Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Cell ; 187(8): 2010-2028.e30, 2024 04 11.
Article in English | MEDLINE | ID: mdl-38569542

ABSTRACT

Gut inflammation involves contributions from immune and non-immune cells, whose interactions are shaped by the spatial organization of the healthy gut and its remodeling during inflammation. The crosstalk between fibroblasts and immune cells is an important axis in this process, but our understanding has been challenged by incomplete cell-type definition and biogeography. To address this challenge, we used multiplexed error-robust fluorescence in situ hybridization (MERFISH) to profile the expression of 940 genes in 1.35 million cells imaged across the onset and recovery from a mouse colitis model. We identified diverse cell populations, charted their spatial organization, and revealed their polarization or recruitment in inflammation. We found a staged progression of inflammation-associated tissue neighborhoods defined, in part, by multiple inflammation-associated fibroblasts, with unique expression profiles, spatial localization, cell-cell interactions, and healthy fibroblast origins. Similar signatures in ulcerative colitis suggest conserved human processes. Broadly, we provide a framework for understanding inflammation-induced remodeling in the gut and other tissues.


Subject(s)
Colitis, Ulcerative , Colitis , Animals , Humans , Mice , Colitis/metabolism , Colitis/pathology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , In Situ Hybridization, Fluorescence/methods , Inflammation/metabolism , Inflammation/pathology , Cell Communication , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology
2.
Immunity ; 57(9): 2077-2094.e12, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38906145

ABSTRACT

Tissues are exposed to diverse inflammatory challenges that shape future inflammatory responses. While cellular metabolism regulates immune function, how metabolism programs and stabilizes immune states within tissues and tunes susceptibility to inflammation is poorly understood. Here, we describe an innate immune metabolic switch that programs long-term intestinal tolerance. Intestinal interleukin-18 (IL-18) stimulation elicited tolerogenic macrophages by preventing their proinflammatory glycolytic polarization via metabolic reprogramming to fatty acid oxidation (FAO). FAO reprogramming was triggered by IL-18 activation of SLC12A3 (NCC), leading to sodium influx, release of mitochondrial DNA, and activation of stimulator of interferon genes (STING). FAO was maintained in macrophages by a bistable switch that encoded memory of IL-18 stimulation and by intercellular positive feedback that sustained the production of macrophage-derived 2'3'-cyclic GMP-AMP (cGAMP) and epithelial-derived IL-18. Thus, a tissue-reinforced metabolic switch encodes durable immune tolerance in the gut and may enable reconstructing compromised immune tolerance in chronic inflammation.


Subject(s)
Immune Tolerance , Interleukin-18 , Macrophages , Nucleotides, Cyclic , Interleukin-18/metabolism , Interleukin-18/immunology , Animals , Mice , Nucleotides, Cyclic/metabolism , Macrophages/immunology , Macrophages/metabolism , Humans , Mice, Inbred C57BL , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice, Knockout , Fatty Acids/metabolism , Intestines/immunology , Immunity, Innate , Inflammation/immunology , Inflammation/metabolism , Glycolysis , Oxidation-Reduction
3.
Immunity ; 56(2): 256-271, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36792572

ABSTRACT

The immune system plays critical roles in both autoimmunity and cancer, diseases at opposite ends of the immune spectrum. Autoimmunity arises from loss of T cell tolerance against self, while in cancer, poor immunity against transformed self fails to control tumor growth. Blockade of pathways that preserve self-tolerance is being leveraged to unleash immunity against many tumors; however, widespread success is hindered by the autoimmune-like toxicities that arise in treated patients. Knowledge gained from the treatment of autoimmunity can be leveraged to treat these toxicities in patients. Further, the understanding of how T cell dysfunction arises in cancer can be leveraged to induce a similar state in autoreactive T cells. Here, we review what is known about the T cell response in autoimmunity and cancer and highlight ways in which we can learn from the nexus of these two diseases to improve the application, efficacy, and management of immunotherapies.


Subject(s)
Autoimmune Diseases , Neoplasms , Humans , Autoimmunity , T-Lymphocytes , Neoplasms/therapy , Immune Tolerance , Self Tolerance , Autoimmune Diseases/therapy
4.
Immunity ; 53(3): 658-671.e6, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32937153

ABSTRACT

Identifying signals in the tumor microenvironment (TME) that shape CD8+ T cell phenotype can inform novel therapeutic approaches for cancer. Here, we identified a gradient of increasing glucocorticoid receptor (GR) expression and signaling from naïve to dysfunctional CD8+ tumor-infiltrating lymphocytes (TILs). Conditional deletion of the GR in CD8+ TILs improved effector differentiation, reduced expression of the transcription factor TCF-1, and inhibited the dysfunctional phenotype, culminating in tumor growth inhibition. GR signaling transactivated the expression of multiple checkpoint receptors and promoted the induction of dysfunction-associated genes upon T cell activation. In the TME, monocyte-macrophage lineage cells produced glucocorticoids and genetic ablation of steroidogenesis in these cells as well as localized pharmacologic inhibition of glucocorticoid biosynthesis improved tumor growth control. Active glucocorticoid signaling associated with failure to respond to checkpoint blockade in both preclinical models and melanoma patients. Thus, endogenous steroid hormone signaling in CD8+ TILs promotes dysfunction, with important implications for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Glucocorticoids/metabolism , Macrophages/metabolism , Melanoma, Experimental/pathology , Tumor Microenvironment/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Hematopoiesis/immunology , Hepatocyte Nuclear Factor 1-alpha/biosynthesis , Immune Checkpoint Inhibitors , Lymphocyte Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Signal Transduction/immunology
5.
Proc Natl Acad Sci U S A ; 120(11): e2220272120, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36881624

ABSTRACT

T cells are present in early stages of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and play a major role in disease outcome and long-lasting immunity. Nasal administration of a fully human anti-CD3 monoclonal antibody (Foralumab) reduced lung inflammation as well as serum IL-6 and C-reactive protein in moderate cases of COVID-19. Using serum proteomics and RNA-sequencing, we investigated the immune changes in patients treated with nasal Foralumab. In a randomized trial, mild to moderate COVID-19 outpatients received nasal Foralumab (100 µg/d) given for 10 consecutive days and were compared to patients that did not receive Foralumab. We found that naïve-like T cells were increased in Foralumab-treated subjects and NGK7+ effector T cells were reduced. CCL5, IL32, CST7, GZMH, GZMB, GZMA, PRF1, and CCL4 gene expression were downregulated in T cells and CASP1 was downregulated in T cells, monocytes, and B cells in subjects treated with Foralumab. In addition to the downregulation of effector features, an increase in TGFB1 gene expression in cell types with known effector function was observed in Foralumab-treated subjects. We also found increased expression of GTP-binding gene GIMAP7 in subjects treated with Foralumab. Rho/ROCK1, a downstream pathway of GTPases signaling was downregulated in Foralumab-treated individuals. TGFB1, GIMAP7, and NKG7 transcriptomic changes observed in Foralumab-treated COVID-19 subjects were also observed in healthy volunteers, MS subjects, and mice treated with nasal anti-CD3. Our findings demonstrate that nasal Foralumab modulates the inflammatory response in COVID-19 and provides a novel avenue to treat the disease.


Subject(s)
Antibodies, Monoclonal , COVID-19 , Animals , Humans , Mice , Administration, Intranasal , Antibodies, Monoclonal/therapeutic use , GTP-Binding Proteins , Membrane Proteins , rho-Associated Kinases , SARS-CoV-2 , T-Lymphocytes , Transforming Growth Factor beta1/genetics
6.
Pain ; 165(4): 811-819, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37943081

ABSTRACT

ABSTRACT: Surgery, burns or surgery-free accident are leading causes of scars with altered tissue consistency, a reduced degree of motion and pain. Autologous fat grafting can dramatically improve tissue consistency and elasticity but less frequently results in the reduction of pain. Therefore, we analyzed different cell populations present within the adipose tissue to be engrafted and correlated them with the reduction of pain after surgery. Here, we identify a population of CD3 - CD4 - CD304 + cells present in grafted adipose tissue, whose abundance highly correlates with pain improvement shortly after surgery ( r2 = 0.7243****) as well as persistently over time (3 months later: r2 = 0.6277****, 1 year later: r2 = 0.5346***, and 4 years later: r2 = 0.5223***). These cells are characterized by the absence of the hematopoietic marker CD45, whereas they express CD90 and CD34, which characterize mesenchymal stem cells (MSCs); the concomitant presence of CD10 and CD73 in the plasma membrane supports a function of these cells in pain reduction. We deduce that the enrichment of this adipose tissue-derived MSC subset could enhance the therapeutic properties of adipose grafts and ameliorate localized pain syndromes.


Subject(s)
Mesenchymal Stem Cells , Humans , Adipose Tissue/transplantation , Pain/metabolism , Syndrome , Cell Differentiation , Cells, Cultured
7.
Cell Rep Med ; 5(7): 101640, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38959885

ABSTRACT

CD8+ T cells must persist and function in diverse tumor microenvironments to exert their effects. Thus, understanding common underlying expression programs could better inform the next generation of immunotherapies. We apply a generalizable matrix factorization algorithm that recovers both shared and context-specific expression programs from diverse datasets to a single-cell RNA sequencing (scRNA-seq) compendium of 33,161 CD8+ T cells from 132 patients with seven human cancers. Our meta-single-cell analyses uncover a pan-cancer T cell dysfunction program that predicts clinical non-response to checkpoint blockade in melanoma and highlights CXCR6 as a pan-cancer marker of chronically activated T cells. Cxcr6 is trans-activated by AP-1 and repressed by TCF1. Using mouse models, we show that Cxcr6 deletion in CD8+ T cells increases apoptosis of PD1+TIM3+ cells, dampens CD28 signaling, and compromises tumor growth control. Our study uncovers a TCF1:CXCR6 axis that counterbalances PD1-mediated suppression of CD8+ cell responses and is essential for effective anti-tumor immunity.


Subject(s)
CD28 Antigens , CD8-Positive T-Lymphocytes , Hepatocyte Nuclear Factor 1-alpha , Receptors, CXCR6 , Animals , Humans , Mice , CD28 Antigens/metabolism , CD28 Antigens/genetics , CD28 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/genetics , Neoplasms/pathology , Receptors, CXCR6/metabolism , Receptors, CXCR6/genetics , Signal Transduction , Single-Cell Analysis/methods , Tumor Microenvironment/immunology
8.
Mucosal Immunol ; 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38925529

ABSTRACT

Dietary proteins are taken up by intestinal dendritic cells (DCs), cleaved into peptides, loaded to major histocompatibility complexes, and presented to T cells to generate an immune response. Amino acid (AA)-diets do not have the same effects because AAs cannot bind to major histocompatibility complex to activate T cells. Here, we show that impairment in regulatory T cell generation and loss of tolerance in mice fed a diet lacking whole protein is associated with major transcriptional changes in intestinal DCs including downregulation of genes related to DC maturation, activation and decreased gene expression of immune checkpoint molecules. Moreover, the AA-diet had a profound effect on microbiome composition, including an increase in Akkermansia muciniphilia and Oscillibacter and a decrease in Lactococcus lactis and Bifidobacterium. Although microbiome transfer experiments showed that AA-driven microbiome modulates intestinal DC gene expression, most of the unique transcriptional change in DC was linked to the absence of whole protein in the diet. Our findings highlight the importance of dietary proteins for intestinal DC function and mucosal tolerance.

9.
J Cell Biochem ; 114(1): 35-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22903691

ABSTRACT

Burkitt's lymphoma (BL), one of the most aggressive tumors affecting humans, characterized by the constitutive activation of the Myc oncogene together with the alteration of many other genetic and epigenetic factors. Among them, the INK4a/ARF locus has been well documented to play a central role in BL. Recently, we have discovered that simultaneous deregulation of both DNA methylation patterns and the ubiquitin-dependent proteolysis system is required to completely inactive the INK4/ARF locus, opening new possibilities for treating Burkitt's lymphoma. In this review, we integrate our discovery with the general view of BL and propose a new comprehensive approach to analyze and manage this aggressive disease.


Subject(s)
ADP-Ribosylation Factors/genetics , Burkitt Lymphoma/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-myc/genetics , ADP-Ribosylation Factors/metabolism , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Methylation , Genetic Loci , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Ubiquitin/genetics , Ubiquitin/metabolism
10.
Cancer Cell ; 41(9): 1662-1679.e7, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37625402

ABSTRACT

Stem-like CD8+ T cells are regulated by T cell factor 1 (TCF1) and are considered requisite for immune checkpoint blockade (ICB) response. However, recent findings indicate that reliance on TCF1+CD8+ T cells for ICB efficacy may differ across tumor contexts. We find that TCF1 is essential for optimal priming of tumor antigen-specific CD8+ T cells and ICB response in poorly immunogenic tumors that accumulate TOX+ dysfunctional T cells, but is dispensable for T cell priming and therapy response in highly immunogenic tumors that efficiently expand transitory effectors. Importantly, improving T cell priming by vaccination or by enhancing antigen presentation on tumors rescues the defective responses of TCF1-deficient CD8+ T cells upon ICB in poorly immunogenic tumors. Our study highlights TCF1's role during the early stages of anti-tumor CD8+ T cell responses with important implications for guiding optimal therapeutic interventions in cancers with low TCF1+CD8+ T cells and low-neo-antigen expression.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , T Cell Transcription Factor 1 , Humans , Antibodies , Antigens, Neoplasm , Immunotherapy , T Cell Transcription Factor 1/genetics , Neoplasms/immunology , Neoplasms/therapy
11.
bioRxiv ; 2023 May 09.
Article in English | MEDLINE | ID: mdl-37214800

ABSTRACT

Gut inflammation involves contributions from immune and non-immune cells, whose interactions are shaped by the spatial organization of the healthy gut and its remodeling during inflammation. The crosstalk between fibroblasts and immune cells is an important axis in this process, but our understanding has been challenged by incomplete cell-type definition and biogeography. To address this challenge, we used MERFISH to profile the expression of 940 genes in 1.35 million cells imaged across the onset and recovery from a mouse colitis model. We identified diverse cell populations; charted their spatial organization; and revealed their polarization or recruitment in inflammation. We found a staged progression of inflammation-associated tissue neighborhoods defined, in part, by multiple inflammation-associated fibroblasts, with unique expression profiles, spatial localization, cell-cell interactions, and healthy fibroblast origins. Similar signatures in ulcerative colitis suggest conserved human processes. Broadly, we provide a framework for understanding inflammation-induced remodeling in the gut and other tissues.

12.
Nat Commun ; 12(1): 4907, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34389726

ABSTRACT

The intestinal mucosa constitutes an environment of closely regulated immune cells. Dendritic cells (DC) interact with the gut microbiome and antigens and are important in maintaining gut homeostasis. Here, we investigate DC transcriptome, phenotype and function in five anatomical locations of the gut lamina propria (LP) which constitute different antigenic environments. We show that DC from distinct gut LP compartments induce distinct T cell differentiation and cytokine secretion. We also find that PD-L1+ DC in the duodenal LP and XCR1+ DC in the colonic LP comprise distinct tolerogenic DC subsets that are crucial for gut homeostasis. Mice lacking PD-L1+ and XCR1+ DC have a proinflammatory gut milieu associated with an increase in Th1/Th17 cells and a decrease in Treg cells and have exacerbated disease in the models of 5-FU-induced mucositis and DSS-induced colitis. Our findings identify PD-L1+ and XCR1+ DC as region-specific physiologic regulators of intestinal homeostasis.


Subject(s)
B7-H1 Antigen/immunology , Dendritic Cells/immunology , Homeostasis/immunology , Intestinal Mucosa/immunology , Receptors, Chemokine/immunology , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/metabolism , Feces/microbiology , Female , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/immunology , Homeostasis/genetics , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcriptome/genetics , Transcriptome/immunology
13.
Sci Immunol ; 5(53)2020 11 06.
Article in English | MEDLINE | ID: mdl-33158974

ABSTRACT

Recent advances have redefined a role for T cell factor 1 (TCF1) that goes beyond T cell development and T memory formation and encompasses new functions in the regulation of T cell biology. Here, we discuss the multifaceted and context-dependent role of TCF1 in peripheral T cells, particularly during disease-induced inflammatory states such as autoimmunity, cancer, and chronic infections. Understanding how TCF1 fine-tunes peripheral T cell biology holds the potential to tailor improved immune-targeted therapies.


Subject(s)
Autoimmune Diseases/immunology , Neoplasms/immunology , T Cell Transcription Factor 1/metabolism , T-Lymphocytes/immunology , Virus Diseases/immunology , Animals , Autoimmune Diseases/drug therapy , Cell Communication/drug effects , Cell Communication/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Chromatin Assembly and Disassembly/drug effects , Chromatin Assembly and Disassembly/immunology , Chronic Disease/drug therapy , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Immunologic Memory/drug effects , Immunologic Memory/genetics , Neoplasms/drug therapy , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Virus Diseases/drug therapy , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/immunology
14.
Biochem Pharmacol ; 130: 1-9, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28017775

ABSTRACT

Glioblastoma remains a fatal tumor despite increased knowledge regarding the complex signalling pathways that drive this devastating disease. Recently, immunotherapeutic approaches have shown remarkable and durable responses in various cancers including metastatic melanoma and advanced non-small cell lung cancer. So far, it remains unclear whether these immunotherapeutics may also work against glioblastoma and other tumors residing in the central nervous system. It is well known that patients with glioblastoma suffer from profound local immunosuppression that represents the major hurdle to overcome in the context of immunotherapy. Several studies have demonstrated that this immunosuppressive phenotype is orchestrated by glioma-derived membrane-bound and soluble factors as well as the particular microenvironment within the brain. Here, we discuss the molecular and cellular pathways involved in glioblastoma-mediated inhibition of the immune system and highlight possible treatment approaches aiming at reinvigorating anti-tumor immune responses.


Subject(s)
Brain Neoplasms/immunology , Glioblastoma/immunology , Brain Neoplasms/therapy , Glioblastoma/therapy , Humans , Immune Tolerance , Immunotherapy
15.
Neuro Oncol ; 18(12): 1610-1621, 2016 12.
Article in English | MEDLINE | ID: mdl-27286797

ABSTRACT

BACKGROUND: The vascular endothelial growth factor (VEGF) and transforming growth factor (TGF)-ß pathways regulate key biological features of glioblastoma. Here we explore whether the TGF-ß pathway, which promotes angiogenesis, invasiveness, and immunosuppression, acts as an escape pathway from VEGF inhibition. METHODS: The role of the TGF-ß pathway in escape from VEGF inhibition was assessed in vitro and in vivo and by gene expression profiling in syngeneic mouse glioma models. RESULTS: We found that TGF-ß is an upstream regulator of VEGF, whereas VEGF pathway activity does not alter the TGF-ß pathway in vitro. In vivo, single-agent activity was observed for the VEGF antibody B20-4.1.1 in 3 and for the TGF-ß receptor 1 antagonist LY2157299 in 2 of 4 models. Reduction of tumor volume and blood vessel density, but not induction of hypoxia, correlated with benefit from B20-4.1.1. Reduction of phosphorylated (p)SMAD2 by LY2157299 was seen in all models but did not predict survival. Resistance to B20 was associated with anti-angiogenesis escape pathway gene expression, whereas resistance to LY2157299 was associated with different immune response gene signatures in SMA-497 and GL-261 on transcriptomic profiling. The combination of B20 with LY2157299 was ineffective in SMA-497 but provided prolongation of survival in GL-261, associated with early suppression of pSMAD2 in tumor and host immune cells, prolonged suppression of angiogenesis, and delayed accumulation of tumor infiltrating microglia/macrophages. CONCLUSIONS: Our study highlights the biological heterogeneity of murine glioma models and illustrates that cotargeting of the VEGF and TGF-ß pathways might lead to improved tumor control only in subsets of glioblastoma.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Lymphotoxin-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/administration & dosage , Animals , Bevacizumab/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Lymphotoxin-alpha/antagonists & inhibitors , Phosphorylation , Pyrazoles/administration & dosage , Quinolines/administration & dosage , Signal Transduction , Smad2 Protein/metabolism
16.
PLoS One ; 8(9): e74466, 2013.
Article in English | MEDLINE | ID: mdl-24147153

ABSTRACT

Glioblastoma multiforme (GBM) is one of the most deadly types of cancer. To date, the best clinical approach for treatment is based on administration of temozolomide (TMZ) in combination with radiotherapy. Much evidence suggests that the intracellular level of the alkylating enzyme O(6)-methylguanine-DNA methyltransferase (MGMT) impacts response to TMZ in GBM patients. MGMT expression is regulated by the methylation of its promoter. However, evidence indicates that this is not the only regulatory mechanism present. Here, we describe a hitherto unknown microRNA-mediated mechanism of MGMT expression regulation. We show that miR-221 and miR-222 are upregulated in GMB patients and that these paralogues target MGMT mRNA, inducing greater TMZ-mediated cell death. However, miR-221/miR-222 also increase DNA damage and, thus, chromosomal rearrangements. Indeed, miR-221 overexpression in glioma cells led to an increase in markers of DNA damage, an effect rescued by re-expression of MGMT. Thus, chronic miR-221/222-mediated MGMT downregulation may render cells unable to repair genetic damage. This, associated also to miR-221/222 oncogenic potential, may poor GBM prognosis.


Subject(s)
DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Glioma/genetics , MicroRNAs/genetics , RNA Interference , RNA, Messenger/genetics , Tumor Suppressor Proteins/genetics , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/genetics , Cell Line, Tumor , DNA Damage/drug effects , DNA Damage/genetics , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Humans , Temozolomide
SELECTION OF CITATIONS
SEARCH DETAIL