (also known as RING1B) is downregulated. Genome-wide chromatin immunoprecipitation (ChIP)-chip experiments in 17q23-amplified breast cancer cells identified many genes, including multiple tumour suppressors, whose promoters were bound by TRIM37 and enriched for ubiquitinated H2A. However, unlike RNF2, which is a subunit of polycomb repressive complex 1 (PRC1), we find that TRIM37 associates with polycomb repressive complex 2 (PRC2). TRIM37, PRC2 and PRC1 are co-bound to specific target genes, resulting in their transcriptional silencing. RNA-interference-mediated knockdown of TRIM37 results in loss of ubiquitinated H2A, dissociation of PRC1 and PRC2 from target promoters, and transcriptional reactivation of silenced genes. Knockdown of TRIM37 in human breast cancer cells containing amplified 17q23 substantially decreases tumour growth in mouse xenografts. Conversely, ectopic expression of TRIM37 renders non-transformed cells tumorigenic. Collectively, our results reveal TRIM37 as an oncogenic H2A ubiquitin ligase that is overexpressed in a subset of breast cancers and promotes transformation by facilitating silencing of tumour suppressors and other genes.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins/genetics , Polycomb Repressive Complex 1/genetics , Animals , Female , Gene Expression Profiling , Gene Knockdown Techniques , Gene Silencing , Heterografts , Histones/metabolism , Humans , MCF-7 Cells , Mice , NIH 3T3 Cells , Oncogene Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
18.
Proc Natl Acad Sci U S A ; 111(35): 12591-8, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25136103

ABSTRACT

X-chromosome inactivation (XCI), the random transcriptional silencing of one X chromosome in somatic cells of female mammals, is a mechanism that ensures equal expression of X-linked genes in both sexes. XCI is initiated in cis by the noncoding Xist RNA, which coats the inactive X chromosome (Xi) from which it is produced. However, trans-acting factors that mediate XCI remain largely unknown. Here, we perform a large-scale RNA interference screen to identify trans-acting XCI factors (XCIFs) that comprise regulators of cell signaling and transcription, including the DNA methyltransferase, DNMT1. The expression pattern of the XCIFs explains the selective onset of XCI following differentiation. The XCIFs function, at least in part, by promoting expression and/or localization of Xist to the Xi. Surprisingly, we find that DNMT1, which is generally a transcriptional repressor, is an activator of Xist transcription. Small-molecule inhibitors of two of the XCIFs can reversibly reactivate the Xi, which has implications for treatment of Rett syndrome and other dominant X-linked diseases. A homozygous mouse knockout of one of the XCIFs, stanniocalcin 1 (STC1), has an expected XCI defect but surprisingly is phenotypically normal. Remarkably, X-linked genes are not overexpressed in female Stc1(-/-) mice, revealing the existence of a mechanism(s) that can compensate for a persistent XCI deficiency to regulate X-linked gene expression.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Glycoproteins/genetics , Methyl-CpG-Binding Protein 2/genetics , RNA, Long Noncoding/genetics , Rett Syndrome/genetics , X Chromosome Inactivation/genetics , Animals , Cerebral Cortex/cytology , Chromones/pharmacology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Embryonic Stem Cells/physiology , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/genetics , Female , Fibroblasts/cytology , Fibroblasts/physiology , Gene Library , Genetic Therapy/methods , Humans , Mammals , Mice , Mice, Knockout , Morpholines/pharmacology , Neurons/cytology , Neurons/physiology , Pyrazoles/pharmacology , RNA, Small Interfering/genetics , Rett Syndrome/therapy , Sulfonamides/pharmacology , Transcriptome , X Chromosome Inactivation/drug effects
19.
Traffic ; 9(6): 871-81, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18331451

ABSTRACT

Exosomes are the newest family member of 'bioactive vesicles' that function to promote intercellular communication. Exosomes are derived from the fusion of multivesicular bodies with the plasma membrane and extracellular release of the intraluminal vesicles. Recent studies have focused on the biogenesis and composition of exosomes as well as regulation of exosome release. Exosomes have been shown to be released by cells of hematopoietic and non-hematopoietic origin, yet their function remains enigmatic. Much of the prior work has focused on exosomes as a source of tumor antigens and in presentation of tumor antigens to T cells. However, new studies have shown that exosomes might also promote cell-to-cell spread of infectious agents. Moreover, exosomes isolated from cells infected with various intracellular pathogens, including Mycobacterium tuberculosis and Toxoplasma gondii, have been shown to contain microbial components and can promote antigen presentation and macrophage activation, suggesting that exosomes may function in immune surveillance. In this review, we summarize our understanding of exosome biogenesis but focus primarily on new insights into exosome function. We also discuss their possible use as disease biomarkers and vaccine candidates.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Endosomes/physiology , Neoplasms/immunology , Animals , Cell Membrane/metabolism , Cytoplasmic Vesicles/metabolism , Endosomes/immunology , Endosomes/microbiology , Forecasting , Macrophage Activation , Macrophages/immunology , Macrophages/microbiology , Models, Biological , Mycobacterium tuberculosis/immunology , Neoplasms/pathology , Toxoplasma/immunology
20.
Blood ; 110(9): 3234-44, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17666571

ABSTRACT

Intracellular pathogens and the molecules they express have limited contact with the immune system. Here, we show that macrophages infected with intracellular pathogens Mycobacterium tuberculosis, M bovis BCG, Salmonella typhimurium, or Toxoplasma gondii release from cells small vesicles known as exosomes which contain pathogen-associated molecular patterns (PAMPs). These exosomes, when exposed to uninfected macrophages, stimulate a proinflammatory response in a Toll-like receptor- and myeloid differentiation factor 88-dependent manner. Further, exosomes isolated from the bronchoalveolar lavage fluid (BALF) of M bovis BCG-infected mice contain the mycobacteria components lipoarabinomannan and the 19-kDa lipoprotein and can stimulate TNF-alpha production in naive macrophages. Moreover, exosomes isolated from M bovis BCG- and M tuberculosis-infected macrophages, when injected intranasally into mice, stimulate TNF-alpha and IL-12 production as well as neutrophil and macrophage recruitment in the lung. These studies identify a previously unknown function for exosomes in promoting intercellular communication during an immune response to intracellular pathogens, and we hypothesize that extracellular release of exosomes containing PAMPs is an important mechanism of immune surveillance.


Subject(s)
Inflammation/etiology , Macrophages/metabolism , Macrophages/microbiology , Macrophages/parasitology , Secretory Vesicles/metabolism , Secretory Vesicles/physiology , Animals , Cells, Cultured , Inflammation/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium bovis/pathogenicity , Mycobacterium tuberculosis/pathogenicity , Salmonella Infections, Animal/metabolism , Salmonella Infections, Animal/pathology , Salmonella typhimurium/pathogenicity , Toxoplasma/pathogenicity , Toxoplasmosis, Animal/metabolism , Toxoplasmosis, Animal/pathology , Tuberculosis/complications , Tuberculosis/metabolism , Tuberculosis/pathology , Tuberculosis/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL
...