Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(29): e2403917121, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38980903

RESUMEN

Pancreatic ductal adenocarcinoma (PDA) is a potentially lethal disease lacking effective treatments. Its immunosuppressive tumor microenvironment (TME) allows it to evade host immunosurveillance and limits response to immunotherapy. Here, using the mouse KRT19-deficient (sgKRT19-edited) PDA model, we find that intratumoral accumulation of natural killer T (NKT) cells is required to establish an immunologically active TME. Mechanistically, intratumoral NKT cells facilitate type I interferon (IFN) production to initiate an antitumor adaptive immune response, and orchestrate the intratumoral infiltration of T cells, dendritic cells, natural killer cells, and myeloid-derived suppressor cells. At the molecular level, NKT cells promote the production of type I IFN through the interaction of their CD40L with CD40 on myeloid cells. To evaluate the therapeutic potential of these observations, we find that administration of folinic acid to mice bearing PDA increases NKT cells in the TME and improves their response to anti-PD-1 antibody treatment. In conclusion, NKT cells have an essential role in the immune response to mouse PDA and are potential targets for immunotherapy.


Asunto(s)
Carcinoma Ductal Pancreático , Células T Asesinas Naturales , Neoplasias Pancreáticas , Microambiente Tumoral , Animales , Ratones , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Células T Asesinas Naturales/inmunología , Microambiente Tumoral/inmunología , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/terapia , Carcinoma Ductal Pancreático/patología , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Inmunoterapia/métodos , Ratones Endogámicos C57BL , Leucovorina/administración & dosificación , Leucovorina/uso terapéutico , Humanos , Células Supresoras de Origen Mieloide/inmunología
2.
Nature ; 581(7806): 100-105, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32376951

RESUMEN

Immune evasion is a major obstacle for cancer treatment. Common mechanisms of evasion include impaired antigen presentation caused by mutations or loss of heterozygosity of the major histocompatibility complex class I (MHC-I), which has been implicated in resistance to immune checkpoint blockade (ICB) therapy1-3. However, in pancreatic ductal adenocarcinoma (PDAC), which is resistant to most therapies including ICB4, mutations that cause loss of MHC-I are rarely found5 despite the frequent downregulation of MHC-I expression6-8. Here we show that, in PDAC, MHC-I molecules are selectively targeted for lysosomal degradation by an autophagy-dependent mechanism that involves the autophagy cargo receptor NBR1. PDAC cells display reduced expression of MHC-I at the cell surface and instead demonstrate predominant localization within autophagosomes and lysosomes. Notably, inhibition of autophagy restores surface levels of MHC-I and leads to improved antigen presentation, enhanced anti-tumour T cell responses and reduced tumour growth in syngeneic host mice. Accordingly, the anti-tumour effects of autophagy inhibition are reversed by depleting CD8+ T cells or reducing surface expression of MHC-I. Inhibition of autophagy, either genetically or pharmacologically with chloroquine, synergizes with dual ICB therapy (anti-PD1 and anti-CTLA4 antibodies), and leads to an enhanced anti-tumour immune response. Our findings demonstrate a role for enhanced autophagy or lysosome function in immune evasion by selective targeting of MHC-I molecules for degradation, and provide a rationale for the combination of autophagy inhibition and dual ICB therapy as a therapeutic strategy against PDAC.


Asunto(s)
Adenocarcinoma/inmunología , Autofagia/inmunología , Carcinoma Ductal Pancreático/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Neoplasias Pancreáticas/inmunología , Escape del Tumor/inmunología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/inmunología , Autofagia/efectos de los fármacos , Autofagia/genética , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/inmunología , Línea Celular Tumoral , Cloroquina/farmacología , Femenino , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Masculino , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Escape del Tumor/efectos de los fármacos
3.
Nature ; 570(7760): 246-251, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31142839

RESUMEN

The identification of lymphocyte subsets with non-overlapping effector functions has been pivotal to the development of targeted therapies in immune-mediated inflammatory diseases (IMIDs)1,2. However, it remains unclear whether fibroblast subclasses with non-overlapping functions also exist and are responsible for the wide variety of tissue-driven processes observed in IMIDs, such as inflammation and damage3-5. Here we identify and describe the biology of distinct subsets of fibroblasts responsible for mediating either inflammation or tissue damage in arthritis. We show that deletion of fibroblast activation protein-α (FAPα)+ fibroblasts suppressed both inflammation and bone erosions in mouse models of resolving and persistent arthritis. Single-cell transcriptional analysis identified two distinct fibroblast subsets within the FAPα+ population: FAPα+THY1+ immune effector fibroblasts located in the synovial sub-lining, and FAPα+THY1- destructive fibroblasts restricted to the synovial lining layer. When adoptively transferred into the joint, FAPα+THY1- fibroblasts selectively mediate bone and cartilage damage with little effect on inflammation, whereas transfer of FAPα+ THY1+ fibroblasts resulted in a more severe and persistent inflammatory arthritis, with minimal effect on bone and cartilage. Our findings describing anatomically discrete, functionally distinct fibroblast subsets with non-overlapping functions have important implications for cell-based therapies aimed at modulating inflammation and tissue damage.


Asunto(s)
Artritis Reumatoide/patología , Fibroblastos/patología , Animales , Huesos/patología , Endopeptidasas , Femenino , Fibroblastos/clasificación , Fibroblastos/metabolismo , Gelatinasas/metabolismo , Humanos , Inflamación/patología , Articulaciones/patología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , RNA-Seq , Serina Endopeptidasas/metabolismo , Análisis de la Célula Individual , Membrana Sinovial/patología , Antígenos Thy-1/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35046049

RESUMEN

Cancer immunotherapy frequently fails because most carcinomas have few T cells, suggesting that cancers can suppress T cell infiltration. Here, we show that cancer cells of human pancreatic ductal adenocarcinoma (PDA), colorectal cancer, and breast cancer are coated with transglutaminase-2 (TGM2)-dependent covalent CXCL12-keratin-19 (KRT19) heterodimers that are organized as filamentous networks. Since a dimeric form of CXCL12 suppresses the motility of human T cells, we determined whether this polymeric CXCL12-KRT19 coating mediated T cell exclusion. Mouse tumors containing control PDA cells exhibited the CXCL12-KRT19 coating, excluded T cells, and did not respond to treatment with anti-PD-1 antibody. Tumors containing PDA cells not expressing either KRT19 or TGM2 lacked the CXCL12-KRT19 coating, were infiltrated with activated CD8+ T cells, and growth was suppressed with anti-PD-1 antibody treatment. Thus, carcinomas assemble a CXCL12-KRT19 coating to evade cancer immune attack.


Asunto(s)
Carcinoma/etiología , Carcinoma/metabolismo , Quimiocina CXCL12/metabolismo , Citotoxicidad Inmunológica , Queratina-19/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Neoplasias de la Mama , Carcinoma/patología , Línea Celular Tumoral , Quimiocina CXCL12/química , Femenino , Humanos , Queratina-19/química , Masculino , Ratones , Repeticiones de Microsatélite , Neoplasias Pancreáticas , Unión Proteica , Multimerización de Proteína , Neoplasias Pancreáticas
5.
Nature ; 551(7681): 512-516, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-29132146

RESUMEN

Pancreatic ductal adenocarcinoma is a lethal cancer with fewer than 7% of patients surviving past 5 years. T-cell immunity has been linked to the exceptional outcome of the few long-term survivors, yet the relevant antigens remain unknown. Here we use genetic, immunohistochemical and transcriptional immunoprofiling, computational biophysics, and functional assays to identify T-cell antigens in long-term survivors of pancreatic cancer. Using whole-exome sequencing and in silico neoantigen prediction, we found that tumours with both the highest neoantigen number and the most abundant CD8+ T-cell infiltrates, but neither alone, stratified patients with the longest survival. Investigating the specific neoantigen qualities promoting T-cell activation in long-term survivors, we discovered that these individuals were enriched in neoantigen qualities defined by a fitness model, and neoantigens in the tumour antigen MUC16 (also known as CA125). A neoantigen quality fitness model conferring greater immunogenicity to neoantigens with differential presentation and homology to infectious disease-derived peptides identified long-term survivors in two independent datasets, whereas a neoantigen quantity model ascribing greater immunogenicity to increasing neoantigen number alone did not. We detected intratumoural and lasting circulating T-cell reactivity to both high-quality and MUC16 neoantigens in long-term survivors of pancreatic cancer, including clones with specificity to both high-quality neoantigens and predicted cross-reactive microbial epitopes, consistent with neoantigen molecular mimicry. Notably, we observed selective loss of high-quality and MUC16 neoantigenic clones on metastatic progression, suggesting neoantigen immunoediting. Our results identify neoantigens with unique qualities as T-cell targets in pancreatic ductal adenocarcinoma. More broadly, we identify neoantigen quality as a biomarker for immunogenic tumours that may guide the application of immunotherapies.


Asunto(s)
Antígenos de Neoplasias/inmunología , Proteínas Bacterianas/inmunología , Supervivientes de Cáncer , Reacciones Cruzadas/inmunología , Neoplasias Pancreáticas/inmunología , Linfocitos T Citotóxicos/inmunología , Adenocarcinoma/sangre , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Antígenos de Neoplasias/genética , Proteínas Bacterianas/sangre , Proteínas Bacterianas/genética , Antígeno Ca-125/genética , Antígeno Ca-125/inmunología , Simulación por Computador , Reacciones Cruzadas/genética , Humanos , Inmunoterapia , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/genética , Pronóstico , Análisis de Supervivencia , Linfocitos T Citotóxicos/citología , Secuenciación del Exoma
6.
Proc Natl Acad Sci U S A ; 117(46): 28960-28970, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33127761

RESUMEN

Inhibition of the chemokine receptor CXCR4 in combination with blockade of the PD-1/PD-L1 T cell checkpoint induces T cell infiltration and anticancer responses in murine and human pancreatic cancer. Here we elucidate the mechanism by which CXCR4 inhibition affects the tumor immune microenvironment. In human immune cell-based chemotaxis assays, we find that CXCL12-stimulated CXCR4 inhibits the directed migration mediated by CXCR1, CXCR3, CXCR5, CXCR6, and CCR2, respectively, chemokine receptors expressed by all of the immune cell types that participate in an integrated immune response. Inhibiting CXCR4 in an experimental cancer medicine study by 1-wk continuous infusion of the small-molecule inhibitor AMD3100 (plerixafor) induces an integrated immune response that is detected by transcriptional analysis of paired biopsies of metastases from patients with microsatellite stable colorectal and pancreatic cancer. This integrated immune response occurs in three other examples of immune-mediated damage to noninfected tissues: Rejecting renal allografts, melanomas clinically responding to anti-PD1 antibody therapy, and microsatellite instable colorectal cancers. Thus, signaling by CXCR4 causes immune suppression in human pancreatic ductal adenocarcinoma and colorectal cancer by impairing the function of the chemokine receptors that mediate the intratumoral accumulation of immune cells.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Inmunidad/inmunología , Páncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores CXCR4/efectos de los fármacos , Receptores CXCR4/metabolismo , Anciano , Bencilaminas , Carcinoma Ductal Pancreático , Quimiocina CXCL12 , Neoplasias Colorrectales/patología , Ciclamas , Femenino , Compuestos Heterocíclicos/antagonistas & inhibidores , Humanos , Inmunoterapia , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/patología , Receptores CCR2/metabolismo , Receptores CXCR3/metabolismo , Receptores CXCR5/metabolismo , Receptores CXCR6/metabolismo , Receptores de Interleucina-8A/metabolismo , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/inmunología , Neoplasias Pancreáticas
7.
Proc Natl Acad Sci U S A ; 116(27): 13490-13497, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31213547

RESUMEN

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.


Asunto(s)
Fibroblastos/patología , Estructuras Linfoides Terciarias/patología , Animales , Modelos Animales de Enfermedad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Interleucina-13/metabolismo , Interleucinas/metabolismo , Linfocitos/patología , Ratones , Glándulas Salivales/patología , Interleucina-22
8.
Br J Cancer ; 125(2): 149-151, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33772153

RESUMEN

A recent Phase 1 clinical study of the immunological effects of inhibiting the chemokine receptor, CXCR4, in patients with pancreatic ductal adenocarcinoma or colorectal cancer suggests that stimulation of CXCR4 on immune cells suppresses the intratumoural immune reaction. Here, we discuss how CXCR4 mediates this response, and how cancer cells elicit it.


Asunto(s)
Neoplasias Colorrectales , Compuestos Heterocíclicos , Neoplasias Pancreáticas , Bencilaminas , Quimiocina CXCL12 , Ciclamas , Movilización de Célula Madre Hematopoyética , Humanos , Receptores CXCR4
9.
Proc Natl Acad Sci U S A ; 115(4): E743-E752, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29311302

RESUMEN

The cancer anorexia cachexia syndrome is a systemic metabolic disorder characterized by the catabolism of stored nutrients in skeletal muscle and adipose tissue that is particularly prevalent in nonsmall cell lung cancer (NSCLC). Loss of skeletal muscle results in functional impairments and increased mortality. The aim of the present study was to characterize the changes in systemic metabolism in a genetically engineered mouse model of NSCLC. We show that a portion of these animals develop loss of skeletal muscle, loss of adipose tissue, and increased inflammatory markers mirroring the human cachexia syndrome. Using noncachexic and fasted animals as controls, we report a unique cachexia metabolite phenotype that includes the loss of peroxisome proliferator-activated receptor-α (PPARα) -dependent ketone production by the liver. In this setting, glucocorticoid levels rise and correlate with skeletal muscle degradation and hepatic markers of gluconeogenesis. Restoring ketone production using the PPARα agonist, fenofibrate, prevents the loss of skeletal muscle mass and body weight. These results demonstrate how targeting hepatic metabolism can prevent muscle wasting in lung cancer, and provide evidence for a therapeutic strategy.


Asunto(s)
Caquexia/prevención & control , Carcinoma de Pulmón de Células no Pequeñas/complicaciones , Fenofibrato/uso terapéutico , Neoplasias Pulmonares/complicaciones , PPAR gamma/agonistas , Aminoácidos/metabolismo , Animales , Caquexia/sangre , Caquexia/etiología , Evaluación Preclínica de Medicamentos , Fenofibrato/farmacología , Gluconeogénesis , Cuerpos Cetónicos/deficiencia , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , PPAR gamma/metabolismo
10.
Oncologist ; 23(1): 116-117, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29021379

RESUMEN

Immune-related radiological and biomarker monitoring in cancer immunotherapy trials permits interrogation of efficacy and reasons for therapeutic failure. We report the results from a cross-sectional analysis of response monitoring in 685 T-cell checkpoint-targeted cancer immunotherapy trials in solid malignancies, as registered on the U.S. National Institutes of Health trial registry by October 2016. Immune-related radiological response criteria were registered for only 25% of clinical trials. Only 38% of trials registered an exploratory immunological biomarker, and registration of immunological biomarkers has decreased over the last 15 years. We suggest that increasing the utilization of immune-related response monitoring across cancer immunotherapy trials will improve analysis of outcomes and facilitate translational efforts to extend the benefit of immunotherapy to a greater proportion of patients with cancer.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Inmunidad Celular/inmunología , Inmunoterapia , Monitorización Inmunológica/estadística & datos numéricos , Neoplasias/tratamiento farmacológico , Estudios Transversales , Humanos , Inmunidad Celular/efectos de los fármacos , Monitorización Inmunológica/métodos , Neoplasias/inmunología , Pronóstico
11.
Adv Exp Med Biol ; 1060: 99-114, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30155624

RESUMEN

The tumor microenvironment comprises a mass of heterogeneous cell types, including immune cells, endothelial cells, and fibroblasts, alongside cancer cells. It is increasingly becoming clear that the development of this support niche is critical to the continued uncontrolled growth of the cancer. The tumor microenvironment contributes to the maintenance of cancer stemness and also directly promotes angiogenesis, invasion, metastasis, and chronic inflammation. In this chapter, we describe on the role of fibroblasts, specifically termed cancer-associated fibroblasts (CAFs), in the promotion and maintenance of cancers. CAFs have a multitude of effects on the growth and maintenance of cancer, and here we focus on their roles in modulating immune cells and responses; CAFs both inhibit immune cell access to the tumor microenvironment and inhibit their functions within the tumor. Finally, we describe the potential modulation of CAF function as an adjunct to bolster the effectiveness of cancer immunotherapies.


Asunto(s)
Células del Estroma/patología , Microambiente Tumoral , Animales , Resistencia a Antineoplásicos , Humanos , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/patología , Neovascularización Patológica/patología , Células del Estroma/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
12.
Proc Natl Acad Sci U S A ; 111(33): 12139-44, 2014 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-25092322

RESUMEN

Fibroblastic reticular cells (FRCs), through their expression of CC chemokine ligand (CCL)19 and CCL21, attract and retain T cells in lymph nodes (LNs), but whether this function applies to both resting and activated T cells has not been examined. Here we describe a model for conditionally depleting FRCs from LNs based on their expression of the diphtheria toxin receptor (DTR) directed by the gene encoding fibroblast activation protein-α (FAP). As expected, depleting FAP(+) FRCs causes the loss of naïve T cells, B cells, and dendritic cells from LNs, and this loss decreases the magnitude of the B- and T-cell responses to a subsequent infection with influenza A virus. In contrast, depleting FAP(+) FRCs during an ongoing influenza infection does not diminish the number or continued response of activated T and B cells in the draining LNs, despite still resulting in the loss of naïve T cells. Therefore, different rules govern the LN trafficking of resting and activated T cells; once a T cell is engaged in antigen-specific clonal expansion, its retention no longer depends on FRCs or their chemokines, CCL19 and CCL21. Our findings suggest that activated T cells remain in the LN because they down-regulate the expression of the sphingosine-1 phosphate receptor-1, which mediates the exit of lymphocytes from secondary lymphoid organs. Therefore, LN retention of naïve lymphocytes and the initiation of an immune response depend on FRCs, but is an FRC independent and possibly cell-autonomous response of activated T cells, which allows the magnitude of clonal expansion to determine LN egress.


Asunto(s)
Linfocitos T CD8-positivos/citología , Ganglios Linfáticos/citología , Animales , Linfocitos T CD8-positivos/inmunología , Cromosomas Artificiales Bacterianos , Femenino , Fibroblastos/citología , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
13.
Proc Natl Acad Sci U S A ; 110(50): 20212-7, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24277834

RESUMEN

An autochthonous model of pancreatic ductal adenocarcinoma (PDA) permitted the analysis of why immunotherapy is ineffective in this human disease. Despite finding that PDA-bearing mice had cancer cell-specific CD8(+) T cells, the mice, like human patients with PDA, did not respond to two immunological checkpoint antagonists that promote the function of T cells: anti-cytotoxic T-lymphocyte-associated protein 4 (α-CTLA-4) and α-programmed cell death 1 ligand 1 (α-PD-L1). Immune control of PDA growth was achieved, however, by depleting carcinoma-associated fibroblasts (CAFs) that express fibroblast activation protein (FAP). The depletion of the FAP(+) stromal cell also uncovered the antitumor effects of α-CTLA-4 and α-PD-L1, indicating that its immune suppressive activity accounts for the failure of these T-cell checkpoint antagonists. Three findings suggested that chemokine (C-X-C motif) ligand 12 (CXCL12) explained the overriding immunosuppression by the FAP(+) cell: T cells were absent from regions of the tumor containing cancer cells, cancer cells were coated with the chemokine, CXCL12, and the FAP(+) CAF was the principal source of CXCL12 in the tumor. Administering AMD3100, a CXCL12 receptor chemokine (C-X-C motif) receptor 4 inhibitor, induced rapid T-cell accumulation among cancer cells and acted synergistically with α-PD-L1 to greatly diminish cancer cells, which were identified by their loss of heterozygosity of Trp53 gene. The residual tumor was composed only of premalignant epithelial cells and inflammatory cells. Thus, a single protein, CXCL12, from a single stromal cell type, the FAP(+) CAF, may direct tumor immune evasion in a model of human PDA.


Asunto(s)
Carcinoma Ductal Pancreático/terapia , Quimiocina CXCL12/metabolismo , Gelatinasas/metabolismo , Inmunoterapia/métodos , Proteínas de la Membrana/metabolismo , Neoplasias Pancreáticas/terapia , Serina Endopeptidasas/metabolismo , Escape del Tumor/genética , Análisis de Varianza , Animales , Secuencia de Bases , Bencilaminas , Carcinoma Ductal Pancreático/inmunología , Ciclamas , Endopeptidasas , Ensayo de Immunospot Ligado a Enzimas , Fibroblastos/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Compuestos Heterocíclicos , Inmunohistoquímica , Ratones , Datos de Secuencia Molecular , Neoplasias Pancreáticas/inmunología , Análisis de Secuencia de ARN
14.
Proc Natl Acad Sci U S A ; 109(33): E2223-9, 2012 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-22745171

RESUMEN

During the primary response, the commitment of the CD8(+) T cell to Blimp-1 expression and the terminal differentiation that Blimp-1 induces must be timed so as not to impair the process of clonal expansion. We determined whether the Hippo pathway, which links cell-cell contact to differentiation in other cell lineages, controls Blimp-1 expression. Activating the CD8(+) T cell with antigen and IL-2 causes expression of the core Hippo pathway components, including the pivotal transcriptional cofactor Yap. Contact between activated CD8(+) T cells induces Hippo pathway-mediated Yap degradation and Blimp-1 expression; a Hippo-resistant, stable form of Yap suppresses Blimp-1 expression. Cytotoxic T lymphocyte antigen 4 (CTLA-4) and CD80 comprise the receptor-ligand pair that mediates contact-dependent Hippo pathway activation. In vivo, CD8(+) T cells expressing Hippo resistant-Yap or lacking CTLA-4 have diminished expression of the senescence marker, KLRG1, during a viral infection. The CTLA-4/Hippo pathway/Blimp-1 system may couple terminal differentiation of CD8(+) T cell with the magnitude of clonal expansion.


Asunto(s)
Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/metabolismo , Activación de Linfocitos/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/inmunología , Factores de Transcripción/metabolismo , Animales , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Linfocitos T CD8-positivos/citología , Diferenciación Celular/inmunología , Membrana Celular/metabolismo , Activación Enzimática , Ligandos , Ratones , Factor 1 de Unión al Dominio 1 de Regulación Positiva
15.
Eur J Immunol ; 43(2): 323-6, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23408319

RESUMEN

Prior murine studies have demonstrated the pivotal role that Blimp-1 has in the exhausted phenotype of T lymphocytes in chronic viral infection. In this issue of the European Journal of Immunology, Seddiki et al. [Eur. J. Immunol. 2013. 43: 510-520] demonstrate the applicability of this research to HIV infection. The authors do so by demonstrating differences in Blimp-1 expression between T lymphocytes isolated from patients with chronic active HIV versus those from long-term nonprogressors and showing that this is matched by differences in the cells' capacity to produce IL-2 and the level of expression of the inhibitory receptor PD-1. The data presented here suggest that this may relate to differential regulation of Blimp-1 by the micro RNA, mIR-9. These findings complement current murine work and fit squarely within the research priorities, as outlined by the International AIDS Society, for determining a cure for AIDS.


Asunto(s)
Infecciones por VIH/inmunología , VIH/inmunología , Proteínas Represoras/inmunología , Animales , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Humanos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Represoras/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología
16.
J Immunol ; 188(5): 2173-8, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22271651

RESUMEN

Persisting infections are often associated with chronic T cell activation. For certain pathogens, this can lead to T cell exhaustion and survival of what is otherwise a cleared infection. In contrast, for herpesviruses, T cells never eliminate infection once it is established. Instead, effective immunity appears to maintain these pathogens in a state of latency. We used infection with HSV to examine whether effector-type T cells undergoing chronic stimulation retained functional and proliferative capacity during latency and subsequent reactivation. We found that latency-associated T cells exhibited a polyfunctional phenotype and could secrete a range of effector cytokines. These T cells were also capable of mounting a recall proliferative response on HSV reactivation and could do so repeatedly. Thus, for this latent infection, T cells subjected to chronic Ag stimulation and periodic reactivation retain the ability to respond to local virus challenge.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/toxicidad , Herpes Simple/inmunología , Herpes Simple/virología , Herpesvirus Humano 1/inmunología , Proteínas del Envoltorio Viral/toxicidad , Activación Viral/inmunología , Latencia del Virus/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/trasplante , Linfocitos T CD8-positivos/virología , Enfermedad Crónica , Epítopos de Linfocito T/administración & dosificación , Epítopos de Linfocito T/inmunología , Ganglios Sensoriales/enzimología , Ganglios Sensoriales/inmunología , Ganglios Sensoriales/patología , Granzimas/biosíntesis , Herpes Simple/patología , Herpesvirus Humano 1/patogenicidad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Envoltorio Viral/administración & dosificación
17.
Nat Genet ; 56(7): 1377-1385, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38886586

RESUMEN

The presence of basal lineage characteristics signifies hyperaggressive human adenocarcinomas of the breast, bladder and pancreas. However, the biochemical mechanisms that maintain this aberrant cell state are poorly understood. Here we performed marker-based genetic screens in search of factors needed to maintain basal identity in pancreatic ductal adenocarcinoma (PDAC). This approach revealed MED12 as a powerful regulator of the basal cell state in this disease. Using biochemical reconstitution and epigenomics, we show that MED12 carries out this function by bridging the transcription factor ΔNp63, a known master regulator of the basal lineage, with the Mediator complex to activate lineage-specific enhancer elements. Consistent with this finding, the growth of basal-like PDAC is hypersensitive to MED12 loss when compared to PDAC cells lacking basal characteristics. Taken together, our genetic screens have revealed a biochemical interaction that sustains basal identity in human cancer, which could serve as a target for tumor lineage-directed therapeutics.


Asunto(s)
Carcinoma Ductal Pancreático , Complejo Mediador , Neoplasias Pancreáticas , Factores de Transcripción , Proteínas Supresoras de Tumor , Humanos , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Complejo Mediador/genética , Complejo Mediador/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , Linaje de la Célula/genética , Elementos de Facilitación Genéticos
18.
Cancer Immunol Res ; : OF1-OF11, 2023 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-37285176

RESUMEN

Stromal fibroblasts reside in inflammatory tissues that are characterized by either immune suppression or activation. Whether and how fibroblasts adapt to these contrasting microenvironments remains unknown. Cancer-associated fibroblasts (CAF) mediate immune quiescence by producing the chemokine CXCL12, which coats cancer cells to suppress T-cell infiltration. We examined whether CAFs can also adopt an immune-promoting chemokine profile. Single-cell RNA sequencing of CAFs from mouse pancreatic adenocarcinomas identified a subpopulation of CAFs with decreased expression of Cxcl12 and increased expression of the T cell-attracting chemokine Cxcl9 in association with T-cell infiltration. TNFα and IFNγ containing conditioned media from activated CD8+ T cells converted stromal fibroblasts from a CXCL12+/CXCL9- immune-suppressive phenotype into a CXCL12-/CXCL9+ immune-activating phenotype. Recombinant IFNγ and TNFα acted together to augment CXCL9 expression, whereas TNFα alone suppressed CXCL12 expression. This coordinated chemokine switch led to increased T-cell infiltration in an in vitro chemotaxis assay. Our study demonstrates that CAFs have a phenotypic plasticity that allows their adaptation to contrasting immune tissue microenvironments.

19.
Cancer Immunol Res ; 2023 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-37216570

RESUMEN

Stromal fibroblasts reside in inflammatory tissues that are characterized by either immune suppression or activation. Whether and how fibroblasts adapt to these contrasting microenvironments remains unknown. Cancer-associated fibroblasts (CAFs) mediate immune quiescence by producing the chemokine CXCL12, which coats cancer cells to suppress T-cell infiltration. We examined whether CAFs can also adopt an immune-promoting chemokine profile. Single-cell RNA-sequencing of CAFs from mouse pancreatic adenocarcinomas identified a sub-population of CAFs with decreased expression of Cxcl12 and increased expression of the T cell-attracting chemokine Cxcl9 in association with T-cell infiltration. TNFα and IFNγ containing conditioned media from activated CD8+ T cells converted stromal fibroblasts from a CXCL12+/CXCL9- immune suppressive phenotype into a CXCL12-/CXCL9+ immune-activating phenotype. Recombinant IFNγ and TNFα acted together to augment CXCL9 expression, whereas TNFα alone suppressed CXCL12 expression. This coordinated chemokine switch led to increased T-cell infiltration in an in vitro chemotaxis assay. Our study demonstrates that CAFs have a phenotypic plasticity that allows their adaptation to contrasting immune tissue microenvironments.

20.
JCI Insight ; 8(21)2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-37751306

RESUMEN

Intratumoral B cell responses are associated with more favorable clinical outcomes in human pancreatic ductal adenocarcinoma (PDAC). However, the antigens driving these B cell responses are largely unknown. We sought to discover these antigens by using single-cell RNA sequencing (scRNA-Seq) and immunoglobulin (Ig) sequencing of tumor-infiltrating immune cells from 7 primary PDAC samples. We identified activated T and B cell responses and evidence of germinal center reactions. Ig sequencing identified plasma cell (PC) clones expressing isotype-switched and hypermutated Igs, suggesting the occurrence of T cell-dependent B cell responses. We assessed the reactivity of 41 recombinant antibodies that represented the products of 235 PCs and 12 B cells toward multiple cell lines and PDAC tissues and observed frequent staining of intracellular self-antigens. Three of these antigens were identified: the filamentous actin (F-actin), the nucleic protein RuvB like AAA ATPase 2 (RUVBL2), and the mitochondrial protein heat shock protein family D (Hsp60) member 1 (HSPD1). Antibody titers against F-actin and HSPD1 were substantially elevated in the plasma of patients with PDAC compared with healthy donors. Thus, PCs in PDAC produce autoantibodies reacting with intracellular self-antigens, which may result from promotion of preexisting, autoreactive B cell responses. These observations indicate the chronic inflammatory microenvironment of PDAC can support the adaptive immune response.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Células Plasmáticas/metabolismo , Autoantígenos , Actinas/metabolismo , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/patología , Microambiente Tumoral , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Proteínas Portadoras , ADN Helicasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA