Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nature ; 532(7598): 245-9, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27049944

ABSTRACT

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.


Subject(s)
Carcinogenesis , Chemokine CXCL1/metabolism , Immune Tolerance , Lectins, C-Type/metabolism , Membrane Proteins/metabolism , Necrosis , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Carcinogenesis/drug effects , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL1/antagonists & inhibitors , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Progression , Female , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lectins, C-Type/immunology , Male , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Up-Regulation , Gemcitabine
3.
J Immunol ; 197(7): 2816-27, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27559045

ABSTRACT

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.


Subject(s)
Concanavalin A/immunology , Hepatitis/immunology , Lectins, C-Type/immunology , Membrane Proteins/immunology , Signal Transduction/immunology , Animals , Disease Models, Animal , Hepatitis/metabolism , Humans , Inflammation/immunology , Lectins, C-Type/deficiency , Leukocytes, Mononuclear , Male , Membrane Proteins/deficiency , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nitrites/metabolism
4.
Gastroenterology ; 147(2): 473-84.e2, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24801349

ABSTRACT

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.


Subject(s)
Cell Proliferation , Hepatocytes/metabolism , Inflammation Mediators/metabolism , Interleukin-17/metabolism , Liver Regeneration , Liver/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/metabolism , Animals , Cells, Cultured , Chemokine CCL20/metabolism , Genotype , Hepatectomy , Hepatocytes/immunology , Humans , Interferon-gamma/metabolism , Interleukin-6/metabolism , Interleukins/metabolism , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Liver/immunology , Liver/surgery , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Antigen, T-Cell, gamma-delta/deficiency , Receptors, Antigen, T-Cell, gamma-delta/genetics , Signal Transduction , T-Lymphocytes/immunology , Time Factors , Interleukin-22
5.
J Immunol ; 190(9): 4640-9, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23536633

ABSTRACT

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.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Fatty Acids/biosynthesis , Animals , Apoptosis/immunology , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Caspase 3/immunology , Caspase 3/metabolism , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Cyclin B1/immunology , Cyclin B1/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Fatty Acids/immunology , Fatty Acids/metabolism , Genes, MHC Class II/immunology , Humans , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-12/immunology , Interleukin-12/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Liver/immunology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/immunology , Mitogen-Activated Protein Kinase Kinases/metabolism , PPAR gamma/immunology , PPAR gamma/metabolism , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , bcl-X Protein/immunology , bcl-X Protein/metabolism
6.
Hepatology ; 58(2): 589-602, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23322710

ABSTRACT

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.


Subject(s)
Cell Communication/physiology , Dendritic Cells/physiology , Disease Progression , Fatty Liver/physiopathology , Liver/physiopathology , Animals , Apoptosis/physiology , CD8-Positive T-Lymphocytes/pathology , Cells, Cultured , Dendritic Cells/pathology , Disease Models, Animal , Fatty Liver/pathology , Kupffer Cells/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Necrosis/physiopathology , Neutrophils/pathology , Toll-Like Receptors/physiology
7.
Gastroenterology ; 143(4): 1061-72, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22705178

ABSTRACT

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.


Subject(s)
Antigens, CD/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lipids/analysis , Liver/immunology , Liver/metabolism , Adipogenesis , Animals , Antigens, CD1d/metabolism , Apoptosis , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , CD11b Antigen/metabolism , CD40 Antigens/metabolism , Cells, Cultured , Dendritic Cells/chemistry , Humans , Immune Tolerance , Intercellular Adhesion Molecule-1/metabolism , Killer Cells, Natural/physiology , Leukocyte Common Antigens/metabolism , Lipid Metabolism , Liver/chemistry , Lymphocyte Activation , Mice , Natural Killer T-Cells/physiology , Phenotype , T-Lymphocytes/physiology , T-Lymphocytes, Regulatory/physiology , Tumor Necrosis Factor-alpha/metabolism
8.
J Exp Med ; 199(7): 1025-31, 2004 Apr 05.
Article in English | MEDLINE | ID: mdl-15067037

ABSTRACT

The role of CD40 ligand (CD40L)/CD40 signaling in T cell-dependent B cell differentiation and maturation has been amply documented. The mechanism of CD40 signaling in B cells has been well established, whereas the signaling mechanism of CD40L in T cell costimulation remains unknown. In this study we show that CD28i, a transmembrane splice variant of CD28 costimulatory receptor, complexes with CD40L in human T cells. The cross-linking of CD40L resulted in the coendocytosis of CD28i with CD40L. The tyrosine phosphorylation of CD28i followed the cross-linking of CD40L, and the overexpression of CD28i augmented the c-Jun NH2-terminal kinase, p21-activated kinase 2, and nuclear factor kappaB activation. These data indicate that CD28i, by functioning as a signaling adaptor, transduces CD40L signaling as well as CD28 signaling in human T cells.


Subject(s)
CD28 Antigens/genetics , CD40 Ligand/metabolism , T-Lymphocytes/immunology , Alternative Splicing , CD28 Antigens/chemistry , CD28 Antigens/metabolism , CD40 Ligand/chemistry , Cell Line , Cross-Linking Reagents , Endocytosis , Humans , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , T-Lymphocytes/metabolism , p21-Activated Kinases
9.
Dev Comp Immunol ; 32(8): 890-7, 2008.
Article in English | MEDLINE | ID: mdl-18262264

ABSTRACT

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.


Subject(s)
Ferrets/immunology , Interferon-gamma/analysis , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Base Sequence , COS Cells , Chlorocebus aethiops , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Interferon-gamma/genetics , Male , Molecular Sequence Data , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology
10.
Cancer Cell ; 34(5): 757-774.e7, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30423296

ABSTRACT

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.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Immune Tolerance/immunology , Macrophages/immunology , Pancreatic Neoplasms/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Humans , Immune Tolerance/genetics , L Cells , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , STAT1 Transcription Factor/metabolism , Th1 Cells/cytology , Th17 Cells/cytology
11.
Nat Med ; 23(5): 556-567, 2017 May.
Article in English | MEDLINE | ID: mdl-28394331

ABSTRACT

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.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Galectins/metabolism , Lectins, C-Type/genetics , Pancreatic Neoplasms/genetics , Tumor Escape/genetics , Animals , Blotting, Western , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Epithelial Cells/metabolism , Flow Cytometry , Gene Knockdown Techniques , Humans , Immune Tolerance/genetics , Immunohistochemistry , Immunoprecipitation , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Mass Spectrometry , Mice , Mice, Knockout , Pancreatic Ducts/cytology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Syk Kinase/genetics , Syk Kinase/metabolism , Tumor Escape/immunology
12.
J Leukoc Biol ; 100(1): 185-94, 2016 07.
Article in English | MEDLINE | ID: mdl-26747838

ABSTRACT

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.


Subject(s)
Inflammation/etiology , Lectins, C-Type/deficiency , Membrane Proteins/deficiency , Spleen/immunology , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Inflammation/metabolism , Inflammation/pathology , Lectins, C-Type/genetics , Lipopolysaccharides/pharmacology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/drug effects , Spleen/drug effects , Spleen/metabolism , Toll-Like Receptor 4/antagonists & inhibitors
14.
Biochem J ; 378(Pt 2): 539-47, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14594451

ABSTRACT

VCAM-1 (vascular cell adhesion molecule-1) plays an important role in the regulation of inflammation in atherosclerosis, asthma, inflammatory bowel disease and transplantation. VCAM-1 activates endothelial cell NADPH oxidase, and this oxidase activity is required for VCAM-1-dependent lymphocyte migration. We reported previously that a mouse microvascular endothelial cell line promotes lymphocyte migration that is dependent on VCAM-1, but not on other known adhesion molecules. Here we have investigated the signalling mechanisms underlying VCAM-1 function. Lymphocyte binding to VCAM-1 on the endothelial cell surface activated an endothelial cell calcium flux that could be inhibited with anti-alpha4-integrin and mimicked by anti-VCAM-1-coated beads. VCAM-1 stimulation of calcium responses could be blocked by an inhibitor of intracellular calcium mobilization, a calcium channel inhibitor or a calcium chelator, resulting in the inhibition of NADPH oxidase activity. Addition of ionomycin overcame the calcium channel blocker suppression of VCAM-1-stimulated NADPH oxidase activity, but could not reverse the inhibitory effect imposed by intracellular calcium blockage, indicating that both intracellular and extracellular calcium mobilization are required for VCAM-1-mediated activation of NADPH oxidase. Furthermore, VCAM-1 specifically activated the Rho-family GTPase Rac1, and VCAM-1 activation of NADPH oxidase was blocked by a dominant negative Rac1. Thus VCAM-1 stimulates the mobilization of intracellular and extracellular calcium and Rac1 activity that are required for the activation of NADPH oxidase.


Subject(s)
Calcium Signaling , Endothelium, Vascular/metabolism , NADPH Oxidases/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Calcium/metabolism , Calcium Channels/metabolism , Cell Adhesion , Cell Line , Cell Movement , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Enzyme Activation , Lymphocytes/immunology , Mice , Mice, Inbred BALB C
15.
Cell Rep ; 13(9): 1909-1921, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26655905

ABSTRACT

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.


Subject(s)
Lectins, C-Type/metabolism , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Toll-Like Receptor 4/metabolism , Animals , Cell Transformation, Neoplastic/drug effects , Cells, Cultured , Chemokine CCL2/blood , Cytokines/metabolism , Diethylnitrosamine/toxicity , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Inflammation , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/toxicity , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Sepsis/etiology , Signal Transduction/drug effects , Thioacetamide/toxicity , Toll-Like Receptor 4/antagonists & inhibitors , Up-Regulation/drug effects
16.
PLoS One ; 10(7): e0132786, 2015.
Article in English | MEDLINE | ID: mdl-26172047

ABSTRACT

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.


Subject(s)
Cachexia/metabolism , Cachexia/mortality , Disease Models, Animal , Immunotherapy , Pancreatic Neoplasms/complications , Transforming Growth Factor beta/immunology , Animals , Antibodies/immunology , Antibodies/therapeutic use , Body Composition , Cachexia/complications , Cachexia/therapy , Cell Line, Tumor , Male , Mice , Mice, Inbred C57BL , Muscular Atrophy/complications , Neoplasm Metastasis , Pancreatic Neoplasms/pathology , Survival Analysis
18.
J Exp Med ; 209(9): 1671-87, 2012 Aug 27.
Article in English | MEDLINE | ID: mdl-22908323

ABSTRACT

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.


Subject(s)
Adenocarcinoma/pathology , Dendritic Cells/metabolism , Myeloid Differentiation Factor 88/metabolism , Pancreatic Neoplasms/pathology , Th2 Cells/pathology , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Adenocarcinoma/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Dendritic Cells/immunology , Humans , Male , Mice , Mice, Mutant Strains , Myeloid Differentiation Factor 88/genetics , Pancreatic Neoplasms/metabolism , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/pathology , Th2 Cells/immunology
19.
J Clin Invest ; 122(11): 4118-29, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23023703

ABSTRACT

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.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cell Transformation, Neoplastic/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Toll-Like Receptor 7/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunity, Innate/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Mutant Strains , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology
20.
Vaccine ; 28(21): 3688-95, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20359561

ABSTRACT

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.


Subject(s)
Adjuvants, Immunologic/pharmacology , CD40 Ligand/immunology , Fas Ligand Protein/immunology , Lymphocyte Activation , Mitogens/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD40 Antigens/metabolism , Cell Line , Cell Proliferation , Humans , Influenza Vaccines/immunology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Positive Regulatory Domain I-Binding Factor 1 , Recombinant Fusion Proteins/immunology , Repressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL