Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Ann Surg Oncol ; 29(11): 7135-7146, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35543908

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is highly recurrent. Cancer-associated fibroblasts (CAFs), a major component of the tumor microenvironment, promote malignancy; however, the mechanisms underlying their actions are obscure. We aimed to identify CAF-specific proteins in HCC and determine whether they could be potential therapeutic targets. METHODS: Using comprehensive proteomic analysis of CAFs and noncancerous fibroblasts (NFs) primary-cultured from resected HCC specimens from the same patients, CAF-specific proteins were identified. Immunohistochemistry for versican (VCAN) was performed on cancerous tissues obtained from 239 patients with HCC. Conditioned medium from CAFs transfected with siRNA for VCAN was analyzed in vitro. RESULTS: CAFs significantly promoted HCC cell proliferation, migration, and invasion (p < 0.01, 0.01, and 0.01, respectively) compared with NFs. VCAN was upregulated in CAFs, and its stromal level correlated with poor differentiation (p = 0.009) and positive vascular invasion (p = 0.003). Stromal VCAN level was also associated with significantly lower overall (p = 0.002) and relapse-free (p < 0.001) survival rates. It also independently predicted prognosis and recurrence. VCAN-knockdown CAFs significantly suppressed HCC cell migration and invasion compared with negative control. CONCLUSIONS: VCAN secreted from CAFs promoted malignant transformation of HCC cells and has potential as a new therapeutic target in HCC.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Hepatocellular , Liver Neoplasms , Cancer-Associated Fibroblasts/pathology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Hepatocyte Growth Factor/metabolism , Humans , Immunologic Factors/metabolism , Liver Neoplasms/pathology , Lymphotoxin-beta/metabolism , Neoplasm Recurrence, Local/pathology , Prognosis , Proteomics , RNA, Small Interfering , Tumor Microenvironment , Versicans/metabolism
2.
Cancer Immunol Res ; 7(11): 1760-1774, 2019 11.
Article in English | MEDLINE | ID: mdl-31624067

ABSTRACT

Head and neck squamous cell carcinomas (HNSCC) promote inflammation in the tumor microenvironment through aberrant NF-κB activation, but the genomic alterations and pathway networks that modulate NF-κB signaling have not been fully dissected. Here, we analyzed genome and transcriptome alterations of 279 HNSCC specimens from The Cancer Genome Atlas (TCGA) cohort and identified 61 genes involved in NF-κB and inflammatory pathways. The top 30 altered genes were distributed across 96% of HNSCC samples, and their expression was often correlated with genomic copy-number alterations (CNA). Ten of the amplified genes were associated with human papilloma virus (HPV) status. We sequenced 15 HPV- and 11 HPV+ human HNSCC cell lines, and three oral mucosa keratinocyte lines, and supervised clustering revealed that 28 of 61 genes exhibit altered expression patterns concordant with HNSCC tissues and distinct signatures related to their HPV status. RNAi screening using an NF-κB reporter line identified 16 genes that are induced by TNFα or Lymphotoxin-ß (LTß) and implicated in the classic and/or alternative NF-κB pathways. Knockdown of TNFR, LTBR, or selected downstream signaling components established cross-talk between the classic and alternative NF-κB pathways. TNFα and LTß induced differential gene expression involving the NF-κB, IFNγ, and STAT pathways, inflammatory cytokines, and metastasis-related genes. Improved survival was observed in HNSCC patients with elevated gene expression in T-cell activation, immune checkpoints, and IFNγ and STAT pathways. These gene signatures of NF-κB activation, which modulate inflammation and responses to the immune therapy, could serve as potential biomarkers in future clinical trials.


Subject(s)
Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , NF-kappa B/metabolism , Signal Transduction/genetics , Cell Line , Cell Proliferation , DNA Copy Number Variations , Databases, Genetic , Female , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/virology , Humans , Inflammation , Interferon-gamma/metabolism , Lymphotoxin-beta/metabolism , Male , NF-kappa B/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/immunology , Papillomavirus Infections/metabolism , Papillomavirus Infections/virology , STAT Transcription Factors/metabolism , Transcriptome , Tumor Necrosis Factor-alpha/metabolism
3.
Gastroenterology ; 156(4): 1190-1205.e14, 2019 03.
Article in English | MEDLINE | ID: mdl-30445013

ABSTRACT

BACKGROUND & AIMS: Cholangiocyte proliferation and ductular reaction contribute to the onset and progression of liver diseases. Little is known about the role of the transcription factor nuclear factor-κB (NF-κB) in this process. We investigated the activities of the RELB proto-oncogene NF-κB subunit in human cholangiocytes and in mouse models of liver disease characterized by a ductular reaction. METHODS: We obtained liver tissue samples from patients with primary sclerosing cholangitis, primary biliary cholangitis, hepatitis B or C virus infection, autoimmune hepatitis, alcoholic liver disease, or without these diseases (controls) from a tissue bank in Germany. Tissues were analyzed by immunohistochemistry for levels of RELB and lymphotoxin ß (LTB). We studied mice with liver parenchymal cell (LPC)-specific disruption of the cylindromatosis (CYLD) lysine 63 deubiquitinase gene (Cyld), with or without disruption of Relb (CyldΔLPC mice and Cyld/RelbΔLPC mice) and compared them with C57BL/6 mice (controls). Mice were fed 5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) or standard chow diets to induce biliary injury or were given injections of CCl4 to induce non-cholestatic liver fibrosis. Liver tissues were analyzed by histology, immunohistochemistry, immunoblots, in situ hybridization, and quantitative real-time polymerase chain reaction. Cholangiocytes were isolated from normal human liver, incubated with LTB receptor agonist, and transfected with small interfering RNAs to knock down RELB. RESULTS: In liver tissues from patients with primary sclerosing cholangitis, primary biliary cholangitis, chronic infection with hepatitis B or C virus, autoimmune hepatitis, or alcoholic liver disease, we detected increased nuclear translocation of RELB and increased levels of LTB in cholangiocytes that formed reactive bile ducts compared with control liver tissues. Human cholangiocytes, but not those with RELB knockdown, proliferated with exposure to LTB. The phenotype of CyldΔLPC mice, which included ductular reaction, oval cell activation, and biliary fibrosis, was completely lost from Cyld/RelbΔLPC mice. Compared with livers from control mice, livers from CyldΔLPC mice (but not Cyld/RelbΔLPC mice) had increased levels of mRNAs encoding cytokines (LTB; CD40; and tumor necrosis factor superfamily [TNFSF] members TNFSF11 [RANKL], TNFSF13B [BAFF], and TNFSF14 [LIGHT]) produced by reactive cholangiocytes. However, these strains of mice developed similar levels of liver fibrosis in response to CCl4 exposure. CyldΔLPC mice and Cyld/RelbΔLPC mice had improved liver function on the DDC diet compared with control mice fed the DDC diet. CONCLUSION: Reactive bile ducts in patients with chronic liver diseases have increased levels of LTB and nuclear translocation of RELB. RELB is required for the ductular reaction and development of biliary fibrosis in CyldΔLPC mice. Deletion of RELB and CYLD from LPCs protects mice from DDC-induced cholestatic liver fibrosis.


Subject(s)
Bile Ducts/metabolism , Bile Ducts/pathology , Cholangitis, Sclerosing/metabolism , Cytokines/genetics , Liver Diseases/metabolism , Transcription Factor RelB/metabolism , Adolescent , Adult , Aged , Animals , Carbon Tetrachloride , Cell Nucleus , Cell Proliferation , Cells, Cultured , Cysteine Endopeptidases/genetics , Deubiquitinating Enzyme CYLD , Dicarbethoxydihydrocollidine , Epithelial Cells/metabolism , Female , Fibrosis , Gene Knockdown Techniques , Humans , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Lymphotoxin beta Receptor/agonists , Lymphotoxin-beta/metabolism , Male , Mice , Middle Aged , Parenchymal Tissue/pathology , Protein Transport , Proto-Oncogene Mas , RNA, Messenger/metabolism , Transcription Factor RelB/genetics , Young Adult
4.
Mol Carcinog ; 58(3): 411-425, 2019 03.
Article in English | MEDLINE | ID: mdl-30488488

ABSTRACT

Head and neck squamous cell carcinomas (HNSCC) preferentially spread to regional cervical tissues and lymph nodes. Here, we hypothesized that lymphotoxin-ß (LTß), receptor LTßR, and NF-κB-inducing kinase (NIK), promote the aberrant activation of alternative NF-κB2/RELB pathway and genes, that enhance migration and invasion of HNSCC. Genomic and expression alterations of the alternative NF-kB pathway were examined in 279 HNSCC tumors from The Cancer Genome Atlas (TCGA) and a panel of HNSCC lines. LTßR is amplified or overexpressed in HNSCC of the larynx or oral cavity, while LTß, NIK, and RELB are overexpressed in cancers arising within lymphoid oropharyngeal and tonsillar sites. Similarly, subsets of HNSCC lines displayed overexpression of LTßR, NIK, and RELB proteins. Recombinant LTß, and siRNA depletion of endogenous LTßR and NIK, modulated expression of LTßR, NIK, and nuclear translocation of NF-κB2(p52)/RELB as well as functional NF-κB promoter reporter activity. Treatment with a NIK inhibitor (1,3[2H,4H]-Iso-Quinoline Dione) reduced the protein expression of NIK and NF-κB2(p52)/RELB, and blocked LTß induced nuclear translocation of RELB. NIK and RELB siRNA knockdown or NIK inhibitor slowed HNSCC migration or invation in vitro. LTß-induces expression of migration and metastasis related genes, including hepatocyte growth/scatter factor receptor MET. Knockdown of NIK or MET similarly inhibited the migration of HNSCC cell lines. This may help explain why HNSCC preferentially migrate to local lymph nodes, where LTß is expressed. Our findings show that LTß/LTßR promotes activation of the alternative NIK-NF-κB2/RELB pathway to enhance MET-mediated cell migration in HNSCC, which could be potential therapeutic targets in HNSCC.


Subject(s)
Carcinoma, Squamous Cell/secondary , Head and Neck Neoplasms/pathology , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-alpha/metabolism , Lymphotoxin-beta/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factor RelB/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Lymphotoxin beta Receptor/genetics , Lymphotoxin-alpha/genetics , Lymphotoxin-beta/genetics , Prognosis , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Transcription Factor RelB/genetics , Tumor Cells, Cultured , NF-kappaB-Inducing Kinase
5.
Eur J Immunol ; 49(3): 428-442, 2019 03.
Article in English | MEDLINE | ID: mdl-30575951

ABSTRACT

IL-4 is critical for differentiation of Th2 cells and antibody isotype switching, but our work demonstrated that it is produced in the peripheral LN under both Type 2, and Type 1 conditions, raising the possibility of other functions. We found that IL-4 is vital for proper positioning of hematopoietic and stromal cells in steady state, and the lack of IL-4 or IL-4Rα correlates with disarrangement of both follicular dendritic cells and CD31+ endothelial cells. We observed a marked disorganization of B cells in these mice, suggesting that the lymphocyte-stromal cell axis is maintained by the IL-4 signaling pathway. This study showed that absence of IL-4 correlates with significant downregulation of Lymphotoxin alpha (LTα) and Lymphotoxin beta (LTß), critical lymphokines for the development and maintenance of lymphoid organs. Moreover, immunization of IL-4 deficient mice with Type 2 antigens failed to induce lymphotoxin production, LN reorganization, or germinal center formation, while this process is IL-4 independent following Type 1 immunization. Additionally, we found that Type 1 antigen mediated LN reorganization is dependent on IFN-γ in the absence of IL-4. Our findings reveal a role of IL-4 in the maintenance of peripheral lymphoid organ microenvironments during homeostasis and antigenic challenge.


Subject(s)
Cell Proliferation , Interleukin-4/immunology , Receptors, Cell Surface/immunology , Stromal Cells/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphotoxin-alpha/immunology , Lymphotoxin-alpha/metabolism , Lymphotoxin-beta/immunology , Lymphotoxin-beta/metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism
6.
Front Immunol ; 9: 2585, 2018.
Article in English | MEDLINE | ID: mdl-30524422

ABSTRACT

Over 1.5 million individuals in the United States are afflicted with inflammatory bowel disease (IBD). While the progression of IBD is multifactorial, chronic, unresolved inflammation certainly plays a key role. Additionally, while multiple immune mediators have been shown to affect pathogenesis, a comprehensive understanding of disease progression is lacking. Previous work has demonstrated that a member of the TNF superfamily, TNFSF14 (LIGHT), which is pro-inflammatory in several contexts, surprisingly plays an important role in protection from inflammation in mouse models of colitis, with LIGHT deficient mice having more severe disease pathogenesis. However, LIGHT is a single member of a complex signaling network. It signals through multiple receptors, including herpes virus entry mediator (HVEM) and lymphotoxin beta receptor (LTßR); these two receptors in turn can bind to other ligands. It remains unknown which receptors and competing ligands can mediate or counteract the outcome of LIGHT-signaling during colitis. Here we demonstrate that LIGHT signaling through LTßR, rather than HVEM, plays a critical role in the progression of DSS-induced colitis, as LTßR deficient mice exhibit a more severe disease phenotype. Further, mice deficient in LTαß do not exhibit differential colitis progression compared to WT mice. However, deletion of both LIGHT and LTαß, but not deletion of both LTαß and LTßR, resulted in a reversal of the adverse effects associated with the loss of LIGHT. In sum, the LIGHT/LTαß/LTßR signaling network contributes to DSS colitis, but there may be additional receptors or indirect effects, and therefore, the relationships between these receptors and ligands remains enigmatic.


Subject(s)
Colitis/immunology , Inflammatory Bowel Diseases/immunology , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-beta/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Animals , Colitis/chemically induced , Dextran Sulfate , Disease Models, Animal , Disease Progression , Humans , Lymphotoxin beta Receptor/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
J Pathol Clin Res ; 4(2): 124-134, 2018 04.
Article in English | MEDLINE | ID: mdl-29665320

ABSTRACT

Cytokine production is essential for follicular dendritic cell (FDC) maintenance and organization of germinal centres. In follicular lymphoma, FDCs are often disarrayed and may lack antigens indicative of terminal differentiation. We investigated the in situ distribution of cells producing lymphotoxin-beta (LTB), lymphotoxin-alpha (LTA), and tumour necrosis factor-alpha (TNFA) transcripts in human reactive lymph nodes and in follicular lymphomas with follicular or diffuse growth pattern. LTB was the cytokine most abundantly produced in germinal centres. LTB was present in nearly 90% of germinal centre cells whereas LTA and TNFA were detected in 30 and 50%, respectively. Moreover, the amount of LTB expressed in reactive germinal centre cells was 80-fold higher than that of LTA and 20-fold higher than that of TNFA. LTB-positive cells were more numerous in the germinal centre dark zone, whereas expression of the FDC proteins CD21, CD23, VCAM, and CXCL13 was more intense in the light zone. Tumour cells of follicular lymphomas produced less LTB than reactive germinal centre cells. The results of the in situ study were confirmed by RT-PCR; LTB was significantly more abundant in reactive lymph nodes than in follicular lymphoma, with the lowest values detected in predominantly diffuse follicular lymphoma. In neoplastic follicles, low production of LTB by tumour B cells was associated with weaker expression of CD21+/CD23+ by FDCs. Our findings detail for the first time the distribution of LTA-, LTB-, and TNFA-producing cells in human reactive germinal centres and in follicular lymphoma. They suggest the possibility that impaired tumour-cell LTB production may represent a determinant of FDC phenotype loss and for defective follicular organization in follicular lymphoma.


Subject(s)
Lymphoma, Follicular/metabolism , Lymphotoxin-beta/metabolism , Adult , Aged , Aged, 80 and over , Child , Dendritic Cells, Follicular/metabolism , Female , Germinal Center/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Lymph Nodes/metabolism , Lymphoma, Follicular/pathology , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Lymphotoxin-beta/genetics , Male , Middle Aged , Phenotype
8.
Proc Natl Acad Sci U S A ; 114(15): 3957-3962, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28348230

ABSTRACT

RelB is an NF-κB family transcription factor activated in the noncanonical pathway downstream of NF-κB-inducing kinase (NIK) and TNF receptor family members including lymphotoxin-ß receptor (LTßR) and CD40. Early analysis suggested that RelB is required for classical dendritic cell (cDC) development based on a severe reduction of cDCs in Relb-/- mice associated with profound myeloid expansion and perturbations in B and T cells. Subsequent analysis of radiation chimeras generated from wild-type and Relb-/- bone marrow showed that RelB exerts cell-extrinsic actions on some lineages, but it has remained unclear whether the impact of RelB on cDC development is cell-intrinsic or -extrinsic. Here, we reevaluated the role of RelB in cDC and myeloid development using a series of radiation chimeras. We found that there was no cell-intrinsic requirement for RelB for development of most cDC subsets, except for the Notch2- and LTßR-dependent subset of splenic CD4+ cDC2s. These results identify a relatively restricted role of RelB in DC development. Moreover, the myeloid expansion in Relb-/- mice resulted from hematopoietic-extrinsic actions of RelB. This result suggests that there is an unrecognized but critical role for RelB within the nonhematopoietic niche that controls normal myelopoiesis.


Subject(s)
Dendritic Cells/physiology , Myeloid Cells/physiology , Transcription Factor RelB/genetics , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Hematopoietic System/cytology , Hematopoietic System/metabolism , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-beta/metabolism , Mice, Inbred C57BL , Mice, Mutant Strains , Protein Serine-Threonine Kinases/metabolism , Spleen/cytology , Spleen/metabolism , Transcription Factor RelB/metabolism , NF-kappaB-Inducing Kinase
9.
Ann Rheum Dis ; 76(1): 235-243, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27098405

ABSTRACT

OBJECTIVES: To investigate whether artery tertiary lymphoid organs (ATLOs) are present in giant cell arteritis (GCA) and that their formation is associated with the ectopic expression of constitutive lymphoid tissue-homing chemokines. METHODS: Reverse transcriptase PCR, immunohistochemical and immunofluorescence analysis were used to determine the presence of ectopic ATLOs in GCA and the expression of chemokines/chemokine receptors and cytokines involved in lymphoneogenesis in the temporal artery samples obtained from 50 patients with GCA and 30 controls. The presence of lymphatic conduits, of follicular dendritic cells (FDCs) precursors and lymphoid tissue inducer cells was also investigated. Finally, expression of CXCL13, B cell activating factor (BAFF), a proliferation-inducing ligand (APRIL) and CCL21 by isolated myofibroblasts was evaluated before and after stimulation with Toll-like receptors (TLRs) agonists and cytokines. RESULTS: ATLOs were observed in the media layer of 60% of patients with GCA in close proximity to high endothelial venules and independently by the age of patients and the presence of atherosclerosis. ATLO formation was also accompanied by the expression of CXCL13, BAFF, a proliferation-inducing ligand (APRIL), lymphotoxin (LT)-ß, interleukin (IL)-17 and IL-7, the presence of FDC precursors and of lymphoid conduits. Stimulation of myofibroblasts with TLR agonists and cytokines resulted in the upregulation of BAFF and CXCL13. CONCLUSIONS: ATLOs occur in the inflamed arteries of patients with GCA possibly representing the immune sites where immune responses towards unknown arterial wall-derived antigens may be organised.


Subject(s)
Chemokines/metabolism , Ectopic Gene Expression/immunology , Giant Cell Arteritis/immunology , Tertiary Lymphoid Structures/immunology , Aged , Aged, 80 and over , B-Cell Activating Factor/metabolism , Biopsy , Cells, Cultured , Chemokine CXCL13/metabolism , Cytokines/metabolism , Female , Giant Cell Arteritis/complications , Giant Cell Arteritis/pathology , Humans , Lymphotoxin-beta/metabolism , Male , Middle Aged , Myofibroblasts/metabolism , Receptors, Chemokine/metabolism , Temporal Arteries/pathology , Tertiary Lymphoid Structures/etiology , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
10.
J Clin Invest ; 126(11): 4331-4345, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27721238

ABSTRACT

Scleroderma is a group of skin-fibrosing diseases for which there are no effective treatments. A feature of the skin fibrosis typical of scleroderma is atrophy of the dermal white adipose tissue (DWAT). Adipose tissue contains adipose-derived mesenchymal stromal cells (ADSCs) that have regenerative and reparative functions; however, whether DWAT atrophy in fibrosis is accompanied by ADSC loss is poorly understood, as are the mechanisms that might maintain ADSC survival in fibrotic skin. Here, we have shown that DWAT ADSC numbers were reduced, likely because of cell death, in 2 murine models of scleroderma skin fibrosis. The remaining ADSCs showed a partial dependence on dendritic cells (DCs) for survival. Lymphotoxin ß (LTß) expression in DCs maintained ADSC survival in fibrotic skin by activating an LTß receptor/ß1 integrin (LTßR/ß1 integrin) pathway on ADSCs. Stimulation of LTßR augmented the engraftment of therapeutically injected ADSCs, which was associated with reductions in skin fibrosis and improved skin function. These findings provide insight into the effects of skin fibrosis on DWAT ADSCs, identify a DC-ADSC survival axis in fibrotic skin, and suggest an approach for improving mesenchymal stromal cell therapy in scleroderma and other diseases.


Subject(s)
Dendritic Cells/metabolism , Dermis/metabolism , Scleroderma, Diffuse/metabolism , Subcutaneous Fat/metabolism , Animals , Cell Survival/genetics , Dendritic Cells/pathology , Dermis/pathology , Disease Models, Animal , Female , Fibrosis , Integrin beta1/genetics , Integrin beta1/metabolism , Lymphotoxin-beta/genetics , Lymphotoxin-beta/metabolism , Mice , Mice, Knockout , Scleroderma, Diffuse/genetics , Scleroderma, Diffuse/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Subcutaneous Fat/pathology
11.
J Autoimmun ; 67: 82-89, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26553386

ABSTRACT

The induction of innate and adaptive immunity is essential for controlling viral infections. Limited or overwhelming innate immunity can negatively impair the adaptive immune response. Therefore, balancing innate immunity separately from activating the adaptive immune response would result in a better antiviral immune response. Recently, we demonstrated that Usp18-dependent replication of virus in secondary lymphatic organs contributes to activation of the innate and adaptive immune responses. Whether specific mechanisms can balance innate and adaptive immunity separately remains unknown. In this study, using lymphocytic choriomeningitis virus (LCMV) and replication-deficient single-cycle LCMV vectors, we found that viral replication of the initial inoculum is essential for activating virus-specific CD8(+) T cells. In contrast, extracellular distribution of virus along the splenic conduits is necessary for inducing systemic levels of type I interferon (IFN-I). Although enforced virus replication is driven primarily by Usp18, B cell-derived lymphotoxin beta contributes to the extracellular distribution of virus along the splenic conduits. Therefore, lymphotoxin beta regulates IFN-I induction independently of CD8(+) T-cell activity. We found that two separate mechanisms act together in the spleen to guarantee amplification of virus during infection, thereby balancing the activation of the innate and adaptive immune system.


Subject(s)
Adaptive Immunity , Immunity, Innate , Virus Replication/immunology , Animals , Disease Models, Animal , Humans , Interferon Type I/biosynthesis , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/virology , Lymphocyte Activation , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Lymphotoxin-beta/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Spleen/immunology , Spleen/metabolism , Spleen/virology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
12.
Kidney Int ; 89(1): 113-26, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26398497

ABSTRACT

Accumulation of inflammatory cells in different renal compartments is a hallmark of progressive kidney diseases including glomerulonephritis (GN). Lymphotoxin ß receptor (LTßR) signaling is crucial for the formation of lymphoid tissue, and inhibition of LTßR signaling has ameliorated several non-renal inflammatory models. Therefore, we tested whether LTßR signaling could also have a role in renal injury. Renal biopsies from patients with GN were found to express both LTα and LTß ligands, as well as LTßR. The LTßR protein and mRNA were localized to tubular epithelial cells, parietal epithelial cells, crescents, and cells of the glomerular tuft, whereas LTß was found on lymphocytes and tubular epithelial cells. Human tubular epithelial cells, mesangial cells, and mouse parietal epithelial cells expressed both LTα and LTß mRNA upon stimulation with TNF in vitro. Several chemokine mRNAs and proteins were expressed in response to LTßR signaling. Importantly, in a murine lupus model, LTßR blockade improved renal function without the reduction of serum autoantibody titers or glomerular immune complex deposition. Thus, a preclinical mouse model and human studies strongly suggest that LTßR signaling is involved in renal injury and may be a suitable therapeutic target in renal diseases.


Subject(s)
Glomerulonephritis, IGA/metabolism , Lupus Nephritis/metabolism , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/metabolism , RNA, Messenger/analysis , Signal Transduction , Adult , Animals , Cell Line , Chemokines/genetics , Chemokines/metabolism , Disease Models, Animal , Epithelial Cells/chemistry , Epithelial Cells/metabolism , Female , Glomerulonephritis, IGA/genetics , Humans , Immunoglobulins/pharmacology , Kidney Glomerulus/chemistry , Kidney Glomerulus/pathology , Kidney Tubules/chemistry , Kidney Tubules/metabolism , Kidney Tubules/pathology , Ligands , Lupus Nephritis/genetics , Lymphocytes/chemistry , Lymphotoxin beta Receptor/analysis , Lymphotoxin beta Receptor/genetics , Lymphotoxin-alpha/analysis , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Lymphotoxin-beta/analysis , Lymphotoxin-beta/genetics , Lymphotoxin-beta/metabolism , Male , Mesangial Cells/metabolism , Mice , Middle Aged , Signal Transduction/drug effects , Transcriptome
13.
Gut ; 65(10): 1765-75, 2016 10.
Article in English | MEDLINE | ID: mdl-26206664

ABSTRACT

OBJECTIVES: The relative contributions of inflammatory signalling and sequential oncogenic dysregulation driving liver cancer pathogenesis remain incompletely understood. Lymphotoxin-ß receptor (LTßR) signalling is critically involved in hepatitis and liver tumorigenesis. Therefore, we explored the interdependence of inflammatory lymphotoxin signalling and specific oncogenic pathways in the progression of hepatic cancer. DESIGN: Pathologically distinct liver tumours were initiated by hydrodynamic transfection of oncogenic V-Akt Murine Thymoma Viral Oncogene Homolog 1 (AKT)/ß-catenin or AKT/Notch expressing plasmids. To investigate the relationship of LTßR signalling and specific oncogenic pathways, LTßR antagonist (LTßR-Fc) or agonist (anti-LTßR) were administered post oncogene transfection. Initiated livers/tumours were investigated for changes in oncogene expression, tumour proliferation, progression, latency and pathology. Moreover, specific LTßR-mediated molecular events were investigated in human liver cancer cell lines and through transcriptional analyses of samples from patients with intrahepatic cholangiocarcinoma (ICC). RESULTS: AKT/ß-catenin-transfected livers displayed increased expression of LTß and LTßR, with antagonism of LTßR signalling reducing tumour progression and enhancing survival. Conversely, enforced LTßR-activation of AKT/ß-catenin-initiated tumours induced robust increases in proliferation and progression of hepatic tumour phenotypes in an AKT-dependent manner. LTßR-activation also rapidly accelerated ICC progression initiated by AKT/Notch, but not Notch alone. Moreover, LTßR-accelerated development coincides with increases of Notch, Hes1, c-MYC, pAKT and ß-catenin. We further demonstrate LTßR signalling in human liver cancer cell lines to be a regulator of Notch, pAKTser473 and ß-catenin. Transcriptome analysis of samples from patients with ICC links increased LTßR network expression with poor patient survival, increased Notch1 expression and Notch and AKT/PI3K signalling. CONCLUSIONS: Our findings link LTßR and oncogenic AKT signalling in the development of ICC.


Subject(s)
Carcinogenesis/metabolism , Cholangiocarcinoma , Liver Neoplasms , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-beta/metabolism , Signal Transduction/physiology , Animals , Cell Proliferation/physiology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Disease Progression , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Statistics as Topic
14.
Iran J Immunol ; 12(3): 188-97, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26412637

ABSTRACT

BACKGROUND: Rotaviruses (RV) are important viral diarrheal agents in calves. Vaccination is an optimum measure to prevent bovine rotaviruses (BRV) infection. However, little research on BRV VP7 vaccine has been done and currently there is no BRV vaccine. OBJECTIVE: To prepare a subunit vaccine of BRV and investigate its efficacy. METHODS: Total RNA was extracted from MA104 cells infected with bovine rotavirus (BRV) strain GSB01. BRV VP7 gene was amplified using real time fluorescence quantitative PCR (qPCR). The pEASY-T3-VP7 plasmid was digested using HindⅢ and BamHI restriction endonucleases, then recombined into the prokaryotic expression vector pET32a. The pET32a-VP7 and pET32a-VP7-LTB (heat-labile enterotoxin B subunit) were transformed into BL21 (DE3) competent cells of Escherichia coli, respectively, and induced with IPTG, then analyzed using SDS-PAGE. Sixty mice were randomly divided into three groups (n=20). Group A mice was used as His-tag control and mice in group B and C were inoculated with pET32a-VP7 and pET32a-VP7-LTB, respectively. VP7 IgG antibody titers and protection efficiency of pET32a-VP7-LTB were further determined in neonatal mice challenged with GSB01 BRV strain. RESULTS: SDS-PAGE analysis showed that the pET32a-VP7 was highly expressed in the BL21 (DE3) cells. PET32a-VP7 and pET32a-VP7-LTB protein could promote VP7 IgG antibody titer(8.33×103 vs. 17.26×103)in mice. Immunization protection ratios of pET32a-VP7 and pET32a-VP7-LTB proteins in the neonatal mice were 86.4% and 91.7%, respectively. CONCLUSION: The fusion protein of pET32a-VP7-LTB had excellent immunogenicity and protected mice from BRV infection. Our findings can be used for further developing of a high-efficiency subunit vaccine of BRV.


Subject(s)
Antigens, Viral/metabolism , Capsid Proteins/metabolism , Enterotoxins/metabolism , Lymphotoxin-beta/metabolism , Recombinant Fusion Proteins/administration & dosage , Rotavirus Infections/prevention & control , Rotavirus/immunology , Viral Vaccines , Animals , Animals, Newborn , Antibodies, Viral , Antigens, Viral/genetics , Capsid Proteins/genetics , Cattle , Enterotoxins/genetics , Lymphotoxin-beta/genetics , Mice , Protein Engineering , Rotavirus Infections/immunology , Vaccines, Subunit
15.
J Biol Chem ; 290(26): 15973-84, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25940088

ABSTRACT

The blockade of tumor necrosis factor (TNF) by etanercept, a soluble version of the human TNF receptor 2 (hTNFR2), is a well established strategy to inhibit adverse TNF-mediated inflammatory responses in the clinic. A similar strategy is employed by poxviruses, encoding four viral TNF decoy receptor homologues (vTNFRs) named cytokine response modifier B (CrmB), CrmC, CrmD, and CrmE. These vTNFRs are differentially expressed by poxviral species, suggesting distinct immunomodulatory properties. Whereas the human variola virus and mouse ectromelia virus encode one vTNFR, the broad host range cowpox virus encodes all vTNFRs. We report the first comprehensive study of the functional and binding properties of these four vTNFRs, providing an explanation for their expression profile among different poxviruses. In addition, the vTNFRs activities were compared with the hTNFR2 used in the clinic. Interestingly, CrmB from variola virus, the causative agent of smallpox, is the most potent TNFR of those tested here including hTNFR2. Furthermore, we demonstrate a new immunomodulatory activity of vTNFRs, showing that CrmB and CrmD also inhibit the activity of lymphotoxin ß. Similarly, we report for the first time that the hTNFR2 blocks the biological activity of lymphotoxin ß. The characterization of vTNFRs optimized during virus-host evolution to modulate the host immune response provides relevant information about their potential role in pathogenesis and may be used to improve anti-inflammatory therapies based on soluble decoy TNFRs.


Subject(s)
Cowpox virus/metabolism , Poxviridae/metabolism , Receptors, Tumor Necrosis Factor, Type II/chemistry , Receptors, Tumor Necrosis Factor, Type II/metabolism , Tumor Necrosis Factor Decoy Receptors/chemistry , Tumor Necrosis Factor Decoy Receptors/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cowpox virus/chemistry , Cowpox virus/genetics , Humans , Lymphotoxin-beta/metabolism , Mice , Molecular Sequence Data , Poxviridae/chemistry , Poxviridae/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Sequence Alignment , Tumor Necrosis Factor Decoy Receptors/genetics , Tumor Necrosis Factors/metabolism , Viral Proteins/genetics
16.
BMC Immunol ; 15: 33, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25266629

ABSTRACT

BACKGROUND: Although a lot is known about how Fibroblastic Reticular Cells (FRCs) can regulate T lymphocytes (T cells), little is understood about whether or how T cells can regulate FRCs. RESULTS: This study shows that the absence of T cells inhibited the secretion of ER-TR7 by splenic FRCs, induced the structural disorder of FRCs, down-regulated the expression of the chemokine ligands CCL21 and CCL19, and weakened the homing ability of T cells to the spleen of nude mice. Transfusion of T cells from BALB/c mice restored the structure and functions of FRCs and recovered them. The expression of lymphotoxin (LT)-B was significantly downregulated in the absence of T cells from nude mice and was recovered after the transfusion of T cells. After the occlusion of the LT-B receptor, the FRCs' structure and functions were not restored by transfusion of T cells. CONCLUSIONS: These data reveal that the absence of T cells will subject spleen FRCs to structural and functional abnormality, and weaken the homing ability of T cells to the spleen. These changes are attributed to the T-cell- derived LT-B.


Subject(s)
Fibroblasts/cytology , Lymphoid Tissue/cytology , Lymphotoxin-beta/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Antibodies, Blocking/pharmacology , Chemokines/metabolism , Down-Regulation , Female , Mice, Inbred BALB C , Mice, Nude , Spleen/metabolism
17.
Science ; 342(6163): 1243-6, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24311691

ABSTRACT

Immunoglobulin A (IgA) production at mucosal surfaces contributes to protection against pathogens and controls intestinal microbiota composition. However, mechanisms regulating IgA induction are not completely defined. We show that soluble lymphotoxin α (sLTα3) produced by RORγt(+) innate lymphoid cells (ILCs) controls T cell-dependent IgA induction in the lamina propria via regulation of T cell homing to the gut. By contrast, membrane-bound lymphotoxin ß (LTα1ß2) produced by RORγt(+) ILCs is critical for T cell-independent IgA induction in the lamina propria via control of dendritic cell functions. Ablation of LTα in RORγt(+) cells abrogated IgA production in the gut and altered microbiota composition. Thus, soluble and membrane-bound lymphotoxins produced by ILCs distinctly organize adaptive immune responses in the gut and control commensal microbiota composition.


Subject(s)
Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestine, Small/immunology , Lymphocyte Subsets/immunology , Lymphotoxin-alpha/immunology , Microbiota/physiology , Adaptive Immunity , Animals , B-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Homeostasis , Immunity, Innate , Immunoglobulin A/biosynthesis , Immunoglobulin Class Switching , Intestine, Small/microbiology , Lymph Nodes/immunology , Lymphocyte Subsets/metabolism , Lymphotoxin-alpha/metabolism , Lymphotoxin-beta/immunology , Lymphotoxin-beta/metabolism , Mice , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
18.
Behav Brain Res ; 257: 129-39, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24095878

ABSTRACT

To identify genes involved in the development/expression of anxiety/fear, we analyzed the gene expression profile in the hippocampus of genetically heterogeneous NIH-HS rats. The NIH-HS rat stock is a unique genetic resource for the fine mapping of quantitative trait loci (QTLs) to very small genomic regions, due to the high amount of genetic recombinants accumulated along more than 50 breeding generations, and for the same reason it can be expected that those genetically heterogeneous rats should be especially useful for studying differential gene expression as a function of anxiety, fearfulness or other complex traits. We selected high- and low-anxious NIH-HS rats according to the number of avoidance responses they performed in a single 50-trial session of the two-way active avoidance task. Rats were also tested in unconditioned anxiety/fearfulness tests, i.e. the elevated zero-maze and a "novel-cage activity" test. Three weeks after behavioral testing, the hippocampus was dissected and prepared for the microarray study. There appeared 29 down-regulated and 37 up-regulated SNC-related genes (fold-change>|2.19|, FDR<0.05) in the "Low-anxious" vs. the "High-anxious" group. Regression analyses (stepwise) revealed that differential expression of some genes could be predictive of anxiety/fear responses. Among those genes for which the present results suggest a link with individual differences in trait anxiety, nine relevant genes (Avpr1b, Accn3, Cd74, Ltb, Nrg2, Oprdl1, Slc10a4, Slc5a7 and RT1-EC12), tested for validation through qRT-PCR, have either neuroendocrinological or neuroinmunological/inflammation-related functions, or have been related with the hippocampal cholinergic system, while some of them have also been involved in the modulation of anxiety or stress-related (neurobiological and behavioral) responses (i.e. Avpr1b, Oprdl1). The present work confirms the usefulness of NIH-HS rats as a good animal model for research on the neurogenetic basis or mechanisms involved in anxiety and/or fear, and suggest that some MHC-(neuroinmunological/inflammation)-related pathways, as well as the cholinergic system within the hippocampus, may play a role in shaping individual differences in trait anxiety.


Subject(s)
Anxiety/pathology , Anxiety/physiopathology , Gene Expression Regulation/genetics , Genetic Heterogeneity , Hippocampus/metabolism , Acid Sensing Ion Channels/genetics , Acid Sensing Ion Channels/metabolism , Animals , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , Anxiety/genetics , Avoidance Learning/physiology , Disease Models, Animal , Exploratory Behavior/physiology , Histocompatibility Antigens/genetics , Histocompatibility Antigens/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Lymphotoxin-beta/genetics , Lymphotoxin-beta/metabolism , Male , Maze Learning/physiology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Rats , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Symporters/genetics , Symporters/metabolism
19.
PLoS Pathog ; 9(3): e1003234, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23555249

ABSTRACT

Exposure to hepatitis C virus (HCV) typically results in chronic infection that leads to progressive liver disease ranging from mild inflammation to severe fibrosis and cirrhosis as well as primary liver cancer. HCV triggers innate immune signaling within the infected hepatocyte, a first step in mounting of the adaptive response against HCV infection. Persistent inflammation is strongly associated with liver tumorigenesis. The goal of our work was to investigate the initiation of the inflammatory processes triggered by HCV viral proteins in their host cell and their possible link with HCV-related liver cancer. We report a dramatic upregulation of the lymphotoxin signaling pathway and more specifically of lymphotoxin-ß in tumors of the FL-N/35 HCV-transgenic mice. Lymphotoxin expression is accompanied by activation of NF-κB, neosynthesis of chemokines and intra-tumoral recruitment of mononuclear cells. Spectacularly, IKKß inactivation in FL-N/35 mice drastically reduces tumor incidence. Activation of lymphotoxin-ß pathway can be reproduced in several cellular models, including the full length replicon and HCV-infected primary human hepatocytes. We have identified NS5B, the HCV RNA dependent RNA polymerase, as the viral protein responsible for this phenotype and shown that pharmacological inhibition of its activity alleviates activation of the pro-inflammatory pathway. These results open new perspectives in understanding the inflammatory mechanisms linked to HCV infection and tumorigenesis.


Subject(s)
Hepacivirus/enzymology , Liver Neoplasms/metabolism , Lymphocytes/immunology , Lymphotoxin-beta/metabolism , RNA-Dependent RNA Polymerase/metabolism , Animals , Cell Line , Chemokines/metabolism , Chemotaxis, Leukocyte , Hepacivirus/pathogenicity , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/virology , Host-Pathogen Interactions , Humans , I-kappa B Kinase/metabolism , Immunity, Innate , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Liver/metabolism , Liver/pathology , Liver/virology , Liver Neoplasms/pathology , Liver Neoplasms/virology , Lymphocyte Activation , Lymphocytes/virology , Male , Mice , Mice, Transgenic , NF-kappa B , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Signal Transduction , Up-Regulation , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Viral Proteins/metabolism
20.
Immunity ; 38(5): 1013-24, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23623380

ABSTRACT

The stromal scaffold of the lymph node (LN) paracortex is built by fibroblastic reticular cells (FRCs). Conditional ablation of lymphotoxin-ß receptor (LTßR) expression in LN FRCs and their mesenchymal progenitors in developing LNs revealed that LTßR-signaling in these cells was not essential for the formation of LNs. Although T cell zone reticular cells had lost podoplanin expression, they still formed a functional conduit system and showed enhanced expression of myofibroblastic markers. However, essential immune functions of FRCs, including homeostatic chemokine and interleukin-7 expression, were impaired. These changes in T cell zone reticular cell function were associated with increased susceptibility to viral infection. Thus, myofibroblasic FRC precursors are able to generate the basic T cell zone infrastructure, whereas LTßR-dependent maturation of FRCs guarantees full immunocompetence and hence optimal LN function during infection.


Subject(s)
Coronavirus Infections/immunology , Lymph Nodes/cytology , Lymph Nodes/metabolism , Myofibroblasts/physiology , T-Lymphocytes/immunology , Animals , Cell Differentiation , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/immunology , Interleukin-7/biosynthesis , Lymph Nodes/immunology , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-beta/biosynthesis , Lymphotoxin-beta/metabolism , Membrane Glycoproteins/biosynthesis , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Murine hepatitis virus/immunology , Myofibroblasts/cytology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...