Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
4.
Cancer Cell ; 34(5): 757-774.e7, 2018 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-30423296

RESUMEN

Pancreatic ductal adenocarcinoma (PDA) is characterized by immune tolerance and immunotherapeutic resistance. We discovered upregulation of receptor-interacting serine/threonine protein kinase 1 (RIP1) in tumor-associated macrophages (TAMs) in PDA. To study its role in oncogenic progression, we developed a selective small-molecule RIP1 inhibitor with high in vivo exposure. Targeting RIP1 reprogrammed TAMs toward an MHCIIhiTNFα+IFNγ+ immunogenic phenotype in a STAT1-dependent manner. RIP1 inhibition in TAMs resulted in cytotoxic T cell activation and T helper cell differentiation toward a mixed Th1/Th17 phenotype, leading to tumor immunity in mice and in organotypic models of human PDA. Targeting RIP1 synergized with PD1-and inducible co-stimulator-based immunotherapies. Tumor-promoting effects of RIP1 were independent of its co-association with RIP3. Collectively, our work describes RIP1 as a checkpoint kinase governing tumor immunity.


Asunto(s)
Carcinoma Ductal Pancreático/inmunología , Tolerancia Inmunológica/inmunología , Macrófagos/inmunología , Neoplasias Pancreáticas/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Linfocitos T Citotóxicos/inmunología , Células TH1/inmunología , Células Th17/inmunología , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Humanos , Tolerancia Inmunológica/genética , Células L , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Factor de Transcripción STAT1/metabolismo , Células TH1/citología , Células Th17/citología
5.
Nat Med ; 23(5): 556-567, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28394331

RESUMEN

The progression of pancreatic oncogenesis requires immune-suppressive inflammation in cooperation with oncogenic mutations. However, the drivers of intratumoral immune tolerance are uncertain. Dectin 1 is an innate immune receptor crucial for anti-fungal immunity, but its role in sterile inflammation and oncogenesis has not been well defined. Furthermore, non-pathogen-derived ligands for dectin 1 have not been characterized. We found that dectin 1 is highly expressed on macrophages in pancreatic ductal adenocarcinoma (PDA). Dectin 1 ligation accelerated the progression of PDA in mice, whereas deletion of Clec7a-the gene encoding dectin 1-or blockade of dectin 1 downstream signaling was protective. We found that dectin 1 can ligate the lectin galectin 9 in mouse and human PDA, which results in tolerogenic macrophage programming and adaptive immune suppression. Upon disruption of the dectin 1-galectin 9 axis, CD4+ and CD8+ T cells, which are dispensable for PDA progression in hosts with an intact signaling axis, become reprogrammed into indispensable mediators of anti-tumor immunity. These data suggest that targeting dectin 1 signaling is an attractive strategy for developing an immunotherapy for PDA.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Galectinas/metabolismo , Lectinas Tipo C/genética , Neoplasias Pancreáticas/genética , Escape del Tumor/genética , Animales , Western Blotting , Carcinogénesis/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/metabolismo , Células Epiteliales/metabolismo , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Humanos , Tolerancia Inmunológica/genética , Inmunohistoquímica , Inmunoprecipitación , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Espectrometría de Masas , Ratones , Ratones Noqueados , Conductos Pancreáticos/citología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Quinasa Syk/genética , Quinasa Syk/metabolismo , Escape del Tumor/inmunología
6.
J Immunol ; 197(7): 2816-27, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27559045

RESUMEN

Con A hepatitis is regarded as a T cell-mediated model of acute liver injury. Mincle is a C-type lectin receptor that is critical in the immune response to mycobacteria and fungi but does not have a well-defined role in preclinical models of non-pathogen-mediated inflammation. Because Mincle can ligate the cell death ligand SAP130, we postulated that Mincle signaling drives intrahepatic inflammation and liver injury in Con A hepatitis. Acute liver injury was assessed in the murine Con A hepatitis model using C57BL/6, Mincle(-/-), and Dectin-1(-/-) mice. The role of C/EBPß and hypoxia-inducible factor-1α (HIF-1α) signaling was assessed using selective inhibitors. We found that Mincle was highly expressed in hepatic innate inflammatory cells and endothelial cells in both mice and humans. Furthermore, sterile Mincle ligands and Mincle signaling intermediates were increased in the murine liver in Con A hepatitis. Most significantly, Mincle deletion or blockade protected against Con A hepatitis, whereas Mincle ligation exacerbated disease. Bone marrow chimeric and adoptive transfer experiments suggested that Mincle signaling in infiltrating myeloid cells dictates disease phenotype. Conversely, signaling via other C-type lectin receptors did not alter disease course. Mechanistically, we found that Mincle blockade decreased the NF-κß-related signaling intermediates C/EBPß and HIF-1α, both of which are necessary in macrophage-mediated inflammatory responses. Accordingly, Mincle deletion lowered production of nitrites in Con A hepatitis and inhibition of both C/EBPß and HIF-1α reduced the severity of liver disease. Our work implicates a novel innate immune driver of Con A hepatitis and, more broadly, suggests a potential role for Mincle in diseases governed by sterile inflammation.


Asunto(s)
Concanavalina A/inmunología , Hepatitis/inmunología , Lectinas Tipo C/inmunología , Proteínas de la Membrana/inmunología , Transducción de Señal/inmunología , Animales , Modelos Animales de Enfermedad , Hepatitis/metabolismo , Humanos , Inflamación/inmunología , Lectinas Tipo C/deficiencia , Leucocitos Mononucleares , Masculino , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Nitritos/metabolismo
7.
Nature ; 532(7598): 245-9, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-27049944

RESUMEN

Neoplastic pancreatic epithelial cells are believed to die through caspase 8-dependent apoptotic cell death, and chemotherapy is thought to promote tumour apoptosis. Conversely, cancer cells often disrupt apoptosis to survive. Another type of programmed cell death is necroptosis (programmed necrosis), but its role in pancreatic ductal adenocarcinoma (PDA) is unclear. There are many potential inducers of necroptosis in PDA, including ligation of tumour necrosis factor receptor 1 (TNFR1), CD95, TNF-related apoptosis-inducing ligand (TRAIL) receptors, Toll-like receptors, reactive oxygen species, and chemotherapeutic drugs. Here we report that the principal components of the necrosome, receptor-interacting protein (RIP)1 and RIP3, are highly expressed in PDA and are further upregulated by the chemotherapy drug gemcitabine. Blockade of the necrosome in vitro promoted cancer cell proliferation and induced an aggressive oncogenic phenotype. By contrast, in vivo deletion of RIP3 or inhibition of RIP1 protected against oncogenic progression in mice and was associated with the development of a highly immunogenic myeloid and T cell infiltrate. The immune-suppressive tumour microenvironment associated with intact RIP1/RIP3 signalling depended in part on necroptosis-induced expression of the chemokine attractant CXCL1, and CXCL1 blockade protected against PDA. Moreover, cytoplasmic SAP130 (a subunit of the histone deacetylase complex) was expressed in PDA in a RIP1/RIP3-dependent manner, and Mincle--its cognate receptor--was upregulated in tumour-infiltrating myeloid cells. Ligation of Mincle by SAP130 promoted oncogenesis, whereas deletion of Mincle protected against oncogenesis and phenocopied the immunogenic reprogramming of the tumour microenvironment that was induced by RIP3 deletion. Cellular depletion suggested that whereas inhibitory macrophages promote tumorigenesis in PDA, they lose their immune-suppressive effects when RIP3 or Mincle is deleted. Accordingly, T cells, which are not protective against PDA progression in mice with intact RIP3 or Mincle signalling, are reprogrammed into indispensable mediators of anti-tumour immunity in the absence of RIP3 or Mincle. Our work describes parallel networks of necroptosis-induced CXCL1 and Mincle signalling that promote macrophage-induced adaptive immune suppression and thereby enable PDA progression.


Asunto(s)
Carcinogénesis , Quimiocina CXCL1/metabolismo , Tolerancia Inmunológica , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/metabolismo , Necrosis , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Adenocarcinoma/inmunología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Apoptosis/efectos de los fármacos , Carcinogénesis/efectos de los fármacos , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL1/antagonistas & inhibidores , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Progresión de la Enfermedad , Femenino , Proteínas Activadoras de GTPasa/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Lectinas Tipo C/inmunología , Masculino , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Regulación hacia Arriba , Gemcitabina
8.
J Leukoc Biol ; 100(1): 185-94, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26747838

RESUMEN

Regulation of Toll-like receptor responses is critical for limiting tissue injury and autoimmunity in both sepsis and sterile inflammation. We found that Mincle, a C-type lectin receptor, regulates proinflammatory Toll-like receptor 4 signaling. Specifically, Mincle ligation diminishes Toll-like receptor 4-mediated inflammation, whereas Mincle deletion or knockdown results in marked hyperresponsiveness to lipopolysaccharide in vitro, as well as overwhelming lipopolysaccharide-mediated inflammation in vivo. Mechanistically, Mincle deletion does not up-regulate Toll-like receptor 4 expression or reduce interleukin 10 production after Toll-like receptor 4 ligation; however, Mincle deletion decreases production of the p38 mitogen-activated protein kinase-dependent inhibitory intermediate suppressor of cytokine signaling 1, A20, and ABIN3 and increases expression of the Toll-like receptor 4 coreceptor CD14. Blockade of CD14 mitigates the increased sensitivity of Mincle(-/-) leukocytes to Toll-like receptor 4 ligation. Collectively, we describe a major role for Mincle in suppressing Toll-like receptor 4 responses and implicate its importance in nonmycobacterial models of inflammation.


Asunto(s)
Inflamación/etiología , Lectinas Tipo C/deficiencia , Proteínas de la Membrana/deficiencia , Bazo/inmunología , Receptor Toll-Like 4/metabolismo , Animales , Células Cultivadas , Inflamación/metabolismo , Inflamación/patología , Lectinas Tipo C/genética , Lipopolisacáridos/farmacología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Bazo/efectos de los fármacos , Bazo/metabolismo , Receptor Toll-Like 4/antagonistas & inhibidores
9.
Cell Rep ; 13(9): 1909-1921, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26655905

RESUMEN

Dectin-1 is a C-type lectin receptor critical in anti-fungal immunity, but Dectin-1 has not been linked to regulation of sterile inflammation or oncogenesis. We found that Dectin-1 expression is upregulated in hepatic fibrosis and liver cancer. However, Dectin-1 deletion exacerbates liver fibro-inflammatory disease and accelerates hepatocarcinogenesis. Mechanistically, we found that Dectin-1 protects against chronic liver disease by suppressing TLR4 signaling in hepatic inflammatory and stellate cells. Accordingly, Dectin-1(-/-) mice exhibited augmented cytokine production and reduced survival in lipopolysaccharide (LPS)-mediated sepsis, whereas Dectin-1 activation was protective. We showed that Dectin-1 inhibits TLR4 signaling by mitigating TLR4 and CD14 expression, which are regulated by Dectin-1-dependent macrophage colony stimulating factor (M-CSF) expression. Our study suggests that Dectin-1 is an attractive target for experimental therapeutics in hepatic fibrosis and neoplastic transformation. More broadly, our work deciphers critical cross-talk between pattern recognition receptors and implicates a role for Dectin-1 in suppression of sterile inflammation, inflammation-induced oncogenesis, and LPS-mediated sepsis.


Asunto(s)
Lectinas Tipo C/metabolismo , Cirrosis Hepática/patología , Neoplasias Hepáticas/patología , Receptor Toll-Like 4/metabolismo , Animales , Transformación Celular Neoplásica/efectos de los fármacos , Células Cultivadas , Quimiocina CCL2/sangre , Citocinas/metabolismo , Dietilnitrosamina/toxicidad , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Inflamación , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/toxicidad , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/metabolismo , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/metabolismo , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Sepsis/etiología , Transducción de Señal/efectos de los fármacos , Tioacetamida/toxicidad , Receptor Toll-Like 4/antagonistas & inhibidores , Regulación hacia Arriba/efectos de los fármacos
10.
PLoS One ; 10(7): e0132786, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26172047

RESUMEN

Cancer cachexia is a debilitating condition characterized by a combination of anorexia, muscle wasting, weight loss, and malnutrition. This condition affects an overwhelming majority of patients with pancreatic cancer and is a primary cause of cancer-related death. However, few, if any, effective therapies exist for both treatment and prevention of this syndrome. In order to develop novel therapeutic strategies for pancreatic cancer cachexia, appropriate animal models are necessary. In this study, we developed and validated a syngeneic, metastatic, murine model of pancreatic cancer cachexia. Using our model, we investigated the ability of transforming growth factor beta (TGF-ß) blockade to mitigate the metabolic changes associated with cachexia. We found that TGF-ß inhibition using the anti-TGF-ß antibody 1D11.16.8 significantly improved overall mortality, weight loss, fat mass, lean body mass, bone mineral density, and skeletal muscle proteolysis in mice harboring advanced pancreatic cancer. Other immunotherapeutic strategies we employed were not effective. Collectively, we validated a simplified but useful model of pancreatic cancer cachexia to investigate immunologic treatment strategies. In addition, we showed that TGF-ß inhibition can decrease the metabolic changes associated with cancer cachexia and improve overall survival.


Asunto(s)
Caquexia/metabolismo , Caquexia/mortalidad , Modelos Animales de Enfermedad , Inmunoterapia , Neoplasias Pancreáticas/complicaciones , Factor de Crecimiento Transformador beta/inmunología , Animales , Anticuerpos/inmunología , Anticuerpos/uso terapéutico , Composición Corporal , Caquexia/complicaciones , Caquexia/terapia , Línea Celular Tumoral , Masculino , Ratones , Ratones Endogámicos C57BL , Atrofia Muscular/complicaciones , Metástasis de la Neoplasia , Neoplasias Pancreáticas/patología , Análisis de Supervivencia
11.
Gastroenterology ; 147(2): 473-84.e2, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24801349

RESUMEN

BACKGROUND & AIMS: Subsets of leukocytes synergize with regenerative growth factors to promote hepatic regeneration. γδT cells are early responders to inflammation-induced injury in a number of contexts. We investigated the role of γδT cells in hepatic regeneration using mice with disruptions in Tcrd (encodes the T-cell receptor δ chain) and Clec7a (encodes C-type lectin domain family 7 member a, also known as DECTIN1). METHODS: We performed partial hepatectomies on wild-type C57BL/6, CD45.1, Tcrd(-/-), or Clec7a(-/-) mice. Cells were isolated from livers of patients and mice via mechanical and enzymatic digestion. γδT cells were purified by fluorescence-activated cell sorting. RESULTS: In mice, partial hepatectomy up-regulated expression of CCL20 and ligands of Dectin-1, which was associated with recruitment and activation of γδT cells and their increased production of interleukin (IL)-17 family cytokines. Recruited γδT cells induced production of IL-6 by antigen-presenting cells and suppressed expression of interferon gamma by natural killer T cells, promoting hepatocyte proliferation. Absence of IL-17-producing γδT cells or deletion of Dectin-1 prevented development of regenerative phenotypes in subsets of innate immune cells. This slowed liver regeneration and was associated with reduced expression of regenerative growth factors and cell cycle regulators. Conversely, exogenous administration of IL-17 family cytokines or Dectin-1 ligands promoted regeneration. More broadly, we found that γδT cells are required for inflammatory responses mediated by IL-17 and Dectin-1. CONCLUSIONS: γδT cells regulate hepatic regeneration by producing IL-22 and IL-17, which have direct mitogenic effects on hepatocytes and promote a regenerative phenotype in hepatic leukocytes, respectively. Dectin-1 ligation is required for γδT cells to promote hepatic regeneration.


Asunto(s)
Proliferación Celular , Hepatocitos/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-17/metabolismo , Regeneración Hepática , Hígado/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/metabolismo , Animales , Células Cultivadas , Quimiocina CCL20/metabolismo , Genotipo , Hepatectomía , Hepatocitos/inmunología , Humanos , Interferón gamma/metabolismo , Interleucina-6/metabolismo , Interleucinas/metabolismo , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Hígado/inmunología , Hígado/cirugía , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Receptores de Antígenos de Linfocitos T gamma-delta/deficiencia , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Transducción de Señal , Linfocitos T/inmunología , Factores de Tiempo , Interleucina-22
13.
J Immunol ; 190(9): 4640-9, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23536633

RESUMEN

Dendritic cells (DC) are professional APCs that regulate innate and adaptive immunity. The role of fatty-acid synthesis in DC development and function is uncertain. We found that blockade of fatty-acid synthesis markedly decreases dendropoiesis in the liver and in primary and secondary lymphoid organs in mice. Human DC development from PBMC precursors was also diminished by blockade of fatty-acid synthesis. This was associated with higher rates of apoptosis in precursor cells and increased expression of cleaved caspase-3 and BCL-xL and downregulation of cyclin B1. Further, blockade of fatty-acid synthesis decreased DC expression of MHC class II, ICAM-1, B7-1, and B7-2 but increased their production of selected proinflammatory cytokines including IL-12 and MCP-1. Accordingly, inhibition of fatty-acid synthesis enhanced DC capacity to activate allogeneic as well as Ag-restricted CD4(+) and CD8(+) T cells and induce CTL responses. Further, blockade of fatty-acid synthesis increased DC expression of Notch ligands and enhanced their ability to activate NK cell immune phenotype and IFN-γ production. Because endoplasmic reticulum (ER) stress can augment the immunogenic function of APC, we postulated that this may account for the higher DC immunogenicity. We found that inhibition of fatty-acid synthesis resulted in elevated expression of numerous markers of ER stress in humans and mice and was associated with increased MAPK and Akt signaling. Further, lowering ER stress by 4-phenylbutyrate mitigated the enhanced immune stimulation associated with fatty-acid synthesis blockade. Our findings elucidate the role of fatty-acid synthesis in DC development and function and have implications to the design of DC vaccines for immunotherapy.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Ácidos Grasos/biosíntesis , Animales , Apoptosis/inmunología , Antígeno B7-1/inmunología , Antígeno B7-1/metabolismo , Antígeno B7-2/inmunología , Antígeno B7-2/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Caspasa 3/inmunología , Caspasa 3/metabolismo , Quimiocina CCL2/inmunología , Quimiocina CCL2/metabolismo , Ciclina B1/inmunología , Ciclina B1/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/metabolismo , Ácidos Grasos/inmunología , Ácidos Grasos/metabolismo , Genes MHC Clase II/inmunología , Humanos , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-12/inmunología , Interleucina-12/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Hígado/inmunología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Quinasas de Proteína Quinasa Activadas por Mitógenos/inmunología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , PPAR gamma/inmunología , PPAR gamma/metabolismo , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Proteína bcl-X/inmunología , Proteína bcl-X/metabolismo
14.
Hepatology ; 58(2): 589-602, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23322710

RESUMEN

UNLABELLED: Nonalcoholic steatohepatitis (NASH) is the most common etiology of chronic liver dysfunction in the United States and can progress to cirrhosis and liver failure. Inflammatory insult resulting from fatty infiltration of the liver is central to disease pathogenesis. Dendritic cells (DCs) are antigen-presenting cells with an emerging role in hepatic inflammation. We postulated that DCs are important in the progression of NASH. We found that intrahepatic DCs expand and mature in NASH liver and assume an activated immune phenotype. However, rather than mitigating the severity of NASH, DC depletion markedly exacerbated intrahepatic fibroinflammation. Our mechanistic studies support a regulatory role for DCs in NASH by limiting sterile inflammation through their role in the clearance of apoptotic cells and necrotic debris. We found that DCs limit CD8(+) T-cell expansion and restrict Toll-like receptor expression and cytokine production in innate immune effector cells in NASH, including Kupffer cells, neutrophils, and inflammatory monocytes. Consistent with their regulatory role in NASH, during the recovery phase of disease, ablation of DC populations results in delayed resolution of intrahepatic inflammation and fibroplasia. CONCLUSION: Our findings support a role for DCs in modulating NASH. Targeting DC functional properties may hold promise for therapeutic intervention in NASH.


Asunto(s)
Comunicación Celular/fisiología , Células Dendríticas/fisiología , Progresión de la Enfermedad , Hígado Graso/fisiopatología , Hígado/fisiopatología , Animales , Apoptosis/fisiología , Linfocitos T CD8-positivos/patología , Células Cultivadas , Células Dendríticas/patología , Modelos Animales de Enfermedad , Hígado Graso/patología , Macrófagos del Hígado/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Necrosis/fisiopatología , Neutrófilos/patología , Receptores Toll-Like/fisiología
15.
J Clin Invest ; 122(11): 4118-29, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23023703

RESUMEN

Pancreatic ductal adenocarcinoma is an aggressive cancer that interacts with stromal cells to produce a highly inflammatory tumor microenvironment that promotes tumor growth and invasiveness. The precise interplay between tumor and stroma remains poorly understood. TLRs mediate interactions between environmental stimuli and innate immunity and trigger proinflammatory signaling cascades. Our finding that TLR7 expression is upregulated in both epithelial and stromal compartments in human and murine pancreatic cancer led us to postulate that carcinogenesis is dependent on TLR7 signaling. In a mouse model of pancreatic cancer, TLR7 ligation vigorously accelerated tumor progression and induced loss of expression of PTEN, p16, and cyclin D1 and upregulation of p21, p27, p53, c-Myc, SHPTP1, TGF-ß, PPARγ, and cyclin B1. Furthermore, TLR7 ligation induced STAT3 activation and interfaced with Notch as well as canonical NF-κB and MAP kinase pathways, but downregulated expression of Notch target genes. Moreover, blockade of TLR7 protected against carcinogenesis. Since pancreatic tumorigenesis requires stromal expansion, we proposed that TLR7 ligation modulates pancreatic cancer by driving stromal inflammation. Accordingly, we found that mice lacking TLR7 exclusively within their inflammatory cells were protected from neoplasia. These data suggest that targeting TLR7 holds promise for treatment of human pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Transformación Celular Neoplásica/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptor Toll-Like 7/metabolismo , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/patología , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inmunidad Innata/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Mutantes , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/inmunología
16.
J Exp Med ; 209(9): 1671-87, 2012 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-22908323

RESUMEN

The transition of chronic pancreatic fibroinflammatory disease to neoplasia is a primary example of the paradigm linking inflammation to carcinogenesis. However, the cellular and molecular mediators bridging these entities are not well understood. Because TLR4 ligation can exacerbate pancreatic inflammation, we postulated that TLR4 activation drives pancreatic carcinogenesis. In this study, we show that lipopolysaccharide accelerates pancreatic tumorigenesis, whereas TLR4 inhibition is protective. Furthermore, blockade of the MyD88-independent TRIF pathway is protective against pancreatic cancer, whereas blockade of the MyD88-dependent pathway surprisingly exacerbates pancreatic inflammation and malignant progression. The protumorigenic and fibroinflammatory effects of MyD88 inhibition are mediated by dendritic cells (DCs), which induce pancreatic antigen-restricted Th2-deviated CD4(+) T cells and promote the transition from pancreatitis to carcinoma. Our data implicate a primary role for DCs in pancreatic carcinogenesis and illustrate divergent pathways in which blockade of TLR4 signaling via TRIF is protective against pancreatic cancer and, conversely, MyD88 inhibition exacerbates pancreatic inflammation and neoplastic transformation by augmenting the DC-Th2 axis.


Asunto(s)
Adenocarcinoma/patología , Células Dendríticas/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Neoplasias Pancreáticas/patología , Células Th2/patología , Receptor Toll-Like 4/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Adenocarcinoma/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Células Dendríticas/inmunología , Humanos , Masculino , Ratones , Ratones Mutantes , Factor 88 de Diferenciación Mieloide/genética , Neoplasias Pancreáticas/metabolismo , Pancreatitis Crónica/metabolismo , Pancreatitis Crónica/patología , Células Th2/inmunología
17.
Gastroenterology ; 143(4): 1061-72, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22705178

RESUMEN

BACKGROUND & AIMS: Immune cells of the liver must be able to recognize and react to pathogens yet remain tolerant to food molecules and other nonpathogens. Dendritic cells (DCs) are believed to contribute to hepatic tolerance. Lipids have been implicated in dysfunction of DCs in cancer. Therefore, we investigated whether high lipid content in liver DCs affects induction of tolerance. METHODS: Mouse and human hepatic nonparenchymal cells were isolated by mechanical and enzymatic digestion. DCs were purified by fluorescence-activated cell sorting or with immunomagnetic beads. DC lipid content was assessed by flow cytometry, immune fluorescence, and electron microscopy and by measuring intracellular component lipids. DC activation was determined from surface phenotype and cytokine profile. DC function was assessed in T-cell, natural killer (NK) cell, and NKT cell coculture assays as well as in vivo. RESULTS: We observed 2 distinct populations of hepatic DCs in mice and humans based on their lipid content and expression of markers associated with adipogenesis and lipid metabolism. This lipid-based dichotomy in DCs was unique to the liver and specific to DCs compared with other hepatic immune cells. However, rather than mediate tolerance, the liver DC population with high concentrations of lipid was immunogenic in multiple models; they activated T cells, NK cells, and NKT cells. Conversely, liver DCs with low levels of lipid induced regulatory T cells, anergy to cancer, and oral tolerance. The immunogenicity of lipid-rich liver DCs required their secretion of tumor necrosis factor α and was directly related to their high lipid content; blocking DC synthesis of fatty acids or inhibiting adipogenesis (by reducing endoplasmic reticular stress) reduced DC immunogenicity. CONCLUSIONS: Human and mouse hepatic DCs are composed of distinct populations that contain different concentrations of lipid, which regulates immunogenic versus tolerogenic responses in the liver.


Asunto(s)
Antígenos CD/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Lípidos/análisis , Hígado/inmunología , Hígado/metabolismo , Adipogénesis , Animales , Antígenos CD1d/metabolismo , Apoptosis , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Antígeno CD11b/metabolismo , Antígenos CD40/metabolismo , Células Cultivadas , Células Dendríticas/química , Humanos , Tolerancia Inmunológica , Molécula 1 de Adhesión Intercelular/metabolismo , Células Asesinas Naturales/fisiología , Antígenos Comunes de Leucocito/metabolismo , Metabolismo de los Lípidos , Hígado/química , Activación de Linfocitos , Ratones , Células T Asesinas Naturales/fisiología , Fenotipo , Linfocitos T/fisiología , Linfocitos T Reguladores/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
18.
Vaccine ; 28(21): 3688-95, 2010 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-20359561

RESUMEN

A large number of infectious diseases caused by viral or bacterial infections are treatable and/or preventable by vaccination. In addition, ongoing research is aimed at the development of vaccines against other types of diseases, including almost all forms of cancer. The efficacy of a vaccine relies on the antigen-specific response by the entire repertoire of immune competent cells. Here, we have generated a powerful mitogen fusion protein, CD40L-FasL-IgFc, which stimulates CD40(+) cells robustly. We found that this specific cell activation is accompanied by increased expression of PRDI-BF1 (Blim-1) RNA, an indicator of terminal B-cell differentiation, in cultures stimulated with CD40L-FasL-IgFc. The addition of specific inhibitors of NF-kappaB and MEK1/2 partially suppressed the observed proliferative effects of CD40L-FasL-IgFc. When tested in vivo, the immune response to influenza HA vaccine was significantly increased by co-administration of CD40L-FasL-IgFc. Moreover, the co-administration of the cDNA expression plasmid encoding CD40L-FasL-IgFc significantly boosted the vaccine response. We now have a unique opportunity to evaluate our novel fusion protein adjuvant, and other similarly constructed fusion proteins, in both protein-based and genetic vaccines.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Ligando de CD40/inmunología , Proteína Ligando Fas/inmunología , Activación de Linfocitos , Mitógenos/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Antígenos CD40/metabolismo , Línea Celular , Proliferación Celular , Humanos , Vacunas contra la Influenza/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Recombinantes de Fusión/inmunología , Proteínas Represoras/metabolismo
19.
Dev Comp Immunol ; 32(8): 890-7, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18262264

RESUMEN

Ferrets (Mustela putorius furo) develop symptoms upon influenza infection that resemble those of humans, including sneezing, body temperature variation and weight loss. Highly pathogenic strains of influenza A, such as H5N1, have the capacity to cause severe illness or death in ferrets. The use of ferrets as a model of influenza infection is currently limited by a lack of species-specific immunological reagents. Interferon gamma (IFN-gamma) plays a key role in the development of innate and adaptive immunity and the regulation of Th1-type immune responses. Here we describe the cloning of the full-length cDNA for ferret IFN-gamma. Multiple sequence alignment of the predicted amino acid sequence with those of other species indicates that the predicted ferret protein shares the highest identity with Eurasian badger IFN-gamma. We raised two hybridoma clones expressing monoclonal antibodies against recombinant ferret IFN-gamma capable of detecting IFN-gamma protein derived from mitogen-stimulated ferret PBMCs by immunoblotting, ELISA and ELISPOT assay. Finally, an ELISA utilizing the ferret-specific antibodies detected elevated levels of IFN-gamma in serum samples from H3N2 influenza A-infected ferrets.


Asunto(s)
Hurones/inmunología , Interferón gamma/análisis , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Secuencia de Bases , Células COS , Chlorocebus aethiops , Clonación Molecular , Ensayo de Inmunoadsorción Enzimática , Interferón gamma/genética , Masculino , Datos de Secuencia Molecular , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología
20.
Blood ; 106(6): 2002-10, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15941918

RESUMEN

OX40 and 4-1BB are members of the tumor necrosis factor (TNF) family of costimulatory receptors whose signaling is important for differential immune responses mediated by CD4+ or CD8+ T cells. Although activated T cells may acquire OX40/4-1BB double-positive phenotype and signaling from each receptor is expected to influence cell functions, the relevance between OX40 and 4-1BB has never been investigated before. While we were investigating the expression of OX40 and 4-1BB on activated human T cells, we found that they colocalize. The study of receptor gene-transfected cells showed that both receptors coendocytose and the complex of OX40 and 4-1BB was detected by specific ligands or antibodies (Abs). The heterodimer of OX40 and 4-1BB was identified by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) under nonreduced conditions and was associated with the tumor receptor-associated factor (TRAF) family proteins in a unique manner. Furthermore, the stimulation of OX40/4-1BB rendered cells sensitive to apoptosis induced by TNF-alpha that accompanied reduced activation of nuclear factor-kappaB (NF-kappaB). Finally, the OX40/4-1BB stimulation repressed the mitogen response in activated CD25+CD4+ T cells and preactivated CD8+ T cells. Thus, the OX40/4-1BB heterodimer appears to represent a unique regulatory receptor in activated T cells.


Asunto(s)
Antígenos CD/análisis , Activación de Linfocitos , Receptores de Factor de Crecimiento Nervioso/análisis , Receptores del Factor de Necrosis Tumoral/análisis , Linfocitos T/química , Antígenos CD/metabolismo , Antígenos CD/fisiología , Apoptosis , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Dimerización , Endocitosis , Humanos , FN-kappa B/metabolismo , Unión Proteica , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores de Factor de Crecimiento Nervioso/fisiología , Receptores OX40 , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores del Factor de Necrosis Tumoral/fisiología , Linfocitos T/inmunología , Transfección , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...