Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell ; 185(11): 1924-1942.e23, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35525247

ABSTRACT

For many solid malignancies, lymph node (LN) involvement represents a harbinger of distant metastatic disease and, therefore, an important prognostic factor. Beyond its utility as a biomarker, whether and how LN metastasis plays an active role in shaping distant metastasis remains an open question. Here, we develop a syngeneic melanoma mouse model of LN metastasis to investigate how tumors spread to LNs and whether LN colonization influences metastasis to distant tissues. We show that an epigenetically instilled tumor-intrinsic interferon response program confers enhanced LN metastatic potential by enabling the evasion of NK cells and promoting LN colonization. LN metastases resist T cell-mediated cytotoxicity, induce antigen-specific regulatory T cells, and generate tumor-specific immune tolerance that subsequently facilitates distant tumor colonization. These effects extend to human cancers and other murine cancer models, implicating a conserved systemic mechanism by which malignancies spread to distant organs.


Subject(s)
Lymph Nodes , Melanoma , Animals , Immune Tolerance , Immunotherapy , Lymphatic Metastasis/pathology , Melanoma/pathology , Mice
2.
Science ; 365(6448)2019 07 05.
Article in English | MEDLINE | ID: mdl-31273097

ABSTRACT

Multiple cytosolic innate sensors form large signalosomes after activation, but this assembly needs to be tightly regulated to avoid accumulation of misfolded aggregates. We found that the eIF2α kinase heme-regulated inhibitor (HRI) controls NOD1 signalosome folding and activation through a process requiring eukaryotic initiation factor 2α (eIF2α), the transcription factor ATF4, and the heat shock protein HSPB8. The HRI/eIF2α signaling axis was also essential for signaling downstream of the innate immune mediators NOD2, MAVS, and TRIF but dispensable for pathways dependent on MyD88 or STING. Moreover, filament-forming α-synuclein activated HRI-dependent responses, which suggests that the HRI pathway may restrict toxic oligomer formation. We propose that HRI, eIF2α, and HSPB8 define a novel cytosolic unfolded protein response (cUPR) essential for optimal innate immune signaling by large molecular platforms, functionally homologous to the PERK/eIF2α/HSPA5 axis of the endoplasmic reticulum UPR.


Subject(s)
Cytosol/enzymology , Cytosol/immunology , Immunity, Innate , Protein Serine-Threonine Kinases/physiology , Unfolded Protein Response/immunology , Activating Transcription Factor 4/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Line , Endoplasmic Reticulum Chaperone BiP , Eukaryotic Initiation Factor-2/metabolism , Fibroblasts , Heat-Shock Proteins/metabolism , Humans , Listeria/immunology , Membrane Proteins/metabolism , Mice , Mice, Mutant Strains , Molecular Chaperones/metabolism , Myeloid Differentiation Factor 88/metabolism , Nod1 Signaling Adaptor Protein/chemistry , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Protein Serine-Threonine Kinases/genetics , Salmonella/immunology , Salmonella Infections , Shigella/immunology , Signal Transduction
3.
Nature ; 567(7746): 56-60, 2019 03.
Article in English | MEDLINE | ID: mdl-30814731

ABSTRACT

The cytokine interferon-γ (IFNγ) is a central coordinator of innate and adaptive immunity, but its highly pleiotropic actions have diminished its prospects for use as an immunotherapeutic agent. Here, we took a structure-based approach to decoupling IFNγ pleiotropy. We engineered an affinity-enhanced variant of the ligand-binding chain of the IFNγ receptor IFNγR1, which enabled us to determine the crystal structure of the complete hexameric (2:2:2) IFNγ-IFNγR1-IFNγR2 signalling complex at 3.25 Å resolution. The structure reveals the mechanism underlying deficits in IFNγ responsiveness in mycobacterial disease syndrome resulting from a T168N mutation in IFNγR2, which impairs assembly of the full signalling complex. The topology of the hexameric complex offers a blueprint for engineering IFNγ variants to tune IFNγ receptor signalling output. Unexpectedly, we found that several partial IFNγ agonists exhibited biased gene-expression profiles. These biased agonists retained the ability to induce upregulation of major histocompatibility complex class I antigen expression, but exhibited impaired induction of programmed death-ligand 1 expression in a wide range of human cancer cell lines, offering a route to decoupling immunostimulatory and immunosuppressive functions of IFNγ for therapeutic applications.


Subject(s)
Drug Design , Interferon-gamma/agonists , Interferon-gamma/immunology , Receptors, Interferon/chemistry , Receptors, Interferon/metabolism , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , Cell Line, Tumor , Drug Partial Agonism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class I/genetics , Humans , Interferon-gamma/chemistry , Interferon-gamma/genetics , Ligands , Models, Molecular , Mutation , Mycobacterium Infections/genetics , Mycobacterium Infections/immunology , Protein Stability , Receptors, Interferon/genetics , Signal Transduction , Structure-Activity Relationship , Interferon gamma Receptor
4.
Cell Rep ; 18(10): 2415-2426, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28273456

ABSTRACT

Insulin resistance is a chronic inflammatory condition accompanying obesity or high fat diets that leads to type 2 diabetes. It is hypothesized that lipids and gut bacterial compounds in particular contribute to metabolic inflammation by activating the immune system; however, the receptors detecting these "instigators" of inflammation remain largely undefined. Here, we show that circulating activators of NOD1, a receptor for bacterial peptidoglycan, increase with high fat feeding in mice, suggesting that NOD1 could be a critical sensor leading to metabolic inflammation. Hematopoietic depletion of NOD1 did not prevent weight gain but protected chimeric mice against diet-induced glucose and insulin intolerance. Mechanistically, while macrophage infiltration of adipose tissue persisted, notably these cells were less pro-inflammatory, had lower CXCL1 production, and consequently, lower neutrophil chemoattraction into the tissue. These findings reveal macrophage NOD1 as a cell-specific target to combat diet-induced inflammation past the step of macrophage infiltration, leading to insulin resistance.


Subject(s)
Hematopoiesis , Inflammation/metabolism , Inflammation/pathology , Insulin Resistance , Nod1 Signaling Adaptor Protein/metabolism , Adipose Tissue/pathology , Animals , Cell Movement/drug effects , Cell Polarity/drug effects , Chemokine CXCL1/metabolism , Chemotactic Factors/pharmacology , Diet, High-Fat , Disease Models, Animal , Gene Deletion , Glucose/metabolism , Hematopoiesis/drug effects , Inflammation/complications , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Neutrophil Infiltration/drug effects , Obesity/blood , Obesity/complications , Obesity/pathology
5.
J Immunol ; 197(10): 3959-3969, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27798168

ABSTRACT

Invariant NKT (iNKT) cells develop and differentiate in the thymus, segregating into iNKT1/2/17 subsets akin to Th1/2/17 classical CD4+ T cells; however, iNKT TCRs recognize Ags in a fundamentally different way. How the biophysical parameters of iNKT TCRs influence signal strength in vivo and how such signals affect the development and differentiation of these cells are unknown. In this study, we manipulated TCRs in vivo to generate clonotypic iNKT cells using TCR retrogenic chimeras. We report that the biophysical properties of CD1d-lipid-TCR interactions differentially impacted the development and effector differentiation of iNKT cells. Whereas selection efficiency strongly correlated with TCR avidity, TCR signaling, cell-cell conjugate formation, and iNKT effector differentiation correlated with the half-life of CD1d-lipid-TCR interactions. TCR binding properties, however, did not modulate Ag-induced iNKT cytokine production. Our work establishes that discrete TCR interaction kinetics influence iNKT cell development and central priming.


Subject(s)
Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/metabolism , Animals , Antigens/immunology , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Cell Differentiation , Cytokines/biosynthesis , Cytokines/immunology , Half-Life , Kinetics , Lipids/immunology , Lymphocyte Activation , Mice , Natural Killer T-Cells/physiology , Protein Binding , Receptors, Antigen, T-Cell/genetics
6.
J Immunol ; 197(11): 4464-4472, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27799307

ABSTRACT

NKT cells are unconventional T cells that respond to self and microbe-derived lipid and glycolipid Ags presented by the CD1d molecule. Invariant NKT (iNKT) cells influence immune responses in numerous diseases. Although only a few studies have examined their role during intestinal inflammation, it appears that iNKT cells protect from Th1-mediated inflammation but exacerbate Th2-mediated inflammation. Studies using iNKT cell-deficient mice and chemically induced dextran sodium sulfate (DSS) colitis have led to inconsistent results. In this study, we show that CD1d-deficient mice, which lack all NKT cells, harbor an altered intestinal microbiota that is associated with exacerbated intestinal inflammation at steady-state and following DSS treatment. This altered microbiota, characterized by increased abundance of the bacterial phyla Proteobacteria, Deferribacteres, and TM7, among which the mucin-eating Mucispirillum, as well as members of the genus Prevotella and segmented filamentous bacteria, was transmissible upon fecal transplant, along with the procolitogenic phenotype. Our results also demonstrate that this proinflammatory microbiota influences iNKT cell function upon activation during DSS colitis. Collectively, alterations of the microbiota have a major influence on colitis outcome and therefore have to be accounted for in such experimental settings and in studies focusing on iNKT cells.


Subject(s)
Colitis/immunology , Colitis/microbiology , Gastrointestinal Microbiome/immunology , Lymphocyte Activation , Natural Killer T-Cells/immunology , Animals , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate/toxicity , Fecal Microbiota Transplantation , Inflammation/chemically induced , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Mice , Mice, Knockout , Natural Killer T-Cells/pathology , Prevotella/immunology , Th2 Cells/immunology , Th2 Cells/pathology
7.
J Exp Med ; 213(13): 2841-2850, 2016 12 12.
Article in English | MEDLINE | ID: mdl-27836928

ABSTRACT

The mammalian gastrointestinal tract hosts a diverse community of microbes including bacteria, fungi, protozoa, helminths, and viruses. Through coevolution, mammals and these microbes have developed a symbiosis that is sustained through the host's continuous sensing of microbial factors and the generation of a tolerant or pro-inflammatory response. While analyzing T cell-driven colitis in nonlittermate mouse strains, we serendipitously identified that a nongenetic transmissible factor dramatically increased disease susceptibility. We identified the protozoan Tritrichomonas muris as the disease-exacerbating element. Furthermore, experimental colonization with T. muris induced an elevated Th1 response in the cecum of naive wild-type mice and accelerated colitis in Rag1-/- mice after T cell transfer. Overall, we describe a novel cross-kingdom interaction within the murine gut that alters immune cell homeostasis and disease susceptibility. This example of unpredicted microbial priming of the immune response highlights the importance of studying trans-kingdom interactions and serves as a stark reminder of the importance of using littermate controls in all mouse research.


Subject(s)
Colitis/immunology , Immunity, Mucosal , Intestinal Mucosa/immunology , T-Lymphocytes/immunology , Tritrichomonas/immunology , Animals , Colitis/genetics , Colitis/parasitology , Colitis/pathology , Disease Susceptibility/immunology , Disease Susceptibility/parasitology , Disease Susceptibility/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Mice , Mice, Knockout
8.
J Immunol ; 191(11): 5646-54, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24163408

ABSTRACT

Invariant NKT (iNKT) cells act at the crossroad between innate and adaptive immunity and are important players in the defense against microbial pathogens. iNKT cells can detect pathogens that trigger innate receptors (e.g., TLRs, Rig-I, Dectin-1) within APCs, with the consequential induction of CD1d-mediated Ag presentation and release of proinflammatory cytokines. We show that the cytosolic peptidoglycan-sensing receptors Nod1 and Nod2 are necessary for optimal IFN-γ production by iNKT cells, as well as NK cells. In the absence of Nod1 and Nod2, iNKT cells had a blunted IFN-γ response following infection by Salmonella enterica serovar Typhimurium and Listeria monocytogenes. For Gram-negative bacteria, we reveal a synergy between Nod1/2 and TLR4 in dendritic cells that potentiates IL-12 production and, ultimately, activates iNKT cells. These findings suggest that multiple innate pathways can cooperate to regulate iNKT cell activation during bacterial infection.


Subject(s)
Listeria monocytogenes/immunology , Listeriosis/immunology , Natural Killer T-Cells/immunology , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Salmonella typhi/immunology , Typhoid Fever/immunology , Animals , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/microbiology , Immunity, Innate/genetics , Interferon-gamma/metabolism , Interleukin-12/metabolism , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/genetics , Toll-Like Receptor 4/metabolism
9.
J Biol Chem ; 286(24): 21137-44, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21518761

ABSTRACT

The nature of inflammatory signals determines the outcome of T cell responses. However, little is known about how inflammatory cytokines provided to human CD8 T cells during activation affects their susceptibility to post-activation cell death. We have examined and compared the effects of the inflammatory cytokine IL-12, as well as the combination of IL-1, IL-6, and IL-23 (IL-1/6/23) on the susceptibility of primary human CD8 T cells to post-activation cell death. Human CD8 T cells activated in the presence of IL-1/6/23 underwent significantly less cell death after activation as compared with those activated in IL-12. This was due to reduced susceptibility to Fas-mediated activation-induced cell death (AICD). Mechanistically, the reduced level of cell death in CD8 T cells activated in IL-1/6/23 was a result of a low level of FasL expression and high level of c-FLIP(S) expression. When the effect of IL-1, IL-6, and IL-23 individually was examined, IL-1 or IL-6 alone was sufficient to inhibit CD8 T cell death that occurs after activation in IL-12. IL-1, but not IL-6, inhibited expression of FasL, whereas IL-6, but not IL-1, increased c-FLIP(S) expression. Our findings show that the presence of IL-1 and/or IL-6 during activation of human CD8 T cells attenuates Fas-mediated AICD, whereas IL-12 increases the susceptibility of activated CD8 T cells to this form of cell death.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , CD8-Positive T-Lymphocytes/cytology , Cytokines/metabolism , Fas Ligand Protein/metabolism , Gene Expression Regulation , fas Receptor/metabolism , Apoptosis , Cell Death , Cell Survival , Humans , Inflammation , Interleukin-1/metabolism , Interleukin-12/metabolism , Interleukin-6/metabolism , Receptors, Antigen, T-Cell/metabolism
10.
Arterioscler Thromb Vasc Biol ; 31(5): 1177-84, 2011 May.
Article in English | MEDLINE | ID: mdl-21330602

ABSTRACT

OBJECTIVE: CD155 is a cell surface protein that has recently been described to exert immune regulatory functions. We have characterized the expression of CD155 on human vascular endothelial cells (ECs) and examined its role in the regulation of T-cell activation. METHODS AND RESULTS: CD155 was expressed on resting human vascular ECs and was upregulated in an interferon-γ (IFNγ)-dependent manner. When the function of CD155 in regulating T-cell activation was examined, antibody-mediated neutralization of CD155 did not affect CD8 T-cell proliferation in response to stimulation with ECs. However, neutralization of CD155 activity or small interfering RNA-mediated inhibition of CD155 expression in ECs increased expression of IFNγ and cytotoxic effector function in activated CD8 T cells. CONCLUSIONS: CD155 is an IFNγ-inducible immune regulatory protein on the surface of human ECs that attenuates the acquisition of effector functions in CD8 T cells.


Subject(s)
ADAM Proteins/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Communication , Endothelial Cells/immunology , Lymphocyte Activation , Membrane Proteins/metabolism , ADAM Proteins/genetics , Antibodies, Neutralizing , Cell Proliferation , Cells, Cultured , Coculture Techniques , Humans , Interferon-gamma/metabolism , Interleukin-1alpha/metabolism , Interleukin-6/metabolism , Membrane Proteins/genetics , RNA Interference , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...