Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
2.
Cancer Immunol Res ; 11(4): 486-500, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36700864

ABSTRACT

Diverse factors contribute to the limited clinical response to radiotherapy (RT) and immunotherapy in metastatic non-small cell lung cancer (NSCLC), among which is the ability of these tumors to recruit a retinue of suppressive immune cells-such as M2 tumor-associated macrophages (TAM)-thereby establishing an immunosuppressive tumor microenvironment that contributes to tumor progression and radio resistance. M2 TAMs are activated by the STAT6 signaling pathway. Therefore, we targeted STAT6 using an antisense oligonucleotide (ASO) along with hypofractionated RT (hRT; 3 fractions of 12 Gy each) to primary tumors in three bilateral murine NSCLC models (Lewis lung carcinoma, 344SQ-parental, and anti-PD-1-resistant 344SQ lung adenocarcinomas). We found that STAT6 ASO plus hRT slowed growth of both primary and abscopal tumors, decreased lung metastases, and extended survival. Interrogating the mechanism of action showed reduced M2 macrophage tumor infiltration, enhanced TH1 polarization, improved T-cell and macrophage function, and decreased TGFß levels. The addition of anti-PD-1 further enhanced systemic antitumor responses. These results provide a preclinical rationale for the pursuit of an alternative therapeutic approach for patients with immune-resistant NSCLC.


Subject(s)
Carcinoma, Lewis Lung , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Mice , Animals , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/radiotherapy , Lung Neoplasms/drug therapy , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Oligonucleotides, Antisense/metabolism , Macrophages , Carcinoma, Lewis Lung/pathology , Tumor Microenvironment , STAT6 Transcription Factor/metabolism
3.
Nat Cell Biol ; 24(7): 1165-1176, 2022 07.
Article in English | MEDLINE | ID: mdl-35773432

ABSTRACT

CD8+ T cells are central mediators of immune responses against infections and cancer. Here we identified Dapl1 as a crucial regulator of CD8+ T cell responses to cancer and infections. Dapl1 deficiency promotes the expansion of tumour-infiltrating effector memory-like CD8+ T cells and prevents their functional exhaustion, coupled with increased antitumour immunity and improved efficacy of adoptive T cell therapy. Dapl1 controls activation of NFATc2, a transcription factor required for the effector function of CD8+ T cells. Although NFATc2 mediates induction of the immune checkpoint receptor Tim3, competent NFATc2 activation prevents functional exhaustion of CD8+ T cells. Interestingly, exhausted CD8+ T cells display attenuated NFATc2 activation due to Tim3-mediated feedback inhibition; Dapl1 deletion rescues NFATc2 activation and thereby prevents dysfunction of exhausted CD8+ T cells in chronic infection and cancer. These findings establish Dapl1 as a crucial regulator of CD8+ T cell immunity and a potential target for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Hepatitis A Virus Cellular Receptor 2/genetics , Humans , Membrane Proteins , NFATC Transcription Factors/genetics , Neoplasms/genetics , Persistent Infection , Transcription Factors
4.
Front Oncol ; 11: 737425, 2021.
Article in English | MEDLINE | ID: mdl-34497773

ABSTRACT

Radiation therapy (RT) is emerging as an interventional modality in the cancer-immunity cycle, augmenting the activation of an adaptive immune response against tumors. RT, particularly in combination with immunotherapy, can enhance immune memory effects and shape the tumor-directed T-cell populations. However, a single cycle of RT delivered to a limited number of polymetastatic lesions is rarely sufficient to achieve systemic control. We hypothesize that several rounds of RT, akin to several rounds of immunotherapeutic drugs, is likely to provide greater clinical benefit to patients with metastatic disease. We propose that the repeated exposure to tumor antigens released by "pulsed-RT" (i.e., treating 2-4 tumor lesions with 3 irradiation cycles given one month apart) may amplify the adaptive immune response by expanding the tumor-specific T-cell receptor repertoire, the production of high-affinity tumor antibodies, and the generation of memory lymphocytes and thereby improve immune control of systemic disease.

5.
Sci Adv ; 7(36): eabh0609, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34516909

ABSTRACT

Microglia have been implicated in neuroinflammatory diseases, including multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE). We demonstrate that microglia mediate EAE disease progression via a mechanism relying on the noncanonical nuclear factor kB (NF-κB) pathway. Microglia-specific deletion of the noncanonical NF-κB-inducing kinase (NIK) impairs EAE disease progression. Although microglial NIK is dispensable for the initial phase of T cell infiltration into the central nervous system (CNS) and EAE disease onset, it is critical for the subsequent CNS recruitment of inflammatory T cells and monocytes. Our data suggest that following their initial CNS infiltration, T cells activate the microglial noncanonical NF-κB pathway, which synergizes with the T cell-derived cytokine granulocyte-macrophage colony-stimulating factor to induce expression of chemokines involved in the second-wave of T cell recruitment and disease progression. These findings highlight a mechanism of microglial function that is dependent on NIK signaling and required for EAE disease progression.

6.
Cell Rep ; 35(8): 109161, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34038725

ABSTRACT

Adipose tissue macrophages (ATMs) regulate the occurrence of obesity and its related diseases. Here, we found that serine/threonine protein kinase 24 (Stk24) expression is downregulated significantly in ATMs in obese subjects or obese subjects with type 2 diabetes and mice fed a high-fat diet (HFD). We further identified that glucolipotoxicity downregulated Stk24 expression in ATMs. Stk24-deficient mice develop severe HFD-induced metabolic disorders and insulin insensitivity. Mechanistically, Stk24 intervenes in NLRP3 inflammasome assembly in ATMs by associating directly with NLRP3, decreasing interleukin-1ß (IL-1ß) secretion. Accordingly, Stk24 deficiency in the hematopoietic system promotes NLRP3 inflammasome activation, which contributes to exacerbation of metabolic disorders. Intriguingly, Stk24 expression correlates negatively with body mass index (BMI) and the levels of glucose, cholesterol, triglycerides, and low-density lipoprotein in human subjects. These findings provide insights into the function and clinical implications of Stk24 in obesity-mediated metabolic disorders.


Subject(s)
Inflammasomes/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Obesity/genetics , Protein Serine-Threonine Kinases/therapeutic use , Animals , Humans , Inflammation/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL
8.
Nat Immunol ; 22(2): 193-204, 2021 02.
Article in English | MEDLINE | ID: mdl-33398181

ABSTRACT

Metabolic reprograming toward aerobic glycolysis is a pivotal mechanism shaping immune responses. Here we show that deficiency in NF-κB-inducing kinase (NIK) impairs glycolysis induction, rendering CD8+ effector T cells hypofunctional in the tumor microenvironment. Conversely, ectopic expression of NIK promotes CD8+ T cell metabolism and effector function, thereby profoundly enhancing antitumor immunity and improving the efficacy of T cell adoptive therapy. NIK regulates T cell metabolism via a NF-κB-independent mechanism that involves stabilization of hexokinase 2 (HK2), a rate-limiting enzyme of the glycolytic pathway. NIK prevents autophagic degradation of HK2 through controlling cellular reactive oxygen species levels, which in turn involves modulation of glucose-6-phosphate dehydrogenase (G6PD), an enzyme that mediates production of the antioxidant NADPH. We show that the G6PD-NADPH redox system is important for HK2 stability and metabolism in activated T cells. These findings establish NIK as a pivotal regulator of T cell metabolism and highlight a post-translational mechanism of metabolic regulation.


Subject(s)
CD8-Positive T-Lymphocytes/enzymology , Colonic Neoplasms/enzymology , Energy Metabolism , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/enzymology , Melanoma, Experimental/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Cytotoxicity, Immunologic , Enzyme Stability , Female , Glucosephosphate Dehydrogenase/metabolism , Glycolysis , Hexokinase/genetics , Hexokinase/metabolism , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/transplantation , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Mice, Knockout , NADP/metabolism , Phenotype , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Microenvironment , NF-kappaB-Inducing Kinase
9.
Transl Oncol ; 14(2): 100983, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33340886

ABSTRACT

Radiotherapy (RT) has been used to control tumors by physically damaging DNA and inducing apoptosis; it also promotes antitumor immune responses via neoantigens release and augmenting immune-oncology agents to elicit systemic response. Tumor regression after RT can recruit inflammatory cells, such as tumor-associated macrophages and CD11b+ myeloid cell populations, a major subset of which may actually be immunosuppressive. However, these inflammatory cells also express Toll-like receptors (TLRs) that can be stimulated to reverse suppressive characteristics and promote systemic antitumor outcomes. Here, we investigated the effects of adding CMP-001, a CpG-A oligodeoxynucleotide TLR9 agonist delivered in a virus-like particle (VLP), to RT in two murine models (344SQ metastatic lung adenocarcinoma and CT26 colon carcinoma). High-dose RT (12Gy x 3 fractions) significantly increased the percentages of plasmacytoid dendritic cells within the tumor islets 3- and 5-days post-RT; adding CMP-001 after RT also enhanced adaptive immunity by increasing the proportion of CD4+ and CD8+ T cells. RT plus CMP-001-mediated activation of the immune system led to significant inhibition of tumor growth at both primary and abscopal tumor sites, thereby suggesting a new combinatorial treatment strategy for systemic disease.

10.
Cell Mol Immunol ; 18(9): 2262-2274, 2021 09.
Article in English | MEDLINE | ID: mdl-33203937

ABSTRACT

Generation and maintenance of antigen-specific effector and memory T cells are central events in immune responses against infections. We show that TNF receptor-associated factor 2 (TRAF2) maintains a survival signaling axis in effector and memory CD8 T cells required for immune responses against infections. This signaling axis involves activation of Tpl2 and its downstream kinase ERK by NF-κB-inducing kinase (NIK) and degradation of the proapoptotic factor Bim. NIK mediates Tpl2 activation by stimulating the phosphorylation and degradation of the Tpl2 inhibitor p105. Interestingly, while NIK is required for Tpl2-ERK signaling under normal conditions, uncontrolled NIK activation due to loss of its negative regulator, TRAF2, causes constitutive degradation of p105 and Tpl2, leading to severe defects in ERK activation and effector/memory CD8 T cell survival. Thus, TRAF2 controls a previously unappreciated signaling axis mediating effector/memory CD8 T cell survival and protective immunity.


Subject(s)
CD8-Positive T-Lymphocytes , Signal Transduction , CD8-Positive T-Lymphocytes/metabolism , NF-kappa B/metabolism , Phosphorylation , TNF Receptor-Associated Factor 2/metabolism
11.
Front Immunol ; 11: 573326, 2020.
Article in English | MEDLINE | ID: mdl-33178201

ABSTRACT

The role of mitochondria in cancer formation and progression has been studied extensively, but much remains to be understood about this complex relationship. Mitochondria regulate many processes that are known to be altered in cancer cells, from metabolism to oxidative stress to apoptosis. Here, we review the evolving understanding of the role of mitochondria in cancer cells, and highlight key evidence supporting the role of mitochondria in cancer immune evasion and the effects of mitochondria-targeted antitumor therapy. Also considered is how knowledge of the role of mitochondria in cancer can be used to design and improve cancer therapies, particularly immunotherapy and radiation therapy. We further offer critical insights into the mechanisms by which mitochondria influence tumor immune responses, not only in cancer cells but also in immune cells. Given the central role of mitochondria in the complex interactions between cancer and the immune system, high priority should be placed on developing rational strategies to address mitochondria as potential targets in future preclinical and clinical studies. We believe that targeting mitochondria may provide additional opportunities in the development of novel antitumor therapeutics.


Subject(s)
Energy Metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Tumor Escape , Animals , Energy Metabolism/drug effects , Energy Metabolism/radiation effects , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Mitochondria/radiation effects , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Escape/drug effects , Tumor Hypoxia , Tumor Microenvironment , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
12.
Gastroenterology ; 159(5): 1793-1806, 2020 11.
Article in English | MEDLINE | ID: mdl-32745468

ABSTRACT

BACKGROUND & AIMS: Intestinal epithelial cells (IECs) regulate intestinal immune cells, particularly development of T-helper 17 (Th17) cells. Deregulation of this process leads to intestinal inflammation and tumorigenesis, via unknown mechanisms. TANK-binding kinase 1 (TBK1) is expressed by IECs and cells in the innate immune system. We studied the functions of TBK1 in the intestinal immune response and tumorigenesis in mice. METHODS: We performed studies of wild-type mice, mice with conditional disruption of Tbk1 (Tbk1IEC-KO), Tbk1IEC-KO mice crossed with ApcMin/+ mice, and Mt-/- mice crossed with ApcMin/+ mice. Some mice were given intraperitoneal injections of a neutralizing antibody against interleukin 17 (IL17) or IL1ß. Intestine tissues were collected from mice and analyzed by histology, for numbers of adenomas and Th17 cells, and expression of inflammatory cytokines by real-time PCR. IECs were isolated from wild-type and Tbk1IEC-KO mice, stimulated with lipopolysaccharide, co-cultured for with bone marrow-derived macrophages, and analyzed by RNA sequencing and biochemical analyses. RESULTS: Compared to ApcMin/+Tbk1WT mice, ApcMin/+Tbk1IEC-KO mice had significant increases in number and size of intestinal polyps, and significantly more Th17 cells in lamina propria. Administration of an antibody against IL17 reduced the number of intestinal polyps in ApcMin/+Tbk1IEC-KO mice to that observed in ApcMin/+Tbk1WT mice. In culture, TBK1-deficient IECs promoted expression of IL1ß by macrophages, which induced differentiation of naïve CD4+ T cells into Th17 cells. RNA sequencing analysis revealed that the TBK1-deficient IECs had increased expression of metallothionein 1 (MT1), an immune regulator that promotes intestinal inflammation. Intestine tissues from ApcMin/+Mt-/- mice had significant fewer Th17 cells than ApcMin/+Mt+/+ mice, and a significantly lower number of polyps. Analyses of colorectal tumors in the Cancer Genome Atlas found colorectal tumors with high levels of MT1 and IL17 mRNAs to be associated with reduced survival times of patients. CONCLUSIONS: Expression of TBK1 by IECs suppresses expression of MT1 and prevents expression of IL1ß by macrophages and differentiation of Th17 cells, to prevent inflammation and tumorigenesis. Strategies to block this pathway might be developed for colorectal tumorigenesis.


Subject(s)
Adenomatous Polyposis Coli/enzymology , Cell Differentiation , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/immunology , Intestinal Mucosa/enzymology , Intestinal Neoplasms/enzymology , Protein Serine-Threonine Kinases/metabolism , Th17 Cells/immunology , Adenomatous Polyposis Coli/immunology , Adenomatous Polyposis Coli/pathology , Animals , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Epithelial Cells/pathology , Genes, APC , Humans , Immunity, Innate , Immunity, Mucosal , Interleukin-17/metabolism , Interleukin-1beta/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Neoplasms/immunology , Intestinal Neoplasms/pathology , Macrophages/immunology , Macrophages/metabolism , Metallothionein/genetics , Metallothionein/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Th17 Cells/metabolism
13.
Nat Cell Biol ; 21(12): 1604-1614, 2019 12.
Article in English | MEDLINE | ID: mdl-31792381

ABSTRACT

TANK-binding kinase 1 (TBK1) responds to microbial stimuli and mediates the induction of type I interferon (IFN). Here, we show that TBK1 is also a central mediator of growth factor signalling; this function of TBK1 relies on a specific adaptor-TBK-binding protein 1 (TBKBP1). TBKBP1 recruits TBK1 to protein kinase C-theta (PKCθ) through a scaffold protein, CARD10. This enables PKCθ to phosphorylate TBK1 at Ser 716, a crucial step for TBK1 activation by growth factors but not by innate immune stimuli. Although the TBK1-TBKBP1 signalling axis is not required for the induction of type I IFN, it mediates mTORC1 activation and oncogenesis. Conditional deletion of either TBK1 or TBKBP1 in lung epithelial cells inhibits tumourigenesis in a mouse model of lung cancer. In addition to promoting tumour growth, the TBK1-TBKBP1 axis facilitates tumour-mediated immunosuppression through a mechanism that involves induction of the checkpoint molecule PD-L1 and stimulation of glycolysis. These findings suggest a PKCθ-TBKBP1-TBK1 growth factor signalling axis that mediates both tumour growth and immunosuppression.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinogenesis/genetics , Immune Tolerance/genetics , Intercellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , A549 Cells , Animals , CARD Signaling Adaptor Proteins/genetics , Cells, Cultured , Epithelial Cells/pathology , HEK293 Cells , Humans , Immunity, Innate/genetics , Interferon Type I/genetics , Lung/pathology , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Inbred C57BL
14.
Cell Res ; 29(6): 474-485, 2019 06.
Article in English | MEDLINE | ID: mdl-31086255

ABSTRACT

NF-κB, a family of transcription factors regulating diverse biological processes including immune responses, is activated by canonical and noncanonical pathways based on degradation of IκBα and processing of the IκB-like protein p100, respectively. Although p100 responds to noncanonical NF-κB stimuli for processing, it does not undergo degradation, but rather becomes accumulated, along with canonical NF-κB activation. We show here that the stability of p100 is tightly controlled by a deubiquitinase, Otub1. Otub1 deficiency not only promotes signal-induced p100 processing and noncanonical NF-κB activation but also causes steady-state p100 degradation, leading to aberrant NF-κB activation in the canonical pathway. B-cell-conditional deletion of Otub1 results in B-cell hyperplasia, antibody hyper-production, and lupus-like autoimmunity. Otub1-deficient B cells display aberrantly activated phenotypes and overproduce the cytokine IL-6, contributing to autoimmunity induction. Thus, maintenance of p100 stability by Otub1 serves as an unusual mechanism of NF-κB regulation that prevents autoimmunity.


Subject(s)
Autoimmunity , Cysteine Endopeptidases/metabolism , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Animals , Cells, Cultured , Cysteine Endopeptidases/deficiency , Deubiquitinating Enzymes , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Stability
15.
Sci Adv ; 5(2): eaav0163, 2019 02.
Article in English | MEDLINE | ID: mdl-30775439

ABSTRACT

Retinoic acid-inducible gene I (RIG-I), a pivotal cytosolic sensor, recognizes viral RNAs to initiate antiviral innate immunity. However, posttranslational regulation of RIG-I signaling is not well understood. We report here that nuclear Dbf2-related kinase 2 (NDR2) functions as a crucial positive regulator of the RIG-I-mediated antiviral immune response. Overexpression of NDR2 or its kinase-inactive mutants potentiates RNA virus-induced production of type I interferons and proinflammatory cytokines and dampens viral replication. NDR2 conditional knockout mice (Lysm+NDR2f/f) show an impaired antiviral immune response. Mechanistically, NDR2 directly associates with RIG-I and TRIM25, thus facilitating the RIG-I/TRIM25 complex and enhancing the TRIM25-mediated K63-linked polyubiquitination of RIG-I, which is required for the RIG-I-mediated antiviral immune response. Furthermore, NDR2 expression is notably down-regulated in peripheral blood from respiratory syncytial virus-infected patients and in virus-infected macrophages. Collectively, these findings provide insights into the function of NDR2 in antiviral immunity and its related clinical significance.


Subject(s)
DEAD Box Protein 58/metabolism , Host-Pathogen Interactions/immunology , Macrophages/immunology , Macrophages/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Virus Diseases/immunology , Virus Diseases/metabolism , Animals , Biomarkers , Cytokines , Disease Models, Animal , Enzyme Activation , Humans , Immunity , Immunomodulation , Macrophages/virology , Mice , Mice, Knockout , Receptors, Immunologic , Signal Transduction , Ubiquitination , Virus Diseases/virology
16.
Cell Mol Immunol ; 16(2): 165-177, 2019 02.
Article in English | MEDLINE | ID: mdl-29503445

ABSTRACT

B cells home to the lymph nodes (LNs) via high endothelial venules (HEVs) under the guidance of chemokines, particularly CXCL13. However, as CXCL13 is not directly made in HEVs, the molecular mechanism mediating B-cell homing to LNs has remained unclear. We show here that nuclear factor (NF)-κB-inducing kinase (NIK), a kinase mediating activation of the noncanonical NF-κB pathway, functions in lymphatic endothelial cells (LECs) to regulate B-cell homing to LNs. LEC-conditional deletion of NIK in mice did not affect the integrity or global function of lymphatic vessels but caused a severe reduction in the frequency of B cells in LNs. The LEC-specific NIK deficiency did not affect the survival of B cells or the frequency of B cells in the spleen. B-cell adoptive transfer studies revealed that the LEC-specific NIK deletion impairs the ability of LNs to recruit B cells. We further show that NIK mediates expression of the chemokines CXCL13 and CCL19 in LECs. Although CCL19 is also expressed in blood endothelial cells (BECs), CXCL13 is not produced in BECs. These results suggest that NIK regulates naive B-cell homing to LNs via mediating production of the B-cell homing chemokine CXCL13 in LECs.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Endothelial Cells/cytology , Lymph Nodes/immunology , Lymphatic Vessels/immunology , Protein Serine-Threonine Kinases/physiology , Animals , B-Lymphocytes/metabolism , Cells, Cultured , Chemokine CXCL13/metabolism , Dendritic Cells/metabolism , Endothelial Cells/metabolism , Lymph Nodes/metabolism , Lymphatic Vessels/metabolism , Mice , Mice, Knockout , NF-kappa B/metabolism , Signal Transduction , NF-kappaB-Inducing Kinase
17.
Nat Immunol ; 19(11): 1224-1235, 2018 11.
Article in English | MEDLINE | ID: mdl-30250187

ABSTRACT

Dendritic cells (DCs) play an integral role in regulating mucosal immunity and homeostasis, but the signaling network mediating this function of DCs is poorly defined. We identified the noncanonical NF-κB-inducing kinase (NIK) as a crucial mediator of mucosal DC function. DC-specific NIK deletion impaired intestinal immunoglobulin A (IgA) secretion and microbiota homeostasis, rendering mice sensitive to an intestinal pathogen, Citrobacter rodentium. DC-specific NIK was required for expression of the IgA transporter polymeric immunoglobulin receptor (pIgR) in intestinal epithelial cells, which in turn relied on the cytokine IL-17 produced by TH17 cells and innate lymphoid cells (ILCs). NIK-activated noncanonical NF-κB induced expression of IL-23 in DCs, contributing to the maintenance of TH17 cells and type 3 ILCs. Consistent with the dual functions of IL-23 and IL-17 in mucosal immunity and inflammation, NIK deficiency also ameliorated colitis induction. Thus, our data suggest a pivotal role for the NIK signaling axis in regulating DC functions in intestinal immunity and homeostasis.


Subject(s)
Dendritic Cells/immunology , Homeostasis/immunology , Immunity, Mucosal/immunology , Intestinal Mucosa/immunology , Protein Serine-Threonine Kinases/immunology , Animals , Colitis/immunology , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology , NF-kappaB-Inducing Kinase
18.
Nat Commun ; 9(1): 2789, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30018336

ABSTRACT

Interferon (IFN)-stimulated genes (ISGs) play crucial roles in the antiviral immune response; however, IFNs also induce negative regulators that attenuate the antiviral response. Here, we show that both viral and bacterial invasion downregulate Nuclear Dbf2-related kinase 1 (NDR1) expression via the type I IFN signaling pathway. NDR1 promotes the virus-induced production of type I IFN, proinflammatory cytokines and ISGs in a kinase-independent manner. NDR1 deficiency also renders mice more susceptible to viral and bacterial infections. Mechanistically, NDR1 enhances STAT1 translation by directly binding to the intergenic region of miR146a, thereby inhibiting miR146a expression and liberating STAT1 from miR146a-mediated translational inhibition. Furthermore, STAT1 binds to the miR146a promoter, thus decreasing its expression. Together, our results suggest that NDR1 promotion of STAT1 translation is an important event for IFN-dependent antiviral immune response, and suggest that NDR1 has an important role in controlling viral infections.


Subject(s)
Bronchiolitis, Viral/genetics , Feedback, Physiological , Herpesvirus 1, Human/genetics , MicroRNAs/genetics , Protein Serine-Threonine Kinases/genetics , STAT1 Transcription Factor/genetics , Vesicular stomatitis Indiana virus/genetics , Animals , Bronchiolitis, Viral/immunology , Bronchiolitis, Viral/virology , Case-Control Studies , Child , Gene Expression Regulation , HEK293 Cells , Herpesvirus 1, Human/immunology , Host-Pathogen Interactions , Humans , Immunity, Innate , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/immunology , Protein Biosynthesis , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/immunology , RAW 264.7 Cells , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/immunology , STAT1 Transcription Factor/immunology , Signal Transduction , Vesicular stomatitis Indiana virus/immunology
19.
J Immunol ; 198(10): 4086-4095, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28411188

ABSTRACT

Raf kinase inhibitor protein (RKIP) protects against host immunological responses in nematodes and Drosophila Whether RKIP functions in innate immune responses in mammals remains unknown. In this article, we report that RKIP preferentially regulates the TLR3-mediated immune response in macrophages. RKIP deficiency or silencing significantly decreases polyinosinic:polycytidylic acid [Poly(I:C)]-induced IFN-ß, IL-6, and TNF-α production without affecting the counterpart induced by LPS or CpG. Compared with their wild-type counterparts, RKIP-deficient mice produce less IFN-ß, IL-6, and TNF-α in serum and display decreased lethality upon peritoneal Poly(I:C) plus d-galactosamine injection. Mechanistically, RKIP interacts with TBK1 and promotes the Poly(I:C)-induced TANK-binding kinase 1/IRF3 activation. Simultaneously, RKIP enhances the Poly(I:C)-induced interaction between TGF-ß-activated kinase 1 and MAPK kinase 3 (MKK3), thus promoting MKK3/6 and p38 activation. We further demonstrated that Poly(I:C) treatment, but not LPS treatment, induces RKIP phosphorylation at S109. This action is required for RKIP to promote TANK-binding kinase 1 activation, as well as the interaction between TGF-ß-activated kinase 1 and MKK3, which lead to activation of the downstream IRF3 and p38, respectively. Therefore, RKIP acts as a positive-feedback regulator of the TLR3-induced inflammatory response and may be a potential therapeutic target for inflammatory disease.


Subject(s)
Inflammation/immunology , Phosphatidylethanolamine Binding Protein/metabolism , Signal Transduction , Toll-Like Receptor 3/immunology , Toll-Like Receptor 3/metabolism , Animals , Cell Line , Gene Expression Regulation , Immunity, Innate , Inflammation/metabolism , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/blood , Interferon-beta/immunology , Interleukin-6/blood , Interleukin-6/immunology , Macrophages/immunology , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylethanolamine Binding Protein/deficiency , Phosphatidylethanolamine Binding Protein/genetics , Phosphorylation , Poly I-C/administration & dosage , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology
20.
EMBO J ; 35(23): 2553-2565, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27753621

ABSTRACT

TANK-binding kinase 1 (TBK1) activation is a central event in type I interferon production in anti-virus innate immunity. However, the regulatory mechanism underlying TBK1 activation remains unclear. Here we report that Raf kinase inhibitory protein (RKIP) is essential for TBK1 activation and type I interferon production triggered by viral infection. Upon viral infection, RKIP is phosphorylated at serine 109 (S109) by TBK1. Phosphorylation of RKIP enhances its interaction with TBK1 and in turn promotes TBK1 autophosphorylation. Mutation of RKIP S109 to alanine abrogates the interaction between RKIP and TBK1, and the anti-viral function of RKIP RKIP deficiency inhibits intracellular double-stranded RNA- or DNA-induced type I interferon production. Consistently, RKIP deficiency renders the mice more susceptible to vesicular stomatitis virus (VSV) and herpes simplex virus (HSV) infections. This study reveals a previously unrecognized positive feedback loop between RKIP and TBK1 that is essential for type I interferon production in anti-viral innate immunity.


Subject(s)
Feedback, Physiological , Immunity, Innate , Interferon Type I/biosynthesis , Phosphatidylethanolamine Binding Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , Simplexvirus/immunology , Vesiculovirus/immunology , Animals , Cells, Cultured , Disease Models, Animal , Herpes Simplex/immunology , Herpes Simplex/pathology , Herpes Simplex/virology , Macrophages/immunology , Mice , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/pathology , Rhabdoviridae Infections/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...