Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cell ; 160(3): 367-80, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25619691

ABSTRACT

The discovery that enhancers are regulated transcription units, encoding eRNAs, has raised new questions about the mechanisms of their activation. Here, we report an unexpected molecular mechanism that underlies ligand-dependent enhancer activation, based on DNA nicking to relieve torsional stress from eRNA synthesis. Using dihydrotestosterone (DHT)-induced binding of androgen receptor (AR) to prostate cancer cell enhancers as a model, we show rapid recruitment, within minutes, of DNA topoisomerase I (TOP1) to a large cohort of AR-regulated enhancers. Furthermore, we show that the DNA nicking activity of TOP1 is a prerequisite for robust eRNA synthesis and enhancer activation and is kinetically accompanied by the recruitment of ATR and the MRN complex, followed by additional components of DNA damage repair machinery to the AR-regulated enhancers. Together, our studies reveal a linkage between eRNA synthesis and ligand-dependent TOP1-mediated nicking-a strategy exerting quantitative effects on eRNA expression in regulating AR-bound enhancer-dependent transcriptional programs.


Subject(s)
DNA Topoisomerases, Type I/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation , Receptors, Androgen/metabolism , Cell Line, Tumor , DNA Breaks, Single-Stranded , DNA Repair , DNA Topoisomerases, Type I/genetics , DNA-Binding Proteins/metabolism , Gene Knockdown Techniques , Homeodomain Proteins/metabolism , Humans , MRE11 Homologue Protein , Transcription Factors/metabolism , Transcription, Genetic
2.
Nat Rev Mol Cell Biol ; 18(8): 471-476, 2017 08.
Article in English | MEDLINE | ID: mdl-28537575

ABSTRACT

The idea that signal-dependent transcription might involve the generation of transient DNA nicks or even breaks in the regulatory regions of genes, accompanied by activation of DNA damage repair pathways, would seem to be counterintuitive, as DNA damage is usually considered harmful to cellular integrity. However, recent studies have generated a substantial body of evidence that now argues that programmed DNA single- or double-strand breaks can, at least in specific cases, have a role in transcription regulation. Here, we discuss the emerging functions of DNA breaks in the relief of DNA torsional stress and in promoter and enhancer activation.


Subject(s)
DNA Damage/genetics , DNA/chemistry , Animals , DNA Breaks, Double-Stranded , DNA Repair/genetics , Gene Expression Regulation , Humans
3.
Cancer Cell ; 7(2): 193-204, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15710331

ABSTRACT

Pten-/- cells display a partially defective checkpoint in response to ionizing radiation (IR). The checkpoint defect was traced to the ability of AKT to phosphorylate CHK1 at serine 280, since a nonphosphorylated mutant of CHK1 (S280A) complemented the checkpoint defect and restored CDC25A degradation. CHK1 phosphorylation at serine 280 led to covalent binding of 1 to 2 molecules of ubiquitin and cytoplasmic CHK1 localization. Primary breast carcinomas lacking PTEN expression and having elevated AKT phosphorylation had increased cytoplasmic CHK1 and displayed aneuploidy (p <0.005). We conclude that loss of PTEN and subsequent activation of AKT impair CHK1 through phosphorylation, ubiquitination, and reduced nuclear localization to promote genomic instability in tumor cells.


Subject(s)
Protein Kinases/genetics , Protein Kinases/physiology , Animals , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Checkpoint Kinase 1 , Cytoplasm/metabolism , DNA Damage , Embryo, Mammalian/cytology , G2 Phase , Growth Substances/metabolism , Humans , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Models, Genetic , Mutation , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Plasmids/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Radiation, Ionizing , Serine/chemistry , Signal Transduction , Stem Cells/cytology , Time Factors , Ubiquitin/metabolism
4.
Cancers (Basel) ; 15(23)2023 Nov 23.
Article in English | MEDLINE | ID: mdl-38067255

ABSTRACT

Epithelial cell adhesion molecule (EpCAM) is a tumor-associated antigen that is frequently overexpressed in various carcinomas. We have developed chimeric antigen receptor (CAR) T cells specifically targeting EpCAM for the treatment of gastric cancer. This study sought to unravel the precise mechanisms by which tumors evade immune surveillance and develop resistance to CAR T cell therapy. Through a combination of whole-body CAR T cell imaging and single-cell multiomic analyses, we uncovered intricate interactions between tumors and tumor-infiltrating lymphocytes (TILs). In a gastric cancer model, tumor-infiltrating CD8 T cells exhibited both cytotoxic and exhausted phenotypes, while CD4 T cells were mainly regulatory T cells. A T cell receptor (TCR) clonal analysis provided evidence of CAR T cell proliferation and clonal expansion within resistant tumors, which was substantiated by whole-body CAR T cell imaging. Furthermore, single-cell transcriptomics showed that tumor cells in mice with refractory or relapsing outcomes were enriched for genes involved in major histocompatibility complex (MHC) and antigen presentation pathways, interferon-γ and interferon-α responses, mitochondrial activities, and a set of genes (e.g., CD74, IDO1, IFI27) linked to tumor progression and unfavorable disease prognoses. This research highlights an approach that combines imaging and multiomic methodologies to concurrently characterize the evolution of tumors and the differentiation of CAR T cells.

5.
Nat Commun ; 14(1): 2068, 2023 04 12.
Article in English | MEDLINE | ID: mdl-37045815

ABSTRACT

The limited number of targetable tumor-specific antigens and the immunosuppressive nature of the microenvironment within solid malignancies represent major barriers to the success of chimeric antigen receptor (CAR)-T cell therapies. Here, using epithelial cell adhesion molecule (EpCAM) as a model antigen, we used alanine scanning of the complementarity-determining region to fine-tune CAR affinity. This allowed us to identify CARs that could spare primary epithelial cells while still effectively targeting EpCAMhigh tumors. Although affinity-tuned CARs showed suboptimal antitumor activity in vivo, we found that inducible secretion of interleukin-12 (IL-12), under the control of the NFAT promoter, can restore CAR activity to levels close to that of the parental CAR. This strategy was further validated with another affinity-tuned CAR specific for intercellular adhesion molecule-1 (ICAM-1). Only in affinity-tuned CAR-T cells was NFAT activity stringently controlled and restricted to tumors expressing the antigen of interest at high levels. Our study demonstrates the feasibility of specifically gearing CAR-T cells towards recognition of solid tumors by combining inducible IL-12 expression and affinity-tuned CAR.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Animals , Mice , Interleukin-12/genetics , Epithelial Cell Adhesion Molecule , Immunotherapy, Adoptive , Neoplasms/genetics , Neoplasms/therapy , Antigens, Neoplasm/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Xenograft Model Antitumor Assays , Cell Line, Tumor , Tumor Microenvironment
6.
Cancer Immunol Res ; 9(10): 1158-1174, 2021 10.
Article in English | MEDLINE | ID: mdl-34341066

ABSTRACT

Adoptive transfer of chimeric antigen receptor (CAR) T cells has demonstrated unparalleled responses in hematologic cancers, yet antigen escape and tumor relapse occur frequently. CAR T-cell therapy for patients with solid tumors faces even greater challenges due to the immunosuppressive tumor environment and antigen heterogeneity. Here, we developed a bispecific CAR to simultaneously target epithelial cell adhesion molecule (EpCAM) and intercellular adhesion molecule 1 (ICAM-1) to overcome antigen escape and to improve the durability of tumor responses. ICAM-1 is an adhesion molecule inducible by inflammatory cytokines and elevated in many types of tumors. Our study demonstrates superior efficacy of bispecific CAR T cells compared with CAR T cells targeting a single primary antigen. Bispecific CAR T achieved more durable antitumor responses in tumor models with either homogenous or heterogenous expression of EpCAM. We also showed that the activation of CAR T cells against EpCAM in tumors led to upregulation of ICAM-1, which rendered tumors more susceptible to ICAM-1 targeting by bispecific CAR T cells. Our strategy of additional targeting of ICAM-1 may have broad applications in augmenting the activity of CAR T cells against primary tumor antigens that are prone to antigen loss or downregulation.


Subject(s)
Epithelial Cell Adhesion Molecule/metabolism , Immunotherapy, Adoptive/methods , Intercellular Adhesion Molecule-1/metabolism , Neoplasms/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , Animals , Antigenic Drift and Shift , CRISPR-Cas Systems , Cell Line, Tumor , Cytotoxicity, Immunologic , Epithelial Cell Adhesion Molecule/genetics , Humans , Immunotherapy, Adoptive/adverse effects , Intercellular Adhesion Molecule-1/genetics , Male , Mice , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
7.
Cancer Cell ; 39(4): 566-579.e7, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33848478

ABSTRACT

Small cell neuroendocrine cancers (SCNCs) are recalcitrant cancers arising from diverse primary sites that lack effective treatments. Using chemical genetic screens, we identified inhibition of ataxia telangiectasia and rad3 related (ATR), the primary activator of the replication stress response, and topoisomerase I (TOP1), nuclear enzyme that suppresses genomic instability, as synergistically cytotoxic in small cell lung cancer (SCLC). In a proof-of-concept study, we combined M6620 (berzosertib), first-in-class ATR inhibitor, and TOP1 inhibitor topotecan in patients with relapsed SCNCs. Objective response rate among patients with SCLC was 36% (9/25), achieving the primary efficacy endpoint. Durable tumor regressions were observed in patients with platinum-resistant SCNCs, typically fatal within weeks of recurrence. SCNCs with high neuroendocrine differentiation, characterized by enhanced replication stress, were more likely to respond. These findings highlight replication stress as a potentially transformative vulnerability of SCNCs, paving the way for rational patient selection in these cancers, now treated as a single disease.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Isoxazoles/pharmacology , Lung Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Pyrazines/pharmacology , Small Cell Lung Carcinoma/drug therapy , Aged , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins/genetics , DNA Replication/drug effects , DNA Topoisomerases, Type I/genetics , Genomic Instability/genetics , Humans , Lung Neoplasms/metabolism , Middle Aged , Neoplasm Recurrence, Local/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Small Cell Lung Carcinoma/metabolism
8.
Ann N Y Acad Sci ; 1073: 317-31, 2006 Aug.
Article in English | MEDLINE | ID: mdl-17102102

ABSTRACT

PTEN, a tumor suppressor gene, is frequently mutated in a variety of human tumors. In mice, monoallelic inactivation of this gene predisposes animals to neoplasia of multiple organs. Interestingly, Pten heterozygous mice develop bilateral hyperplasia of the adrenal medulla. In this report we demonstrate that these neoplasms are hormonally active pheochromocytomas that secrete increased amounts of bioactive catecholamines: norepinephrine and epinephrine. To test a possibility that PTEN might be one of the genes responsible for human sporadic pheochromocytoma, we performed mutation analysis of DNA obtained from tumors of 29 patients. However, direct sequencing of all nine exons of the PTEN gene, including the splice junctions, revealed no mutations. Examination of protein expression by immunohistochemistry using 8 normal adrenals and 11 sporadic pheochromocytomas showed no decrease in the PTEN protein expression in the tumor tissue, but upregulation of insulin-like growth factor II, a peptide implicated in growth of adrenal tissue, was observed in four cases (36%).


Subject(s)
Adrenal Gland Neoplasms/genetics , Mutation , PTEN Phosphohydrolase/genetics , Pheochromocytoma/genetics , Animals , Base Sequence , DNA Primers , Immunohistochemistry , Mice
9.
Cell Cycle ; 8(14): 2198-210, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19502790

ABSTRACT

Chromosomes in PTEN deficient cells display both numerical as well as structural alterations including regional amplification. We found that PTEN deficient cells displayed a normal DNA damage response (DDR) as evidenced by the ionizing radiation (IR)-induced phosphorylation of Ataxia Telangiectasia Mutated (ATM) as well as its effectors. PTEN deficient cells also had no defect in Rad51 expression or DNA damage repair kinetics post irradiation. In contrast, caffeine treatment specifically increased IR-induced chromosome aberrations and mitotic index only in cells with PTEN, and not in cells deficient for PTEN, suggesting that their checkpoints were defective. Furthermore, PTEN-deficient cells were unable to maintain active spindle checkpoint after taxol treatment. Genomic instability in PTEN deficient cells could not be attributed to lack of PTEN at centromeres, since no interaction was detected between centromeric DNA and PTEN in wild type cells. These results indicate that PTEN deficiency alters multiple cell cycle checkpoints possibly leaving less time for DNA damage repair and/or chromosome segregation as evidenced by the increased structural as well as numerical alterations seen in PTEN deficient cells.


Subject(s)
Cell Cycle , DNA Repair , Genomic Instability , PTEN Phosphohydrolase/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Ataxia Telangiectasia Mutated Proteins , Caffeine/pharmacology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromosome Aberrations , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Humans , Infrared Rays , Karyotyping , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Paclitaxel/pharmacology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/metabolism , Rad51 Recombinase/metabolism , Telomere/metabolism , Tumor Suppressor Proteins/metabolism
10.
Mol Cell ; 27(4): 609-21, 2007 Aug 17.
Article in English | MEDLINE | ID: mdl-17707232

ABSTRACT

Deciphering the epigenetic "code" remains a central issue in transcriptional regulation. Here, we report the identification of a JAMM/MPN(+) domain-containing histone H2A deubiquitinase (2A-DUB, or KIAA1915/MYSM1) specific for monoubiquitinated H2A (uH2A) that has permitted delineation of a strategy for specific regulatory pathways of gene activation. 2A-DUB regulates transcription by coordinating histone acetylation and deubiquitination, and destabilizing the association of linker histone H1 with nucleosomes. 2A-DUB interacts with p/CAF in a coregulatory protein complex, with its deubiquitinase activity modulated by the status of acetylation of nucleosomal histones. Consistent with this mechanistic role, 2A-DUB participates in transcriptional regulation events in androgen receptor-dependent gene activation, and the levels of uH2A are dramatically decreased in prostate tumors, serving as a cancer-related mark. We suggest that H2A ubiquitination represents a widely used mechanism for many regulatory transcriptional programs and predict that various H2A ubiquitin ligases/deubiquitinases will be identified for specific cohorts of regulated transcription units.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Histones/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Ubiquitins/metabolism , Acetylation/drug effects , Androgens/pharmacology , Animals , Cell Line , Chromatography, Affinity , DNA-Binding Proteins/isolation & purification , Gene Expression Regulation/drug effects , Histone Acetyltransferases/metabolism , Humans , Mice , Models, Genetic , Nucleosomes/drug effects , Nucleosomes/metabolism , Phosphorylation/drug effects , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Trans-Activators , Transcription Factors/isolation & purification , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Ubiquitin-Specific Proteases
12.
Cell Cycle ; 4(7): 927-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15970699

ABSTRACT

CHK1 is an essential kinase involved in the regulation of the cell cycle progression and preservation of genomic integrity. Inhibition of CHK1 leads to the accumulation of double-stranded DNA breaks. Loss of PTEN impairs CHK1-mediated checkpoint activation due to cytoplasmic sequestration of ubiquitinated CHK1. Here, we provide evidence that another consequence of reduced CHK1 function in PTEN deficient cells is the accumulation of double-stranded DNA breaks. Moreover, we show that the site of CHK1 ubiquitination (K274) is near the site of AKT phosphorylation (S280). Overall, these data demonstrate that lack of PTEN generates DNA damage due to inappropriate inactivation of CHK1.DNA damage due to the loss of PTEN is likely to stimulate tumor development.


Subject(s)
DNA Damage/genetics , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Protein Kinases/metabolism , Amino Acid Sequence , Animals , Checkpoint Kinase 1 , Down-Regulation , Mice , Molecular Sequence Data , Mutation/genetics , Oncogene Protein v-akt/metabolism , Protein Binding , Protein Kinases/chemistry , Stem Cells/radiation effects , Ubiquitin/metabolism , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL