Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
MAbs ; 13(1): 1868066, 2021.
Article in English | MEDLINE | ID: mdl-33404287

ABSTRACT

LIGHT is a member of the tumor necrosis factor superfamily, which has been claimed to mediate anti-tumor activity on the basis of cancer cures observed in immunocompetent mice bearing transgenic LIGHT-expressing tumors. The preclinical development of a LIGHT-based therapeutic has been hindered by the lack of functional stability exhibited by this protein. Here, we describe the cloning, expression, and characterization of five antibody-LIGHT fusion proteins, directed against the alternatively spliced extra domain A of fibronectin, a conserved tumor-associated antigen. Among the five tested formats, only the sequential fusion of the F8 antibody in single-chain diabody format, followed by the LIGHT homotrimer expressed as a single polypeptide, yielded a protein (termed "F8-LIGHT") that was not prone to aggregation. A quantitative biodistribution analysis in tumor-bearing mice, using radio-iodinated protein preparations, confirmed that F8-LIGHT was able to preferentially accumulate at the tumor site, with a tumor-to-blood ratio of ca. five to one 24 hours after intravenous administration. Tumor therapy experiments, performed in two murine tumor models (CT26 and WEHI-164), featuring different levels of lymphocyte infiltration into the neoplastic mass, revealed that F8-LIGHT could significantly reduce tumor-cell growth and was more potent than a similar fusion protein (KSF-LIGHT), directed against hen egg lysozyme and serving as negative control of irrelevant specificity in the mouse. At a mechanistic level, the activity of F8-LIGHT was mainly due to an intratumoral expansion of natural killer cells, whereas there was no evidence of expansion of CD8 + T cells, neither in the tumor, nor in draining lymph nodes. Abbreviations: CTLA-4: Cytotoxic T-lymphocytes-associated protein 4; EGFR: Epidermal growth factor receptor; HVEM: Herpesvirus entry mediator; IFNγ: Interferon-gamma; LIGHT: Lymphotoxin, exhibits inducible expression and competes with HSV glycoprotein D for binding to herpesvirus entry mediator, a receptor expressed on T lymphocytes; LTßR: Lymphotoxin beta receptor; NF-κB: Nuclear factor "kappa-light-chain-enhancer" of activated B cells; NK: Natural killer cells; PD-1: Programmed cell death protein 1; PD-L1: Programmed death-ligand 1; TNF: Tumor necrosis factor.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Killer Cells, Natural/drug effects , Neoplasms/drug therapy , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Animals , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/metabolism , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Disease Progression , Humans , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/metabolism , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics , Tissue Distribution , Tumor Burden/drug effects , Tumor Burden/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
2.
Front Immunol ; 11: 922, 2020.
Article in English | MEDLINE | ID: mdl-32499782

ABSTRACT

Tumor necrosis factor superfamily member 14 (LIGHT) has been in pre-clinical development for over a decade and shows promise as a modality of enhancing treatment approaches in the field of cancer immunotherapy. To date, LIGHT has been used to combat cancer in multiple tumor models where it can be combined with other immunotherapy modalities to clear established solid tumors as well as treat metastatic events. When LIGHT molecules are delivered to or expressed within tumors they cause significant changes in the tumor microenvironment that are primarily driven through vascular normalization and generation of tertiary lymphoid structures. These changes can synergize with methods that induce or support anti-tumor immune responses, such as checkpoint inhibitors and/or tumor vaccines, to greatly improve immunotherapeutic strategies against cancer. While investigators have utilized multiple vectors to LIGHT-up tumor tissues, there are still improvements needed and components to be found within a human tumor microenvironment that may impede translational efforts. This review addresses the current state of this field.


Subject(s)
Immunotherapy/methods , Neoplasms/therapy , Tumor Microenvironment/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 14/therapeutic use , Animals , Clinical Trials as Topic , Combined Modality Therapy , Humans , Immunity , Mice , Neoplasms/immunology , Tumor Microenvironment/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology
3.
Rheumatology (Oxford) ; 59(4): 860-868, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31497844

ABSTRACT

OBJECTIVE: To address heterogeneity complicating primary SS (pSS) clinical trials, research and care by characterizing and clustering patients by their molecular phenotypes. METHODS: pSS patients met American-European Consensus Group classification criteria and had at least one systemic manifestation and stimulated salivary flow of ⩾0.1 ml/min. Correlated transcriptional modules were derived from gene expression microarray data from blood (n = 47 with appropriate samples). Patients were clustered based on this molecular information using an unbiased random forest modelling approach. In addition, multiplex, bead-based assays and ELISAs were used to assess 30 serum cytokines, chemokines and soluble receptors. Eleven autoantibodies, including anti-Ro/SSA and anti-La/SSB, were measured by Bio-Rad Bioplex 2200. RESULTS: Transcriptional modules distinguished three clusters of pSS patients. Cluster 1 showed no significant elevation of IFN or inflammation modules. Cluster 2 showed strong IFN and inflammation modular network signatures, as well as high plasma protein levels of IP-10/CXCL10, MIG/CXCL9, BLyS (BAFF) and LIGHT. Cluster 3 samples exhibited moderately elevated IFN modules, but with suppressed inflammatory modules, increased IP-10/CXCL10 and B cell-attracting chemokine 1/CXCL13 and trends toward increased MIG/CXCL9, IL-1α, and IL-21. Anti-Ro/SSA and anti-La/SSB were present in all three clusters. CONCLUSION: Molecular profiles encompassing IFN, inflammation and other signatures can be used to separate patients with pSS into distinct clusters. In the future, such profiles may inform patient selection for clinical trials and guide treatment decisions.


Subject(s)
Gene Expression , Sjogren's Syndrome/genetics , Adult , Antibodies, Antinuclear/immunology , Autoantibodies/immunology , B-Cell Activating Factor/genetics , B-Cell Activating Factor/immunology , B-Cell Activating Factor/metabolism , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Chemokine CXCL13/genetics , Chemokine CXCL13/immunology , Chemokine CXCL13/metabolism , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Chemokine CXCL9/metabolism , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Gene Regulatory Networks , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interferons/genetics , Interferons/immunology , Interferons/metabolism , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Interleukin-1alpha/metabolism , Interleukins/genetics , Interleukins/immunology , Interleukins/metabolism , Male , Middle Aged , Models, Statistical , Phenotype , Sjogren's Syndrome/classification , Sjogren's Syndrome/immunology , Sjogren's Syndrome/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
4.
J Immunol ; 202(10): 2999-3007, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30952816

ABSTRACT

The lymphatic vasculature is an important route for dendritic cell (DC) or tumor cell migration from peripheral tissues to draining lymph nodes (DLNs). However, the underlying molecular and cellular mechanisms remain poorly understood. In this study, using conventional bone marrow chimeric mice and additional UVB radiation, we found that deficiency of LIGHT but not lymphotoxin (LT) α1ß2, likely on radioresistant Langerhans cells (LCs), resulted in impaired skin DC migration to DLNs during LPS-induced inflammation. In addition, LT ß receptor (LTßR), but not herpes virus entry mediator, was found to be the receptor of LIGHT controlling DC migration. Furthermore, conditional deficiency of LTßR in Tie2 cre or Lyve1 cre mice, but not in LTßR-deficient bone marrow chimeric mice, impaired DC migration, suggesting an important role of LTßR in radioresistant lymphatic endothelial cells (LECs), although the role of LTßR in blood endothelial cells remains intriguing. Mechanistically, the gene expression of both CCL21 and CCL19 was found to be reduced in skin LECs isolated from LC-LIGHT-conditionally deficient or Lyve1 cre Ltbr fl/fl mice compared with their controls upon LPS stimulation. Soluble recombinant LIGHT was able to upregulate CCL21 and CCL19 gene expression on SVEC4-10 endothelial cells. Doxycycline, an inhibitor of soluble LIGHT release in the inflamed skin, impaired skin CCL21 and CCL19 expression and DC migration. In addition, melanoma cell metastasis to DLNs was also inhibited in LC-LIGHT-conditionally deficient or Lyve1 cre Ltbr fl/fl mice. Together, our data suggest, to our knowledge, a previously unrecognized scenario in which LCs activate LECs via the LIGHT-LTßR signaling axis to promote DC migration or tumor cell metastasis.


Subject(s)
Endothelial Cells/immunology , Langerhans Cells/immunology , Lymphatic Vessels/immunology , Lymphotoxin beta Receptor/immunology , Signal Transduction/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Animals , Endothelial Cells/pathology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Langerhans Cells/pathology , Lipopolysaccharides/toxicity , Lymphatic Vessels/pathology , Lymphotoxin beta Receptor/genetics , Mice , Mice, Transgenic , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
5.
J Immunol ; 200(8): 2894-2904, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29514949

ABSTRACT

The transition of effector T cells or memory precursors into distinct long-lived memory T cell subsets is not well understood. Although many molecules made by APCs can contribute to clonal expansion and effector cell differentiation, it is not clear if clonal contraction and memory development is passive or active. Using respiratory virus infection, we found that CD8 T cells that cannot express the TNF family molecule lymphotoxin-like, exhibits inducible expression, competes with HSV glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes (LIGHT) are unimpaired in their initial response and clonally expand to form effector cell pools. Thereafter, LIGHT-deficient CD8 T cells undergo strikingly enhanced clonal contraction with resultant compromised accumulation of both circulating and tissue-resident memory cells. LIGHT expression at the peak of the effector response regulates the balance of several pro- and antiapoptotic genes, including Akt, and has a preferential impact on the development of the peripheral memory population. These results underscore the importance of LIGHT activity in programming memory CD8 T cell development, and suggest that CD8 effector T cells can dictate their own fate into becoming memory cells by expressing LIGHT.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Respiratory Tract Infections/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Virus Diseases/immunology , Animals , Cell Differentiation/immunology , Female , Mice , Mice, Inbred C57BL , Respiratory Tract Infections/virology
6.
J Bone Miner Res ; 33(4): 704-719, 2018 04.
Article in English | MEDLINE | ID: mdl-29178458

ABSTRACT

Multiple cytokines produced by immune cells induce remodeling and aid in maintaining bone homeostasis through differentiation of bone-forming osteoblasts and bone-resorbing osteoclasts. Here, we investigate bone remodeling controlled by the tumor necrosis factor (TNF) superfamily cytokine LIGHT. LIGHT-deficient mice (Tnfsf14-/- ) exhibit spine deformity and reduced femoral cancellous bone mass associated with an increase in the osteoclast number and a slight decrease of osteoblasts compared with WT mice. The effect of LIGHT in bone cells can be direct or indirect, mediated by both the low expression of the anti-osteoclastogenic osteoprotegerin (OPG) in B and T cells and reduced levels of the pro-osteoblastogenic Wnt10b in CD8+ T cells in Tnfsf14-/- mice. LIGHT stimulation increases OPG levels in B, CD8+ T, and osteoblastic cells, as well as Wnt10b expression in CD8+ T cells. The high bone mass in Light and T- and B-cell-deficient mice (Rag- /Tnfsf14- ) supports the cooperative role of the immune system in bone homeostasis. These results implicate LIGHT as a potential target in bone disease. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Bone Remodeling/immunology , Cancellous Bone/immunology , Femur/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/deficiency , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Bone Remodeling/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cancellous Bone/pathology , Femur/physiology , Mice , Mice, Knockout , Osteoblasts/immunology , Osteoclasts/immunology , Osteoclasts/pathology , Osteoprotegerin/genetics , Osteoprotegerin/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Wnt Proteins/genetics , Wnt Proteins/immunology
7.
Biochem Biophys Res Commun ; 490(3): 732-738, 2017 08 26.
Article in English | MEDLINE | ID: mdl-28642135

ABSTRACT

Inflammation is critical for the progression of hyperlipidemia. Although the exact mechanism through which inflammation affects hyperlipidemia is not very clear, evidence suggests that the tumor necrosis factor superfamily member 14 (TNFSF14/LIGHT)LIGHT might regulate lipid metabolism. In this study we investigated the expression of LIGHT in patients with different stages of coronary disease. The expression of lipid metabolism-related enzymes and inflammation-related proteins were further explored in oxidized low-density lipoproteins (oxLDL)-induced THP-1 macrophages. We found that LIGHT is highly expressed and companied with severe inflammations in patients with coronary disease. LIGHT significantly enhanced inflammation response in oxLDL-induced THP-1 macrophages. We further demonstrated that LIGHT markedly decreased the levels of lipolytic genes and increased the expressions of lipogenic genes in oxLDL-induced THP-1 macrophages. In addition, our results showed that LIGHT exerts its pro-inflammatory and pro-lipogenesis roles through activating nuclear factor-kappa B (NF-κB) signaling pathway. Taken together our study has demonstrated that LIGHT NF-κB-dependently exacerbates inflammation response and promotes lipid accumulation, and provided a new potential target for treatment of hyperlipidemia-related disease.


Subject(s)
Coronary Artery Disease/immunology , Hyperlipidemias/immunology , Inflammation/immunology , Lipoproteins, LDL/immunology , Macrophages/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Cell Line , Coronary Artery Disease/complications , Humans , Hyperlipidemias/complications , Inflammation/complications , NF-kappa B/immunology , Signal Transduction
8.
Cancer Res ; 77(8): 1880-1891, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28249900

ABSTRACT

The majority of patients with colon cancer will develop advanced disease, with the liver being the most common site of metastatic disease. Patients with increased numbers of tumor-infiltrating lymphocytes in primary colon tumors and liver metastases have improved outcomes. However, the molecular factors that could empower antitumor immune responses in this setting remain to be elucidated. We reported that the immunostimulatory cytokine LIGHT (TNFSF14) in the microenvironment of colon cancer metastases associates with improved patient survival, and here we demonstrate in an immunocompetent murine model that colon tumors expressing LIGHT stimulate lymphocyte proliferation and tumor cell-specific antitumor immune responses. In this model, increasing LIGHT expression in the microenvironment of either primary tumors or liver metastases triggered regression of established tumors and slowed the growth of liver metastases, driven by cytotoxic T-lymphocyte-mediated antitumor immunity. These responses corresponded with significant increases in tumor-infiltrating lymphocytes and increased expression of lymphocyte-homing signals in the metastatic tumors. Furthermore, we demonstrated evidence of durable tumor-specific antitumor immunity. In conclusion, increasing LIGHT expression increased T-cell proliferation, activation, and infiltration, resulting in enhanced tumor-specific immune-mediated tumor regressions in primary tumors and colorectal liver metastases. Mechanisms to increase LIGHT in the colon cancer microenvironment warrant further investigation and hold promise as an immunotherapeutic strategy. Cancer Res; 77(8); 1880-91. ©2017 AACR.


Subject(s)
Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/biosynthesis , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Animals , Cell Line, Tumor , Colonic Neoplasms/pathology , Female , HEK293 Cells , Humans , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology
9.
Clin Pharmacol Drug Dev ; 6(3): 292-301, 2017 May.
Article in English | MEDLINE | ID: mdl-27545119

ABSTRACT

LIGHT, a member of the tumor necrosis factor superfamily, is potentially involved in mucosal inflammation associated with inflammatory bowel disease. The safety and pharmacokinetics of the fully human monoclonal anti-LIGHT antibody, SAR252067, was evaluated in healthy volunteers in a phase 1 study as a potential treatment for diseases related to LIGHT-mediated mucosal inflammation. This double-blind, randomized, placebo-controlled, sequential ascending single-dose, single-center, 16-week study randomized 48 subjects to a single subcutaneous dose of SAR252067 (40, 120, 300, 600, 900, or 1200 mg) or placebo. Safety assessments included adverse events (AEs), injection-site reactions, and antidrug antibody (ADA) titer. Pharmacokinetic end points were serum parameters of SAR252067 (Cmax , AUC0-∞ , tmax , t1/2z ). Serum-soluble LIGHT concentrations were also determined. Safety analyses included all 48 participants; pharmacokinetic analyses included 36 subjects who received SAR252067. No serious AEs were reported, and no dose-effect relationship was apparent. Injection-site reactions were minimal. ADAs were not clinically relevant. SAR252067 exposure increased in a near-dose-proportional manner, median tmax ranged from 5.0 to 8.5 days, and t1/2z ranged from 18.0 to 27.0 days. Serum-soluble LIGHT significantly increased after SAR252067 administration with the 40-mg dose only. SAR252067 had a good safety profile, was well tolerated in healthy humans, and displayed a predictable pharmacokinetic profile.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Adult , Aged , Antibodies, Monoclonal, Humanized , Area Under Curve , Double-Blind Method , Drug Administration Schedule , Female , Half-Life , Healthy Volunteers , Humans , Injections, Subcutaneous , Male , Middle Aged
10.
MAbs ; 8(3): 478-90, 2016.
Article in English | MEDLINE | ID: mdl-26752542

ABSTRACT

Tumor necrosis factor (TNF)/TNF receptor (TNFR) superfamily members play essential roles in the development of the different phases of the immune response. Mouse LIGHT (TNFSF14) is a type II transmembrane protein with a C-terminus extracellular TNF homology domain (THD) that assembles in homotrimers and regulates the course of the immune responses by signaling through 2 receptors, the herpes virus entry mediator (HVEM, TNFRSF14) and the lymphotoxin ß receptor (LTßR, TNFRSF3). LIGHT is a membrane-bound protein transiently expressed on activated T cells, natural killer (NK) cells and immature dendritic cells that can be proteolytically cleaved by a metalloprotease and released to the extracellular milieu. The immunotherapeutic potential of LIGHT blockade was evaluated in vivo. Administration of an antagonist of LIGHT interaction with its receptors attenuated the course of graft-versus-host reaction and recapitulated the reduced cytotoxic activity of LIGHT-deficient T cells adoptively transferred into non-irradiated semiallogeneic recipients. The lack of LIGHT expression on donor T cells or blockade of LIGHT interaction with its receptors slowed down the rate of T cell proliferation and decreased the frequency of precursor alloreactive T cells, retarding T cell differentiation toward effector T cells. The blockade of LIGHT/LTßR/HVEM pathway was associated with delayed downregulation of interleukin-7Rα and delayed upregulation of inducible costimulatory molecule expression on donor alloreactive CD8 T cells that are typical features of impaired T cell differentiation. These results expose the relevance of LIGHT/LTßR/HVEM interaction for the potential therapeutic control of the allogeneic immune responses mediated by alloreactive CD8 T cells that can contribute to prolong allograft survival.


Subject(s)
Adoptive Transfer , CD8-Positive T-Lymphocytes , Cell Proliferation , Signal Transduction/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, Interleukin-7/immunology , Signal Transduction/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
11.
Wei Sheng Wu Xue Bao ; 55(4): 492-500, 2015 Apr 04.
Article in Chinese | MEDLINE | ID: mdl-26211324

ABSTRACT

OBJECTIVE: To study the role of lymphotoxin-like inducible protein that competes with glycoprotein D for herpesvirus entry on T cells (LIGHT) in the development of protective immunity and pathology during Chlamydia Muridarum urogenital infection in mice. METHODS: C57BL/6J wild type (wt) and mice deficient in LIGHT (LIGHT KO) were inoculated intravaginally with 1 x 10(4) IFUs of live C. muridarum organisms. Half mice of each group were reinfected on day 49 after primary infection. We took mice vaginal swabs every 3 or 4 days to monitor live organism shedding. On day 80 after the primary infection, mice were sacrificed, the vaginal tract was isolated for pathology analysis. The spleen cells were collected and IL-4, IL-5, IL-17 and IFN-y were detected by ELISA in the spleen cells culture supernatant after restimulated by UV-MoPn EB. The titers of different Ab isotypes were measured in mice serum by Indirect Immunofluorescence Assay. RESULTS: The chlamydia shedding time of LIGHT KO mice was similar to wild type mice, which cleared the organisms within 28 days after primary infection, and acquired protective immunity against C. muridarum reinfection. All mice regardless of genotypes developed severe upper genital tract pathology and showed no significant difference between LIGHT KO and wild type mice. All mice developed robust anti-C. muridarum organism IgG antibody responses and the ratios of IgG2a versus IgG1 showed no significant difference between LIGHT KO and wild type mice. Splenocytes from MoPn-infected LIGHT KO and wild type mice produced high levels of IFN-gamma and IL-17, but IL-4 and IL-5 couldn't be detected. CONCLUSIONS: LIGHT signal pathway may not correlated with protection against C. muridarum urogenital tract infection and urogenital tract pathology induced by C. muridarum.


Subject(s)
Chlamydia Infections/immunology , Chlamydia muridarum/physiology , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Vagina/immunology , Animals , Chlamydia Infections/genetics , Chlamydia Infections/microbiology , Female , Humans , Interferon-gamma/immunology , Interleukin-4/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Vagina/microbiology
12.
J Immunol ; 195(1): 194-202, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26026056

ABSTRACT

We previously showed that LIGHT and its receptor herpes virus entry mediator (HVEM) are important for development of optimal CD4(+) Th1 cell immunity and resistance to primary Leishmania major infection in mice. In this study, we further characterized the contributions of this molecule in dendritic cell (DC) maturation, initiation, and maintenance of primary immunity and secondary anti-Leishmania immunity. Flow-cytometric studies showed that CD8α(+) DC subset was mostly affected by HVEM-Ig and lymphotoxin ß receptor-Ig treatment. LIGHT signaling is required at both the priming and the maintenance stages of primary anti-Leishmania immunity but is completely dispensable during secondary immunity in wild type mice. However, LIGHT blockade led to impaired IL-12 and IFN-γ responses and loss of resistance in healed CD40-deficient mice after L. major challenge. The protective effect of LIGHT was mediated primarily via its interaction with lymphotoxin ß receptor on CD8α(+) DCs. Collectively, our results show that although LIGHT is critical for maintenance of primary Th1 response, it is dispensable during secondary anti-Leishmania immunity in the presence of functional CD40 signaling as seen in wild type mice.


Subject(s)
CD40 Antigens/deficiency , Host-Pathogen Interactions/immunology , Leishmaniasis, Cutaneous/immunology , Th1 Cells/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/parasitology , Bone Marrow Cells/pathology , CD40 Antigens/genetics , CD40 Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/parasitology , Dendritic Cells/pathology , Female , Gene Expression Regulation , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Leishmania major/immunology , Leishmaniasis, Cutaneous/genetics , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Cutaneous/pathology , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/immunology , Mice , Signal Transduction , Th1 Cells/parasitology , Th1 Cells/pathology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
13.
J Allergy Clin Immunol ; 136(3): 757-68, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25680454

ABSTRACT

BACKGROUND: Pulmonary fibrosis is characterized by excessive accumulation of collagen and α-smooth muscle actin in the lung. The key molecules that promote these phenotypes are of clinical interest. OBJECTIVES: Thymic stromal lymphopoietin (TSLP) has been found at high levels in patients with asthma and idiopathic pulmonary fibrosis, and TSLP has been proposed as a primary driver of lung fibrotic disease. We asked whether tumor necrosis factor superfamily protein 14 (TNFSF14) (aka LIGHT) controls TSLP production to initiate fibrosis. METHODS: Expression of TSLP and initiation of pulmonary fibrosis induced by bleomycin were assessed in mice deficient in LIGHT. The ability of recombinant LIGHT, given intratracheally to naive mice, to promote TSLP and fibrosis was also determined. RESULTS: Genetic deletion of LIGHT abolished lung TSLP expression driven by bleomycin, accompanied by near-complete absence of accumulation of lung collagen and α-smooth muscle actin. Furthermore, recombinant LIGHT administered in vivo induced lung expression of TSLP in the absence of other inflammatory stimuli, and strikingly reproduced the primary features of bleomycin-driven disease in a TSLP-dependent manner. Blockade of LIGHT binding to either of its receptors, herpes virus entry mediator and lymphotoxin beta receptor, inhibited clinical symptoms of pulmonary fibrosis, and correspondingly both receptors were found on human bronchial epithelial cells, a primary source of TSLP. Moreover, LIGHT induced TSLP directly in human bronchial epithelial cells and synergized with IL-13 and TGF-ß in vivo to promote TSLP in the lungs and drive fibrosis. CONCLUSIONS: These results show that LIGHT is a profibrogenic cytokine that may be a key driver of TSLP production during the initiation and development of lung fibrotic disease.


Subject(s)
Cytokines/immunology , Lung/immunology , Pulmonary Fibrosis/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Actins/genetics , Actins/immunology , Animals , Bleomycin , Cell Line , Collagen/genetics , Collagen/immunology , Cytokines/genetics , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Fibroblasts/immunology , Fibroblasts/pathology , Gene Expression Regulation , Herpesviridae/immunology , Humans , Lung/pathology , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/immunology , Mice , Mice, Knockout , Protein Binding , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Receptors, Virus/genetics , Receptors, Virus/immunology , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Thymic Stromal Lymphopoietin
14.
Biochem J ; 465(2): 271-9, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25351958

ABSTRACT

Macrophages switch to an anti-inflammatory, 'regulatory'-like phenotype characterized by the production of high levels of interleukin (IL)-10 and low levels of pro-inflammatory cytokines to promote the resolution of inflammation. A potential therapeutic strategy for the treatment of chronic inflammatory diseases would be to administer drugs that could induce the formation of 'regulatory'-like macrophages at sites of inflammation. In the present study, we demonstrate that the clinically approved cancer drugs bosutinib and dasatinib induce several hallmark features of 'regulatory'-like macrophages. Treatment of macrophages with bosutinib or dasatinib elevates the production of IL-10 while suppressing the production of IL-6, IL-12p40 and tumour necrosis factor α (TNFα) in response to Toll-like receptor (TLR) stimulation. Moreover, macrophages treated with bosutinib or dasatinib express higher levels of markers of 'regulatory'-like macrophages including LIGHT, SPHK1 and arginase 1. Bosutinib and dasatinib were originally developed as inhibitors of the protein tyrosine kinases Bcr-Abl and Src but we show that, surprisingly, the effects of bosutinib and dasatinib on macrophage polarization are the result of the inhibition of the salt-inducible kinases. Consistent with the present finding, bosutinib and dasatinib induce the dephosphorylation of CREB-regulated transcription co-activator 3 (CRTC3) and its nuclear translocation where it induces a cAMP-response-element-binding protein (CREB)-dependent gene transcription programme including that of IL-10. Importantly, these effects of bosutinib and dasatinib on IL-10 gene expression are lost in macrophages expressing a drug-resistant mutant of salt-inducible kinase 2 (SIK2). In conclusion, our study identifies the salt-inducible kinases as major targets of bosutinib and dasatinib that mediate the effects of these drugs on the innate immune system and provides novel mechanistic insights into the anti-inflammatory properties of these drugs.


Subject(s)
Aniline Compounds/pharmacology , Immunity, Innate/drug effects , Macrophages/immunology , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Quinolines/pharmacology , Thiazoles/pharmacology , Animals , Arginase/immunology , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/immunology , Cytokines/immunology , Dasatinib , Macrophages/cytology , Mice , Phosphotransferases (Alcohol Group Acceptor)/immunology , Protein Serine-Threonine Kinases/immunology , Transcription Factors/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology
15.
Proc Natl Acad Sci U S A ; 111(52): E5688-96, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25512551

ABSTRACT

Interactions between natural killer (NK) cells and dendritic cells (DCs) aid DC maturation and promote T-cell responses. Here, we have analyzed the response of human NK cells to tumor cells, and we identify a pathway by which NK-DC interactions occur. Gene expression profiling of tumor-responsive NK cells identified the very rapid induction of TNF superfamily member 14 [TNFSF14; also known as homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for HVEM, a receptor expressed by T lymphocytes (LIGHT)], a cytokine implicated in the enhancement of antitumor responses. TNFSF14 protein expression was induced by three primary mechanisms of NK cell activation, namely, via the engagement of CD16, by the synergistic activity of multiple target cell-sensing NK-cell activation receptors, and by the cytokines IL-2 and IL-15. For antitumor responses, TNFSF14 was preferentially produced by the licensed NK-cell population, defined by the expression of inhibitory receptors specific for self-MHC class I molecules. In contrast, IL-2 and IL-15 treatment induced TNFSF14 production by both licensed and unlicensed NK cells, reflecting the ability of proinflammatory conditions to override the licensing mechanism. Importantly, both tumor- and cytokine-activated NK cells induced DC maturation in a TNFSF14-dependent manner. The coupling of TNFSF14 production to tumor-sensing NK-cell activation receptors links the tumor immune surveillance function of NK cells to DC maturation and adaptive immunity. Furthermore, regulation by NK cell licensing helps to safeguard against TNFSF14 production in response to healthy tissues.


Subject(s)
Cell Communication/immunology , Dendritic Cells/immunology , Immunologic Surveillance , Killer Cells, Natural/immunology , Lymphocyte Activation , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Coculture Techniques , Dendritic Cells/cytology , Female , GPI-Linked Proteins/immunology , Humans , Interleukin-15/immunology , Interleukin-2/immunology , K562 Cells , Killer Cells, Natural/cytology , Male , Neoplasms/immunology , Receptors, IgG/immunology
16.
Eur J Immunol ; 44(12): 3708-16, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25316186

ABSTRACT

Environmental signals shape the phenotype and function of activated macrophages. Here, we show that the neuropeptide calcitonin gene-related peptide (CGRP), which is released from sensory nerves, modulates the phenotype of TLR4-activated murine macrophages by enhancing expression of the regulatory macrophage markers IL-10, sphingosine kinase 1 (SPHK1), and LIGHT (lymphotoxin-like, exhibits inducible expression and competes with HSV glycoprotein D for herpesvirus entry mediator, a receptor expressed by T lymphocytes). In contrast, CGRP inhibits production of cytokines characteristic of inflammatory macrophages and does not affect expression of wound-healing macrophage markers upon TLR4 engagement. In IL-4-stimulated macrophages, CGRP increased LIGHT expression, but failed to induce IL-10 and SPHK1. The stimulatory effect of CGRP on IL-10 production required activation of protein kinase A and was linked to prolonged phosphorylation of CREB and sustained nuclear accumulation of CRTC2 and CRTC3 (where CRTC is CREB-regulated transcriptional cofactor). CGRP enhanced expression of regulatory macrophage markers during the early, but not late, phase of LPS-stimulation and this effect was independent of autocrine type-I IFN activity. In contrast, autocrine type-I IFN activity and treatment of macrophages with IFN-ß promoted late-phase IL-10 production, but had only minor influence on LIGHT and SPHK1 expression. Together, the results identify neuroimmunological communication through CGRP as a novel costimulatory pathway promoting the development of a regulatory phenotype of TLR4-stimulated macrophages. CGRP appears to act through a mechanism that involves sustained activation of CREB-dependent gene transcription.


Subject(s)
Calcitonin Gene-Related Peptide/immunology , Macrophage Activation/physiology , Macrophages/immunology , Toll-Like Receptor 4/immunology , Animals , Calcitonin Gene-Related Peptide/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Macrophages/cytology , Mice , Mice, Knockout , Phosphorylation/genetics , Phosphorylation/immunology , Toll-Like Receptor 4/genetics , Transcription Factors/genetics , Transcription Factors/immunology , Transcription, Genetic/genetics , Transcription, Genetic/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology
17.
Vaccine ; 32(35): 4565-4570, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-24951859

ABSTRACT

LIGHT, a TNF superfamily member (TNFSF14), is a type II transmembrane protein expressed on activated T cells and immature dendritic cells (DCs). However, the expression of LIGHT on mature DCs is down-regulated. Recent studies demonstrated that LIGHT provides potent costimulatory activity for T cells, enhancing proliferation and the production of Th1 cytokines independently of the B7-CD28 pathway. Here, we evaluated the effectiveness of peptide-pulsed DC-mediated antiviral immunity in HBV transgenic mice and the immunoadjuvant effect of LIGHT. The bone marrow-derived DCs were modified in vitro with an adenovirus (Ad) vector expressing mouse LIGHT (Ad-LIGHT), the expression of costimulatory molecules was up-regulated and the secretion of cytokines IL-12 and IFN-γ increased. LIGHT-modified DCs enhanced allostimulation for T cells in mixed lymphocyte reaction (MLR). HBV peptide-pulsed DCs elicited HBV specific CD8+ T cell response and reduced the level of HBsAg and HBV DNA in sera of HBV transgenic mice. Importantly, LIGHT-modified DCs could induce stronger antiviral immunity. These results support the concept that genetic modification of DCs with a recombinant LIGHT adenovirus vector may be a useful strategy for antiviral immunotherapy.


Subject(s)
Adenoviridae/genetics , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Genetic Vectors , Hepatitis B Vaccines/immunology , Immunotherapy/methods , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Animals , DNA, Viral/blood , Female , Hepatitis B Surface Antigens/blood , Hepatitis B Vaccines/administration & dosage , Interferon-gamma/metabolism , Interleukin-12/metabolism , Mice, Inbred BALB C , Mice, Transgenic , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
18.
Am J Transplant ; 13(3): 541-51, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23356438

ABSTRACT

The exchange of information during interactions of T cells with dendritic cells, B cells or other T cells regulates the course of T, B and DC-cell activation and their differentiation into effector cells. The tumor necrosis factor superfamily member LIGHT (homologous to lymphotoxin, exhibits inducible expression and competes with HSV glycoprotein D for binding to herpesvirus entry mediator, a receptor expressed on T lymphocytes) is transiently expressed upon T cell activation and modulates CD8 T cell-mediated alloreactive responses upon herpes virus entry mediator (HVEM) and lymphotoxin ß receptor (LTßR) engagement. LIGHT-deficient mice, or WT mice treated with LIGHT-targeting decoy receptors HVEM-Ig, LTßR-Ig or sDcR3-Ig, exhibit prolonged graft survival compared to untreated controls, suggesting that LIGHT modulates the course and severity of graft rejection. Therefore, targeting the interaction of LIGHT with HVEM and/or LTßR using recombinant soluble decoy receptors or monoclonal antibodies represent an innovative therapeutic strategy for the prevention and treatment of allograft rejection and for the promotion of donor-specific tolerance.


Subject(s)
Antibodies, Monoclonal/pharmacology , Graft Rejection/immunology , Graft vs Host Disease/immunology , Lymphotoxin beta Receptor/antagonists & inhibitors , Organ Transplantation , Tumor Necrosis Factor Ligand Superfamily Member 14/antagonists & inhibitors , Animals , Humans , Lymphotoxin beta Receptor/immunology , Lymphotoxin beta Receptor/metabolism , Mice , Protein Binding , Transplantation, Homologous , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology
19.
Curr Opin Pharmacol ; 12(4): 478-85, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22445654

ABSTRACT

The regulation of the immune system is controlled by many cell surface receptors. A prominent representative is the 'molecular switch' HVEM (herpes virus entry mediator) that can activate either proinflammatory or inhibitory signaling pathways. HVEM ligands belong to two distinct families: the TNF-related cytokines LIGHT and lymphotoxin-α, and the Ig-related membrane proteins BTLA and CD160. HVEM and its ligands have been involved in the pathogenesis of various autoimmune and inflammatory diseases, but recent reports indicate that this network may also be involved in tumor progression and resistance to immune response. Here we summarize the recent advances made regarding the knowledge on HVEM and its ligands in cancer cells, and their potential roles in tumor progression and escape to immune responses. Blockade or enhancement of these pathways may help improving cancer therapy.


Subject(s)
Neoplasms/immunology , Receptors, Tumor Necrosis Factor, Member 14/immunology , Animals , Antigens, CD/immunology , GPI-Linked Proteins/immunology , Humans , Neoplasms/drug therapy , Receptors, Immunologic/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology
20.
Immunol Rev ; 244(1): 75-84, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22017432

ABSTRACT

Lymph nodes (LNs) maintain active homeostasis at steady state. However, in response to changes in the local environment, such as local infection, cancer, vaccination, and autoimmune disease, dramatic remodeling of LN occurs. This remodeling includes changes in size, lymph and blood flow, immune cell trafficking and cellularity, lymphatic and blood vessel growth and activation, as well as microarchitecture. Therefore, inflammatory conditions often lead to enlarged nodes; after local inflammation resolves, LNs actively regress in size and return to steady state. Remodeling of lymphatic vessels (LVs) and blood vessels (BVs) during both the expansion and regression phases are key steps in controlling LN size as well as function. The cells, membrane-associated molecules, and soluble cytokines that are essential for LV and BV homeostasis as well as dynamic changes in the expansion and regression phases have not been well defined. Understanding the underlying cellular and molecular mechanisms behind LN remodeling would help us to better control undesired immune responses (e.g. inflammation and autoimmune diseases) or promote desired responses (e.g. antitumor immunity and vaccination). In this review, we focus on how the closely related tumor necrosis factor (TNF) members: LIGHT (TNFSF14), lymphotoxin-αß, and TNF-α contribute to the remodeling of LNs at various stages of inflammation.


Subject(s)
Blood Vessels/immunology , Immunity, Innate , Lymph Nodes/immunology , Lymphatic Vessels/immunology , Lymphotoxin alpha1, beta2 Heterotrimer/immunology , Signal Transduction/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Blood Vessels/cytology , Blood Vessels/metabolism , Cell Movement , Cellular Microenvironment/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Gene Expression , Homeostasis/immunology , Humans , Inflammation , Lymph Nodes/blood supply , Lymph Nodes/cytology , Lymph Nodes/metabolism , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Lymphotoxin alpha1, beta2 Heterotrimer/genetics , Lymphotoxin alpha1, beta2 Heterotrimer/metabolism , Mice , Mice, Knockout , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...