Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Sci Adv ; 9(48): eadh2708, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-38019914

RESUMO

Mature lymphoid stromal cells (LSCs) are key organizers of immune responses within secondary lymphoid organs. Similarly, inflammation-driven tertiary lymphoid structures depend on immunofibroblasts producing lymphoid cytokines and chemokines. Recent studies have explored the origin and heterogeneity of LSC/immunofibroblasts, yet the molecular and epigenetic mechanisms involved in their commitment are still unknown. This study explored the transcriptomic and epigenetic reprogramming underlying LSC/immunofibroblast commitment. We identified the induction of lysine demethylase 6B (KDM6B) as the primary epigenetic driver of early immunofibroblast differentiation. In addition, we observed an enrichment for KDM6B gene signature in murine inflammatory fibroblasts and pathogenic stroma of patients with autoimmune diseases. Last, KDM6B was required for the acquisition of LSC/immunofibroblast functional properties, including the up-regulation of CCL2 and the resulting recruitment of monocytes. Overall, our results reveal epigenetic mechanisms that participate in the early commitment and immune properties of immunofibroblasts and support the use of epigenetic modifiers as fibroblast-targeting strategies in chronic inflammation.


Assuntos
Epigênese Genética , Células Estromais , Animais , Humanos , Camundongos , Diferenciação Celular/genética , Inflamação , Histona Desmetilases com o Domínio Jumonji/genética , Regulação para Cima
2.
Artigo em Inglês | MEDLINE | ID: mdl-36266053

RESUMO

BACKGROUND AND OBJECTIVES: Tertiary lymphoid structures and aggregates are reported in the meninges of patients with multiple sclerosis (MS), especially at the progressive stage, and are strongly associated with cortical lesions and disability. Besides B cells, these structures comprise follicular helper T (Tfh) cells that are crucial to support B-cell differentiation. Tfh cells play a pivotal role in amplifying autoreactive B cells and promoting autoantibody production in several autoimmune diseases, but very few are known in MS. In this study, we examined the phenotype, frequency, and transcriptome of circulating cTfh cells in the blood and CSF of patients with relapsing-remitting MS (RRMS). METHODS: The phenotype and frequency of cTfh cells were analyzed in the blood of 39 healthy controls and 41 untreated patients with RRMS and in the CSF and paired blood of 10 patients with drug-naive RRMS at diagnosis by flow cytometry. Using an in vitro model of blood-brain barrier, we assessed the transendothelial migratory abilities of the different cTfh-cell subsets. Finally, we performed an RNA sequencing analysis of paired CSF cTfh cells and blood cTfh cells in 8 patients sampled at their first demyelinating event. RESULTS: The blood phenotype and frequency of cTfh cells were not significantly modified in patients with RRMS. In the CSF, we found an important infiltration of Tfh1 cells, with a high proportion of activated PD1+ cells. We demonstrated that the specific subset of Tfh1 cells presents increased migration abilities to cross an in vitro model of blood-brain barrier. Of interest, even at the first demyelinating event, cTfh cells in the CSF display specific characteristics with upregulation of EOMES gene and proinflammatory/cytotoxic transcriptomic signature able to efficiently distinguish cTfh cells from the CSF and blood. Finally, interactome analysis revealed potential strong cross talk between pathogenic B cells and CSF cTfh cells, pointing out the CSF as opportune supportive compartment and highlighting the very early implication of B-cell helper T cells in MS pathogenesis. DISCUSSION: Overall, CSF enrichment in activated Tfh1 as soon as disease diagnosis, associated with high expression of EOMES, and a predicted high propensity to interact with CSF B cells suggest that these cells probably contribute to disease onset and/or activity.


Assuntos
Esclerose Múltipla , Linfócitos T Auxiliares-Indutores , Humanos , Linfócitos B , Ativação Linfocitária , Contagem de Linfócitos , Esclerose Múltipla/patologia , Proteínas com Domínio T/metabolismo , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Auxiliares-Indutores/patologia , Células Th1
3.
Cytotherapy ; 24(5): 500-507, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35219585

RESUMO

The therapeutic potential of culture-adapted adipose-derived stromal cells (ASCs) is largely related to their production of immunosuppressive factors that are inducible in vitro by priming with inflammatory stimuli, in particular tumor necrosis factor-α (TNFα) and interferon-γ (IFNγ). In vivo, obesity is associated with chronic inflammation of white adipose tissue, including accumulation of neutrophils, infiltration by IFNγ/TNFα-producing immune cells, and ASC dysfunction. In the current study, we identified in obese patients a simultaneous upregulation of CD40Lin the adipose tissue stroma vascular fraction (AT-SVF), correlated with the Th1 gene signature, and an overexpression of CD40 by native ASCs. Moreover, activated CD4+ T cells upregulated CD40 on culture-expanded ASCs and triggered their production of IL-8 in a CD40L-dependent manner, leading to an increased capacity to recruit neutrophils. Finally, activation of ASCs by sCD40L or CD40L-expressing CD4+ T cells relies on both canonical and non-canonical NF-κB pathways, and IL-8 was found to be coregulated with NF-κB family members in AT-SVF. These data identify the CD40-CD40L axis as a priming mechanism of ASCs, able to modulate their cross talk with neutrophils in an inflammatory context, and their functional capacity for therapeutic applications.


Assuntos
Ligante de CD40 , NF-kappa B , Tecido Adiposo , Linfócitos T CD4-Positivos/metabolismo , Ligante de CD40/genética , Ligante de CD40/metabolismo , Células Cultivadas , Quimiocinas/metabolismo , Humanos , Interleucina-8/metabolismo , NF-kappa B/metabolismo , Obesidade , Células Estromais/patologia , Linfócitos T , Fator de Necrose Tumoral alfa/metabolismo
5.
Immunity ; 54(8): 1788-1806.e7, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34166622

RESUMO

Lymphoid stromal cells (LSCs) are essential organizers of immune responses. We analyzed tonsillar tissue by combining flow cytometry, in situ imaging, RNA sequencing, and functional assays, defining three distinct human LSC subsets. The integrin CD49a designated perivascular stromal cells exhibiting features of local committed LSC precursors and segregated cytokine and chemokine-producing fibroblastic reticular cells (FRCs) supporting B and T cell survival. The follicular dendritic cell transcriptional profile reflected active responses to B cell and non-B cell stimuli. We therefore examined the effect of B cell stimuli on LSCs in follicular lymphoma (FL). FL B cells interacted primarily with CD49a+ FRCs. Transcriptional analyses revealed LSC reprogramming in situ downstream of the cytokines tumor necrosis factor (TNF) and transforming growth factor ß (TGF-ß), including increased expression of the chemokines CCL19 and CCL21. Our findings define human LSC populations in healthy tissue and reveal bidirectional crosstalk between LSCs and malignant B cells that may present a targetable axis in lymphoma.


Assuntos
Linfócitos B/imunologia , Células Dendríticas/imunologia , Linfoma Folicular/imunologia , Linfoma Folicular/patologia , Tonsila Palatina/imunologia , Células Estromais/imunologia , Células Cultivadas , Quimiocina CCL19/metabolismo , Quimiocina CCL21/metabolismo , Humanos , Integrina alfa1/metabolismo , Tonsila Palatina/citologia , Transdução de Sinais/imunologia , Células Estromais/citologia , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
6.
Blood ; 138(1): 57-70, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-33881493

RESUMO

Follicular lymphoma (FL) originates in the lymph nodes (LNs) and infiltrates bone marrow (BM) early in the course of the disease. BM FL B cells are characterized by a lower cytological grade, decreased proliferation, and a specific phenotypic and subclonal profile. Mesenchymal stromal cells (MSCs) obtained from FL BM display a specific gene expression profile (GEP), including enrichment for a lymphoid stromal cell signature, and an increased capacity to sustain FL B-cell growth. However, the mechanisms triggering the formation of the medullar FL permissive stromal niche have not been identified. In the current work, we demonstrate that FL B cells produce extracellular vesicles (EVs) that can be internalized by BM-MSCs, making them more efficient to support FL B-cell survival and quiescence. Accordingly, EVs purified from FL BM plasma activate transforming growth factor ß-dependent and independent pathways in BM-MSCs and modify their GEP, triggering an upregulation of factors classically associated with hematopoietic stem cell niche, including CXCL12 and angiopoietin-1. Moreover, we provide the first characterization of BM FL B-cell GEP, allowing the definition of the landscape of molecular interactions they could engage with EV-primed BM-MSCs. This work identifies FL-derived EVs as putative mediators of BM stroma polarization and supports further investigation of their clinical interest for targeting the crosstalk between BM-MSCs and malignant B cells.


Assuntos
Linfócitos B/patologia , Células da Medula Óssea/patologia , Polaridade Celular , Vesículas Extracelulares/patologia , Linfoma Folicular/patologia , Sequência de Bases , Células da Medula Óssea/metabolismo , Comunicação Celular , Diferenciação Celular , Endocitose , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/ultraestrutura , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Humanos , Linfoma Folicular/genética , Heterotrímero de Linfotoxina alfa1 e beta2/metabolismo , Células-Tronco Mesenquimais/metabolismo , Fenótipo , Transdução de Sinais , Células Estromais/metabolismo , Células Estromais/patologia , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/genética
7.
Mol Cell ; 81(7): 1469-1483.e8, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33609448

RESUMO

We demonstrate that DNA hypomethylating agent (HMA) treatment can directly modulate the anti-tumor response and effector function of CD8+ T cells. In vivo HMA treatment promotes CD8+ T cell tumor infiltration and suppresses tumor growth via CD8+ T cell-dependent activity. Ex vivo, HMAs enhance primary human CD8+ T cell activation markers, effector cytokine production, and anti-tumor cytolytic activity. Epigenomic and transcriptomic profiling shows that HMAs vastly regulate T cell activation-related transcriptional networks, culminating with over-activation of NFATc1 short isoforms. Mechanistically, demethylation of an intragenic CpG island immediately downstream to the 3' UTR of the short isoform was associated with antisense transcription and alternative polyadenylation of NFATc1 short isoforms. High-dimensional single-cell mass cytometry analyses reveal a selective effect of HMAs on a subset of human CD8+ T cell subpopulations, increasing both the number and abundance of a granzyme Bhigh, perforinhigh effector subpopulation. Overall, our findings support the use of HMAs as a therapeutic strategy to boost anti-tumor immune response.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Ilhas de CpG/imunologia , Metilação de DNA/efeitos dos fármacos , Decitabina/farmacologia , Granzimas/imunologia , Ativação Linfocitária/efeitos dos fármacos , Metilação de DNA/imunologia , Humanos , Fatores de Transcrição NFATC/imunologia , Perforina/imunologia
8.
Epigenomics ; 12(2): 157-169, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31849241

RESUMO

The tumor microenvironment (TME) plays a central role in tumor development and drug resistance. Within TME, the stromal cell subset, called cancer-associated fibroblasts, is a heterogeneous population originating from poorly characterized precursors. Since cancer-associated fibroblasts do not acquire somatic mutations, other mechanisms like epigenetic regulation, could be involved in the development of these cells and in the acquisition of tumor supportive phenotypes. Moreover, such epigenetic modulations have been correlated to the emergence of an immunosuppressive microenvironment facilitating tumor evasion. These findings underline the need to deepen our knowledge on epigenetic mechanisms driving TME development and function, and to understand the impact of epigenetic drugs that could be used in future to target both tumor cells and their TME.


Assuntos
Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Neoplasias/genética , Microambiente Tumoral , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Fibroblastos Associados a Câncer/metabolismo , Epigênese Genética/efeitos dos fármacos , Humanos , Tolerância Imunológica , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/metabolismo , Microambiente Tumoral/efeitos dos fármacos
9.
Stem Cells ; 38(1): 146-159, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31502731

RESUMO

Clinical-grade mesenchymal stromal cells (MSCs) can be expanded from bone marrow and adipose tissue to treat inflammatory diseases and degenerative disorders. However, the influence of their tissue of origin on their functional properties, including their immunosuppressive activity, remains unsolved. In this study, we produced paired bone marrow-derived mesenchymal stromal cell (BM-MSC) and adipose-derived stromal cell (ASC) batches from 14 healthy donors. We then compared them using transcriptomic, phenotypic, and functional analyses and validated our results on purified native MSCs to infer which differences were really endowed by tissue of origin. Cultured MSCs segregated together owing to their tissue of origin based on their gene expression profile analyzed using differential expression and weighted gene coexpression network analysis. This translated into distinct immune-related gene signatures, phenotypes, and functional cell interactions. Importantly, sorted native BM-MSCs and ASCs essentially displayed the same distinctive patterns than their in vitro-expanded counterparts. As a whole, ASCs exhibited an immune profile consistent with a stronger inhibition of immune response and a lower immunogenicity, supporting the use of adipose tissue as a valuable source for clinical applications.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Transcriptoma/genética , Adulto , Diferenciação Celular , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Adulto Jovem
10.
Proc Natl Acad Sci U S A ; 116(27): 13490-13497, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31213547

RESUMO

Resident fibroblasts at sites of infection, chronic inflammation, or cancer undergo phenotypic and functional changes to support leukocyte migration and, in some cases, aggregation into tertiary lymphoid structures (TLS). The molecular programming that shapes these changes and the functional requirements of this population in TLS development are unclear. Here, we demonstrate that external triggers at mucosal sites are able to induce the progressive differentiation of a population of podoplanin (pdpn)-positive stromal cells into a network of immunofibroblasts that are able to support the earliest phases of TLS establishment. This program of events, that precedes lymphocyte infiltration in the tissue, is mediated by paracrine and autocrine signals mainly regulated by IL13. This initial fibroblast network is expanded and stabilized, once lymphocytes are recruited, by the local production of the cytokines IL22 and lymphotoxin. Interfering with this regulated program of events or depleting the immunofibroblasts in vivo results in abrogation of local pathology, demonstrating the functional role of immunofibroblasts in supporting TLS maintenance in the tissue and suggesting novel therapeutic targets in TLS-associated diseases.


Assuntos
Fibroblastos/patologia , Estruturas Linfoides Terciárias/patologia , Animais , Modelos Animais de Doenças , Citometria de Fluxo , Imunofluorescência , Humanos , Interleucina-13/metabolismo , Interleucinas/metabolismo , Linfócitos/patologia , Camundongos , Glândulas Salivares/patologia , Interleucina 22
11.
Nature ; 563(7732): 579-583, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30429608

RESUMO

The use of liquid biopsies for cancer detection and management is rapidly gaining prominence1. Current methods for the detection of circulating tumour DNA involve sequencing somatic mutations using cell-free DNA, but the sensitivity of these methods may be low among patients with early-stage cancer given the limited number of recurrent mutations2-5. By contrast, large-scale epigenetic alterations-which are tissue- and cancer-type specific-are not similarly constrained6 and therefore potentially have greater ability to detect and classify cancers in patients with early-stage disease. Here we develop a sensitive, immunoprecipitation-based protocol to analyse the methylome of small quantities of circulating cell-free DNA, and demonstrate the ability to detect large-scale DNA methylation changes that are enriched for tumour-specific patterns. We also demonstrate robust performance in cancer detection and classification across an extensive collection of plasma samples from several tumour types. This work sets the stage to establish biomarkers for the minimally invasive detection, interception and classification of early-stage cancers based on plasma cell-free DNA methylation patterns.


Assuntos
Ácidos Nucleicos Livres/sangue , Ácidos Nucleicos Livres/metabolismo , Metilação de DNA , DNA de Neoplasias/sangue , DNA de Neoplasias/metabolismo , Detecção Precoce de Câncer/métodos , Neoplasias/classificação , Neoplasias/genética , Adenocarcinoma/sangue , Adenocarcinoma/genética , Animais , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/sangue , Neoplasias Colorretais/genética , Análise Mutacional de DNA , Epigênese Genética , Feminino , Xenoenxertos , Humanos , Biópsia Líquida , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Neoplasias/sangue , Especificidade de Órgãos , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/genética
12.
Oncoimmunology ; 5(3): e1090077, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27141349

RESUMO

Recent studies have demonstrated that DNA demethylation agents can mimic a viral infection by induction of dsRNAs. This viral mimicry leads to an antiviral response mediated by the cytosolic pattern recognition receptor MDA5, followed by MAVS (IPS1) activation, IRF7 nuclear translocation and upregulation of type III Interferon and interferon-stimulated genes.

13.
Oncotarget ; 7(23): 34664-87, 2016 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-27129173

RESUMO

Malignant mesothelioma (MM) is one of the worst cancers in terms of clinical outcome, urging the need to establish and characterize new preclinical tools for investigation of the tumorigenic process, improvement of early diagnosis and evaluation of new therapeutic strategies. For these purposes, we characterized a collection of 27 cell lines established from F344 rats, after 136 to 415 days of induction with crocidolite asbestos administered intraperitoneally. Four mesotheliomas were distinguished from 23 preneoplastic mesothelial cell lines (PN) according to their propensity to generate tumors after orthotopic transplantation into syngeneic rats, their growth pattern, and the expression profile of three genes. PN cell lines were further discriminated into groups / subgroups according to morphology in culture and the expression profiles of 14 additional genes. This approach was completed by analysis of positive and negative immunohistochemical MM markers in the four tumors, of karyotype alterations in the most aggressive MM cell line in comparison with a PN epithelioid cell line, and of human normal mesothelial and mesothelioma cells and a tissue array. Our results showed that both the rat and human MM cell lines shared in common a dramatic decrease in the relative expression of Cdkn2a and of epigenetic regulators, in comparison with PN and normal human mesothelial cells, respectively. In particular, we identified the involvement of the relative expression of the Ten-Eleven Translocation (TET) family of dioxygenases and Dnmt3a in relation to the 5-hydroxymethylcytosine level in malignant transformation and the acquisition of metastatic potential.


Assuntos
5-Metilcitosina/análogos & derivados , Transformação Celular Neoplásica/patologia , Inibidor de Quinase Dependente de Ciclina p18/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Neoplasias Pulmonares/patologia , Mesotelioma/patologia , Oxigenases de Função Mista/metabolismo , Lesões Pré-Cancerosas/patologia , Proteínas Proto-Oncogênicas/metabolismo , 5-Metilcitosina/metabolismo , Animais , Asbesto Crocidolita/toxicidade , Biomarcadores Tumorais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina , DNA Metiltransferase 3A , Células Epiteliais/patologia , Epitélio/patologia , Humanos , Cariótipo , Neoplasias Pulmonares/induzido quimicamente , Mesotelioma/induzido quimicamente , Mesotelioma Maligno , Ratos , Ratos Endogâmicos F344
14.
Oncotarget ; 7(13): 15772-86, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26908456

RESUMO

Chronic periodontitis (CP) is a chronic inflammatory disease independently associated with higher incidence of oral cavity squamous cell carcinoma (OSCC). However, the molecular mechanism responsible for this increased incidence is unknown. Here we profiled the DNA methylome of CP patients and healthy controls and compared to a large set of OSCC samples from TCGA. We observed a significant overlap between the altered DNA methylation patterns in CP and in OSCC, suggesting an emergence of a pre-neoplastic epigenome in CP. Remarkably, the hypermethylated CpGs in CP were significantly enriched for enhancer elements. This aberrant enhancer methylation is functional and able to disrupt enhancer activity by preventing the binding of chromatin looping factors. This study provides new insights on the molecular mechanisms linking chronic inflammation and tumor predisposition, highlighting the role of epigenetic disruption of transcriptional enhancers.


Assuntos
Periodontite Crônica/genética , Elementos Facilitadores Genéticos/genética , Inflamação/genética , Lesões Pré-Cancerosas/genética , Adulto , Carcinoma de Células Escamosas/genética , Metilação de DNA , Epigênese Genética , Feminino , Neoplasias de Cabeça e Pescoço/genética , Humanos , Inflamação/complicações , Masculino , Neoplasias Bucais/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço
15.
Oncotarget ; 6(42): 44892-904, 2015 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-26539644

RESUMO

Attenuated measles virus (MV) is currently being evaluated as an oncolytic virus in clinical trials and could represent a new therapeutic approach for malignant pleural mesothelioma (MPM). Herein, we screened the sensitivity to MV infection and replication of twenty-two human MPM cell lines and some healthy primary cells. We show that MV replicates in fifteen of the twenty-two MPM cell lines. Despite overexpression of CD46 by a majority of MPM cell lines compared to healthy cells, we found that the sensitivity to MV replication did not correlate with this overexpression. We then evaluated the antiviral type I interferon (IFN) responses of MPM cell lines and healthy cells. We found that healthy cells and the seven insensitive MPM cell lines developed a type I IFN response in presence of the virus, thereby inhibiting replication. In contrast, eleven of the fifteen sensitive MPM cell lines were unable to develop a complete type I IFN response in presence of MV. Finally, we show that addition of type I IFN onto MV sensitive tumor cell lines inhibits replication. These results demonstrate that defects in type I IFN response are frequent in MPM and that MV takes advantage of these defects to exert oncolytic activity.


Assuntos
Interferon Tipo I/metabolismo , Vírus do Sarampo/crescimento & desenvolvimento , Mesotelioma/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/crescimento & desenvolvimento , Neoplasias Pleurais/terapia , Replicação Viral , Antígenos CD/metabolismo , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Interações Hospedeiro-Patógeno , Humanos , Interferon Tipo I/imunologia , Vírus do Sarampo/imunologia , Vírus do Sarampo/metabolismo , Proteína Cofatora de Membrana/metabolismo , Mesotelioma/imunologia , Mesotelioma/metabolismo , Mesotelioma/virologia , Vírus Oncolíticos/imunologia , Vírus Oncolíticos/metabolismo , Neoplasias Pleurais/imunologia , Neoplasias Pleurais/metabolismo , Neoplasias Pleurais/virologia , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Fatores de Tempo
16.
Cell ; 162(5): 961-73, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-26317465

RESUMO

DNA-demethylating agents have shown clinical anti-tumor efficacy via an unknown mechanism of action. Using a combination of experimental and bioinformatics analyses in colorectal cancer cells, we demonstrate that low-dose 5-AZA-CdR targets colorectal cancer-initiating cells (CICs) by inducing viral mimicry. This is associated with induction of dsRNAs derived at least in part from endogenous retroviral elements, activation of the MDA5/MAVS RNA recognition pathway, and downstream activation of IRF7. Indeed, disruption of virus recognition pathways, by individually knocking down MDA5, MAVS, or IRF7, inhibits the ability of 5-AZA-CdR to target colorectal CICs and significantly decreases 5-AZA-CdR long-term growth effects. Moreover, transfection of dsRNA into CICs can mimic the effects of 5-AZA-CdR. Together, our results represent a major shift in understanding the anti-tumor mechanisms of DNA-demethylating agents and highlight the MDA5/MAVS/IRF7 pathway as a potentially druggable target against CICs.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Azacitidina/farmacologia , Células Cultivadas , RNA Helicases DEAD-box/metabolismo , Metilação de DNA/efeitos dos fármacos , Decitabina , Retrovirus Endógenos/metabolismo , Humanos , Fator Regulador 7 de Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon , Camundongos , RNA de Cadeia Dupla/metabolismo , Receptores do Ácido Retinoico/metabolismo , Transdução de Sinais
17.
Biomed Res Int ; 2013: 871936, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23509794

RESUMO

MUC1 glycoprotein is often found overexpressed and hypoglycosylated in tumor cells from numerous cancer types. Since its discovery MUC1 has been an attractive target for antitumor immunotherapy. Indeed, in vitro and in vivo experiments have shown T-cell-specific responses against MUC1 in an HLA-restricted and HLA-unrestricted manner, although some animal models have highlighted the possible development of tolerogenic responses against this antigen. These observations permit the development of new T-cell vaccine strategies capable of inducing an MUC1-specific cytotoxic T cell response in cancer patients. Some of these strategies are now being tested in clinical trials against different types of cancer. To date, encouraging clinical responses have been observed with some MUC1 vaccines in phase II/III clinical trials. This paper compiles knowledge regarding MUC1 as a promising tumor antigen for antitumor therapeutic vaccines applicable to numerous cancers. We also summarize the results of MUC1-vaccine-based clinical trials.


Assuntos
Regulação Neoplásica da Expressão Gênica , Mucina-1/metabolismo , Neoplasias/terapia , Linfócitos T Citotóxicos/citologia , Animais , Antígenos de Neoplasias/imunologia , Antineoplásicos/farmacologia , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Antígenos HLA/metabolismo , Humanos , Imunoterapia/métodos , Camundongos , Neoplasias/imunologia , Linfócitos T Citotóxicos/imunologia
18.
Clin Cancer Res ; 19(5): 1147-58, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23339127

RESUMO

PURPOSE: Plasmacytoid dendritic cells (pDC) are antigen-presenting cells specialized in antiviral response. The measles virus vaccine is proposed as an antitumor agent to target and specifically kill tumor cells without infecting healthy cells. EXPERIMENTAL DESIGN: Here, we investigated, in vitro, the effects of measles virus vaccine-infected tumor cells on the phenotype and functions of human pDC. We studied maturation and tumor antigen cross-presentation by pDC, exposed either to the virus alone, or to measles virus vaccine-infected or UV-irradiated tumor cells. RESULTS: We found that only measles virus vaccine-infected cells induced pDC maturation with a strong production of IFN-α, whereas UV-irradiated tumor cells were unable to activate pDC. This IFN-α production was triggered by the interaction of measles virus vaccine single-stranded RNA (ssRNA) with TLR7. We observed that measles virus vaccine-infected tumor cells were phagocytosed by pDC. Interestingly, we showed cross-presentation of the tumor antigen NYESO-1 to a specific CD8(+) T-cell clone when pDC were cocultured with measles virus vaccine-infected tumor cells, whereas pDC were unable to cross-present NYESO-1 after coculture with UV-irradiated tumor cells. CONCLUSIONS: Altogether, our results suggest that the use of measles virus vaccine in antitumor virotherapy induces immunogenic tumor cell death, allowing pDC to mature, produce high amounts of IFN-α, and cross-present tumor antigen, thus representing a mode of recruiting these antigen-presenting cells in the immune response. Clin Cancer Res; 19(5); 1147-58. ©2012 AACR.


Assuntos
Antígenos de Neoplasias/imunologia , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Vacina contra Sarampo/farmacologia , Neoplasias/imunologia , Terapia Viral Oncolítica , Fagocitose/imunologia , Western Blotting , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Proliferação de Células , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Imunofluorescência , Humanos , Interferon-alfa/metabolismo , Interleucina-3/farmacologia , Proteína Cofatora de Membrana/imunologia , Proteína Cofatora de Membrana/metabolismo , Neoplasias/terapia , Neoplasias/virologia , RNA Mensageiro/genética , RNA Viral/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 7 Toll-Like/metabolismo
19.
J Thorac Oncol ; 7(5): 883-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22729037

RESUMO

INTRODUCTION: Malignant pleural mesothelioma (MPM) is a highly aggressive tumor with poor prognosis. One major challenge for this disease is the development of new, early, and highly reliable diagnostic markers. The aim of this study was to compare the diagnostic value of the chemokine chemokine (C-C motif) ligand 2 (CCL2), galectin-3, and the secretory leukocyte peptidase inhibitor (SLPI) with soluble mesothelin-related peptides (SMRP), and to evaluate the diagnostic performance of marker combinations. METHODS: The levels of the different markers were measured by enzyme-linked immunosorbent assay in pleural fluids from patients with MPM (n = 61), adenocarcinomas (ADCA, n = 25), or with benign pleural effusions (BPE, n = 15). RESULTS: SMRP, SLPI, and CCL2 concentrations were significantly higher in pleural effusions from mesothelioma patients. Conversely, galectin-3 levels seemed to be elevated in patients with pulmonary ADCA. Receiver operating characteristic curve analysis revealed that SMRP (area under the curve [AUC] = 0.9059), CCL2 (AUC = 0.7912), galectin-3 (AUC = 0.7584), and SLPI (AUC = 0.7219) were potentially interesting biomarkers for the differentiation of MPM patients from those with BPE or ADCA. Of interest, we showed that the combination of SMRP/CCL2/galectin-3 greatly improved MPM diagnosis (AUC = 0.9680), when compared with SMRP alone. CONCLUSION: The combination of SMRP/CCL2/galectin-3 seems to represent a promising panel of biomarkers for the reliable diagnosis of MPM in pleural fluids.


Assuntos
Adenocarcinoma/diagnóstico , Quimiocina CCL2/metabolismo , Galectina 3/metabolismo , Proteínas de Membrana/metabolismo , Mesotelioma/diagnóstico , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Derrame Pleural/diagnóstico , Adenocarcinoma/metabolismo , Idoso , Área Sob a Curva , Biomarcadores Tumorais/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Masculino , Mesotelioma/metabolismo , Proteínas do Tecido Nervoso , Derrame Pleural/metabolismo , Prognóstico , Curva ROC , Sensibilidade e Especificidade
20.
Eur J Immunol ; 42(3): 783-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22144115

RESUMO

Research into new treatments against malignant pleural mesothelioma (MPM) is of great interest, as this aggressive cancer is often resistant to conventional therapies. One potential strategy is the use of epigenetic drugs, such as 5-aza-2'-deoxycytidine (5-azaCdR), a DNA-hypomethylating drug, and valproate (VPA), a histone deacetylase inhibitor (HDACi). Indeed, these drugs not only trigger MPM cell death, but also induce the expression of cancer testis antigens recognized by CD8(+) T cells, such as New York-esophageal cancer-1 (NY-ESO-1). The objective of this study was to assess effects of these drugs on the expression and recognition by CD8(+) T cells of Mucin1 (MUC1), a tumor-associated antigen that is overexpressed by MPM. MPM tumor cell lines were treated with epigenetic drugs, alone or in combination. MUC1 expression by MPM cells, and its recognition by a MUC1-specific CD8(+) T-cell clone, was downregulated by HDACi when used alone or in combination with 5-azaCdR. This effect was not due to a blocking of the HLA class I presentation pathway in treated MPM cells, as NY-ESO-1 induced by 5-azaCdR alone, or with VPA, was recognized by a NY-ESO-1-specific T-cell clone. This study suggests that the choice of tumor antigens could be critical for strategies combining epigenetic drugs with immunotherapy.


Assuntos
Azacitidina/análogos & derivados , Linfócitos T CD8-Positivos/imunologia , Inibidores de Histona Desacetilases/farmacologia , Mesotelioma/imunologia , Mucina-1/imunologia , Ácido Valproico/farmacologia , Antígenos de Neoplasias/imunologia , Azacitidina/farmacologia , Linhagem Celular Tumoral , Decitabina , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Quimioterapia Combinada , Citometria de Fluxo , Humanos , Cinética , Proteínas de Membrana/imunologia , Mesotelioma/tratamento farmacológico , Mesotelioma/genética , Mucina-1/genética , RNA/química , RNA/genética , Reação em Cadeia da Polimerase em Tempo Real , Estatísticas não Paramétricas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA