Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Discov ; 12(11): 2606-2625, 2022 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-36027053

RESUMO

It is currently accepted that cancer-associated fibroblasts (CAF) participate in T-cell exclusion from tumor nests. To unbiasedly test this, we used single-cell RNA sequencing coupled with multiplex imaging on a large cohort of lung tumors. We identified four main CAF populations, two of which are associated with T-cell exclusion: (i) MYH11+αSMA+ CAF, which are present in early-stage tumors and form a single cell layer lining cancer aggregates, and (ii) FAP+αSMA+ CAF, which appear in more advanced tumors and organize in patches within the stroma or in multiple layers around tumor nests. Both populations orchestrate a particular structural tissue organization through dense and aligned fiber deposition compared with T cell-permissive CAF. Yet they produce distinct matrix molecules, including collagen IV (MYH11+αSMA+ CAF) and collagen XI/XII (FAP+αSMA+ CAF). Hereby, we uncovered unique molecular programs of CAF driving T-cell marginalization, whose targeting should increase immunotherapy efficacy in patients bearing T cell-excluded tumors. SIGNIFICANCE: The cellular and molecular programs driving T-cell marginalization in solid tumors remain unclear. Here, we describe two CAF populations associated with T-cell exclusion in human lung tumors. We demonstrate the importance of pairing molecular and spatial analysis of the tumor microenvironment, a prerequisite to developing new strategies targeting T cell-excluding CAF. See related commentary by Sherman, p. 2501. This article is highlighted in the In This Issue feature, p. 2483.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias Pulmonares , Humanos , Fibroblastos Associados a Câncer/patologia , Linfócitos T , Microambiente Tumoral , Imunoterapia/métodos , Neoplasias Pulmonares/patologia , Fibroblastos
2.
Front Oncol ; 10: 1683, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33042810

RESUMO

Inflammasomes are molecular complexes that trigger an inflammatory response upon detection of pathogens or danger signals. Recent studies suggest that they are also involved in cancer progression. However, their roles during tumorigenesis remain poorly understood and controversial. Here, we investigated whether inflammasome activation supports mammary tumor growth. Using mouse models of invasive breast cancer, our results demonstrate that the absence of a functional inflammasome impairs tumor growth. Importantly, tumors implanted into inflammasome-deficient mice recruited significantly less neutrophils and more natural killer (NK) cells, and these latter cells displayed a more active phenotype. Interestingly, NK cell depletion abolished the anti-tumoral effect observed in inflammasome-deficient mice, although inflammasome-regulated cytokine neutralization had no effect. Thus, our work identifies a novel role for the inflammasome in supporting mammary tumor growth by attenuating NK cell recruitment and activity. These results suggest that inflammasome inhibition could be a putative target for treating invasive breast cancers.

3.
EMBO Mol Med ; 8(8): 863-77, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27378792

RESUMO

In a number of human cancers, NTN1 upregulation inhibits apoptosis induced by its so-called dependence receptors DCC and UNC5H, thus promoting tumor progression. In other cancers however, the selective inhibition of this dependence receptor death pathway relies on the silencing of pro-apoptotic effector proteins. We show here that a substantial fraction of human breast tumors exhibits simultaneous DNA methylation-dependent loss of expression of NTN1 and of DAPK1, a serine threonine kinase known to transduce the netrin-1 dependence receptor pro-apoptotic pathway. The inhibition of DNA methylation by drugs such as decitabine restores the expression of both NTN1 and DAPK1 in netrin-1-low cancer cells. Furthermore, a combination of decitabine with NTN1 silencing strategies or with an anti-netrin-1 neutralizing antibody potentiates tumor cell death and efficiently blocks tumor growth in different animal models. Thus, combining DNA methylation inhibitors with netrin-1 neutralizing agents may be a valuable strategy for combating cancer.


Assuntos
Neoplasias da Mama/patologia , Metilação de DNA , Regulação para Baixo , Fatores de Crescimento Neural/biossíntese , Proteínas Supressoras de Tumor/biossíntese , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proteínas Quinases Associadas com Morte Celular/biossíntese , Humanos , Netrina-1
4.
Cancer Res ; 73(15): 4629-40, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23722543

RESUMO

Plasmacytoid dendritic cells (pDC) are key regulators of antiviral immunity. In previous studies, we reported that pDC-infiltrating human primary breast tumors represent an independent prognostic factor associated with poor outcome. To understand this negative impact of tumor-associated pDC (TApDC), we developed an orthotopic murine mammary tumor model that closely mimics the human pathology, including pDC and regulatory T cell (Treg) infiltration. We showed that TApDC are mostly immature and maintain their ability to internalize antigens in vivo and to activate CD4(+) T cells. Most importantly, TApDC were specifically altered for cytokine production in response to Toll-like receptor (TLR)-9 ligands in vitro while preserving unaltered response to TLR7 ligands (TLR7L). In vivo pDC depletion delayed tumor growth, showing that TApDC provide an immune-subversive environment, most likely through Treg activation, thus favoring tumor progression. However, in vivo intratumoral administration of TLR7L led to TApDC activation and displayed a potent curative effect. Depletion of pDC and type I IFN neutralization prevented TLR7L antitumoral effect. Our results establish a direct contribution of TApDC to primary breast tumor progression and rationalize the application of TLR7 ligands to restore TApDC activation in breast cancer. Cancer Res; 73(15); 4629-40. ©2013 AACR.


Assuntos
Células Dendríticas/imunologia , Ativação Linfocitária/imunologia , Neoplasias Mamárias Experimentais/imunologia , Glicoproteínas de Membrana/imunologia , Receptor 7 Toll-Like/imunologia , Animais , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Feminino , Ligantes , Teste de Cultura Mista de Linfócitos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Receptor 7 Toll-Like/metabolismo
5.
Cancer Res ; 72(20): 5188-97, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22836755

RESUMO

Infiltration and dysfunction of immune cells have been documented in many types of cancers. We previously reported that plasmacytoid dendritic cells (pDC) within primary breast tumors correlate with an unfavorable prognosis for patients. The role of pDC in cancer remains unclear but they have been shown to mediate immune tolerance in other pathophysiologic contexts. We postulated that pDC may interfere with antitumor immune response and favor tolerance in breast cancer. The present study was designed to decipher the mechanistic basis for the deleterious impact of pDC on the clinical outcome. Using fresh human breast tumor biopsies (N = 60 patients), we observed through multiparametric flow cytometry increased tumor-associated (TA) pDC (TApDC) rates in aggressive breast tumors, i.e., those with high mitotic index and the so-called triple-negative breast tumors (TNBT). Furthermore, TApDC expressed a partially activated phenotype and produced very low amounts of IFN-α following toll-like receptor activation in vitro compared with patients' blood pDC. Within breast tumors, TApDC colocalized and strongly correlated with TA regulatory T cells (TATreg), especially in TNBT. Of most importance, the selective suppression of IFN-α production endowed TApDC with the unique capacity to sustain FoxP3(+) Treg expansion, a capacity that was reverted by the addition of exogenous IFN-α. These findings indicate that IFN-α-deficient TApDC accumulating in aggressive tumors are involved in the expansion of TATreg in vivo, contributing to tumor immune tolerance and poor clinical outcome. Thus, targeting pDC to restore their IFN-α production may represent an attractive therapeutic strategy to overcome immune tolerance in breast cancer.


Assuntos
Neoplasias da Mama/imunologia , Células Dendríticas/metabolismo , Interferon-alfa/biossíntese , Linfócitos T Reguladores/imunologia , Neoplasias da Mama/patologia , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Microambiente Tumoral
6.
PLoS Genet ; 8(5): e1002723, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22654675

RESUMO

The epithelial-mesenchymal transition (EMT) is an embryonic transdifferentiation process consisting of conversion of polarized epithelial cells to motile mesenchymal ones. EMT-inducing transcription factors are aberrantly expressed in multiple tumor types and are known to favor the metastatic dissemination process. Supporting oncogenic activity within primary lesions, the TWIST and ZEB proteins can prevent cells from undergoing oncogene-induced senescence and apoptosis by abolishing both p53- and RB-dependent pathways. Here we show that they also downregulate PP2A phosphatase activity and efficiently cooperate with an oncogenic version of H-RAS in malignant transformation of human mammary epithelial cells. Thus, by down-regulating crucial tumor suppressor functions, EMT inducers make cells particularly prone to malignant conversion. Importantly, by analyzing transformed cells generated in vitro and by characterizing novel transgenic mouse models, we further demonstrate that cooperation between an EMT inducer and an active form of RAS is sufficient to trigger transformation of mammary epithelial cells into malignant cells exhibiting all the characteristic features of claudin-low tumors, including low expression of tight and adherens junction genes, EMT traits, and stem cell-like characteristics. Claudin-low tumors are believed to be the most primitive breast malignancies, having arisen through transformation of an early epithelial precursor with inherent stemness properties and metaplastic features. Challenging this prevailing view, we propose that these aggressive tumors arise from cells committed to luminal differentiation, through a process driven by EMT inducers and combining malignant transformation and transdifferentiation.


Assuntos
Neoplasias da Mama , Transformação Celular Neoplásica , Claudinas , Transição Epitelial-Mesenquimal , Glândulas Mamárias Humanas/metabolismo , Proteína Fosfatase 2 , Proteína 1 Relacionada a Twist/genética , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Diferenciação Celular , Linhagem Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Claudinas/genética , Claudinas/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Genes ras , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Humanas/citologia , Camundongos , Camundongos Transgênicos , Proteína Fosfatase 2/antagonistas & inibidores , Proteína Fosfatase 2/metabolismo , Proteína do Retinoblastoma/metabolismo , Telomerase/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco
7.
Blood ; 118(19): 5130-40, 2011 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21937703

RESUMO

Absent in peripheral tissues during homeostasis, human plasmacytoid dendritic cells (pDCs) are described in inflamed skin or mucosa. Here, we report that, unlike blood pDCs, a subset of tonsil pDCs express functional CCR6 and CCR10, and their respective ligands CCL20 and CCL27are detected in inflamed epithelia contacting blood dendritic cell antigen 2(+) pDCs. Moreover, pDCs are recruited to imiquimod-treated skin tumors in WT but not CCR6-deficient mice, and competitive adoptive transfers reveal that CCR6-deficient pDCs are impaired in homing to inflamed skin tumors after intravenous transfer. On IL-3 culture, CCR6 and CCR10 expression is induced on human blood pDCs that become responsive to CCL20 and CCL27/CCL28, respectively. Interestingly, unlike myeloid DC, blood pDCs initially up-regulate CCR7 expression and CCL19 responsiveness on IL-3 ± CpG-B and then acquire functional CCR6 and CCR10. Finally, IL-3-differentiated CCR6(+) CCR10(+) pDCs secrete high levels of IFN-α in response to virus. Overall, we propose an unexpected pDCs migratory model that may best apply for mucosal-associated lymphoid tissues. After CCR7-mediated extravasation into lymphoid tissues draining inflamed epithelia, blood pDCs may be instructed to up-regulate CCR6 and/or CCR10 allowing their homing into inflamed epithelia (in mucosae or skin). At this site, pDCs can then produce IFN-α contributing to pathogen clearance and/or local inflammation.


Assuntos
Células Dendríticas/imunologia , Inflamação/imunologia , Receptores CCR10/metabolismo , Receptores CCR6/metabolismo , Transferência Adotiva , Animais , Diferenciação Celular/imunologia , Movimento Celular/imunologia , Quimiocina CCL19/farmacologia , Quimiocina CCL20/farmacologia , Células Dendríticas/patologia , Epitélio/imunologia , Epitélio/patologia , Feminino , Humanos , Inflamação/patologia , Interferon-alfa/biossíntese , Interleucina-3/farmacologia , Ligantes , Tecido Linfoide/imunologia , Tecido Linfoide/patologia , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Imunológicos , Tonsila Palatina/citologia , Tonsila Palatina/imunologia , Receptores CCR6/deficiência , Receptores CCR6/genética , Receptor 7 Toll-Like/metabolismo
8.
Cancer Cell ; 14(1): 79-89, 2008 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-18598946

RESUMO

Twist1 and Twist2 are major regulators of embryogenesis. Twist1 has been shown to favor the metastatic dissemination of cancer cells through its ability to induce an epithelial-mesenchymal transition (EMT). Here, we show that a large fraction of human cancers overexpress Twist1 and/or Twist2. Both proteins override oncogene-induced premature senescence by abrogating key regulators of the p53- and Rb-dependent pathways. Twist1 and Twist2 cooperate with Ras to transform mouse embryonic fibroblasts. Interestingly, in epithelial cells, the oncogenic cooperation between Twist proteins and activated mitogenic oncoproteins, such as Ras or ErbB2, leads to complete EMT. These findings suggest an unanticipated direct link between early escape from failsafe programs and the acquisition of invasive features by cancer cells.


Assuntos
Transdiferenciação Celular , Transformação Celular Neoplásica/metabolismo , Senescência Celular , Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Animais , Linhagem Celular , Transdiferenciação Celular/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Senescência Celular/genética , Cães , Ativação Enzimática , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Fibroblastos/enzimologia , Fibroblastos/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Glândulas Mamárias Humanas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Nus , Camundongos Transgênicos , Invasividade Neoplásica , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Proteínas Nucleares/genética , Interferência de RNA , Proteínas Repressoras/genética , Proteína do Retinoblastoma/metabolismo , Transfecção , Transplante Heterólogo , Proteína Supressora de Tumor p53/metabolismo , Proteína 1 Relacionada a Twist/genética , Regulação para Cima , Proteínas ras/metabolismo
9.
Int J Cancer ; 111(5): 653-61, 2004 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-15252833

RESUMO

Interleukin-6 (IL-6) is produced at high levels by renal cell carcinoma cell lines. The molecular mechanisms involved in its possible role as an autocrine growth factor were investigated. IL-6 and IL-6 receptor expression was investigated in 8 renal cell carcinoma (RCC) cell lines. The modulation of RCC cell line proliferation by an anti-IL-6 Ab, an IL-6 antisense oligonucleotide (ASON) directed against the second exon of IL-6 and cytokines inhibiting IL-6 production (IL-4 and IL-13) was investigated. All 8 RCC cell lines expressed IL-6 mRNA, produced IL-6 and expressed the soluble and membrane-bound gp130 chain of IL-6 receptor. The gp80 chain of IL-6 receptor was undetectable at the surface of the 8 RCC cell lines tested, while the soluble form of gp80 was detectable in the supernatant of one of these cell lines. The addition of a blocking IL-6 Ab did not inhibit the proliferation of any of the 8 RCC cell lines. In contrast, IL-6 ASON inhibited specifically IL-6 production and the proliferation of all RCC cell lines. Exogenous IL-6 failed to restore RCC cell line proliferation blocked by ASON, indicating that IL-6 acts through an intracrine loop in RCC cell lines. IL-13 and IL-4 inhibited the proliferation of 7 of the 8 cell lines without interfering with IL-6 or IL-6 receptor expression. IL-6 ASON inhibited the proliferation of the 8 RCC cell lines tested additively with IL-4 or IL-13. IL-6 is an intracrine growth factor in renal cell carcinoma cell lines.


Assuntos
Carcinoma de Células Renais/fisiopatologia , Interleucina-6/biossíntese , Interleucina-6/farmacologia , Neoplasias Renais/fisiopatologia , Receptores de Interleucina-6/biossíntese , Humanos , Interleucina-3/farmacologia , Interleucina-4/farmacologia , Oligonucleotídeos Antissenso/farmacologia , Receptores de Interleucina-6/fisiologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...