Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Oncol Rep ; 45(1): 107-118, 2021 01.
Article in English | MEDLINE | ID: mdl-33155663

ABSTRACT

Intrahepatic cholangiocarcinoma (ICC) is a type of cancer that is difficult to cure; chemoresistance of cholangiocarcinoma cells affect the prognosis of patients who cannot be treated with surgery. The mechanism underlying this chemoresistance remains unknown. Mesenchymal stem cells (MSCs) are known to be important components of the tumor microenvironment. In the present study, a large number of MSCs were observed to infiltrate the tumor sites of ICC; thus, MSCs were isolated from ICC tumor tissues. It was revealed that herpesvirus entry mediator (HVEM) was overexpressed in ICC­MSCs. The present study then investigated the role of HVEM­overexpressing MSCs in the chemoresistance of cholangiocarcinoma cells. It was demonstrated that HVEM­overexpressing MSCs could support cell survival of chemotherapeutic cholangiocarcinoma cells and inhibited their apoptosis. Further investigations revealed that HVEM­overexpressing MSCs could secrete IL­6 and also activated AMPK/mTOR­dependent autophagy of cholangiocarcinoma cells. Thus, it was concluded that ICC­MSC­induced autophagy is the primary cause of chemoresistance in ICC.


Subject(s)
Autophagy/physiology , Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Mesenchymal Stem Cells/physiology , AMP-Activated Protein Kinases/physiology , Adult , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cholangiocarcinoma/pathology , Drug Resistance, Neoplasm , Female , Humans , Interleukin-6/physiology , Male , Middle Aged , Receptors, Tumor Necrosis Factor, Member 14/physiology , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology
2.
mBio ; 11(3)2020 05 12.
Article in English | MEDLINE | ID: mdl-32398314

ABSTRACT

Ocular herpes simplex virus 1 (HSV-1) infection leads to an immunopathogenic disease called herpes stromal keratitis (HSK), in which CD4+ T cell-driven inflammation contributes to irreversible damage to the cornea. Herpesvirus entry mediator (HVEM) is an immune modulator that activates stimulatory and inhibitory cosignals by interacting with its binding partners, LIGHT (TNFSF14), BTLA (B and T lymphocyte attenuator), and CD160. We have previously shown that HVEM exacerbates HSK pathogenesis, but the involvement of its binding partners and its connection to the pathogenic T cell response have not been elucidated. In this study, we investigated the role of HVEM and its binding partners in mediating the T cell response using a murine model of ocular HSV-1 infection. By infecting mice lacking the binding partners, we demonstrated that multiple HVEM binding partners were required for HSK pathogenesis. Surprisingly, while LIGHT-/-, BTLA-/-, and CD160-/- mice did not show differences in disease compared to wild-type mice, BTLA-/- LIGHT-/- and CD160-/- LIGHT-/- double knockout mice showed attenuated disease characterized by decreased clinical symptoms, increased retention of corneal sensitivity, and decreased infiltrating leukocytes in the cornea. We determined that the attenuation of disease in HVEM-/-, BTLA-/- LIGHT-/-, and CD160-/- LIGHT-/- mice correlated with a decrease in gamma interferon (IFN-γ)-producing CD4+ T cells. Together, these results suggest that HVEM cosignaling through multiple binding partners induces a pathogenic Th1 response to promote HSK. This report provides new insight into the mechanism of HVEM in HSK pathogenesis and highlights the complexity of HVEM signaling in modulating the immune response following ocular HSV-1 infection.IMPORTANCE Herpes simplex virus 1 (HSV-1), a ubiquitous human pathogen, is capable of causing a progressive inflammatory ocular disease called herpes stromal keratitis (HSK). HSV-1 ocular infection leads to persistent inflammation in the cornea resulting in outcomes ranging from significant visual impairment to complete blindness. Our previous work showed that herpesvirus entry mediator (HVEM) promotes the symptoms of HSK independently of viral entry and that HVEM expression on CD45+ cells correlates with increased infiltration of leukocytes into the cornea during the chronic inflammatory phase of the disease. Here, we elucidated the role of HVEM in the pathogenic Th1 response following ocular HSV-1 infection and the contribution of HVEM binding partners in HSK pathogenesis. Investigating the molecular mechanisms of HVEM in promoting corneal inflammation following HSV-1 infection improves our understanding of potential therapeutic targets for HSK.


Subject(s)
Herpesvirus 1, Human/physiology , Keratitis, Herpetic/immunology , Keratitis, Herpetic/pathology , Receptors, Tumor Necrosis Factor, Member 14/physiology , Virus Internalization , Animals , Cornea/immunology , Cornea/pathology , Cornea/virology , Disease Models, Animal , Female , Herpesvirus 1, Human/immunology , Host Microbial Interactions/immunology , Inflammation , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Member 14/immunology , Signal Transduction , T-Lymphocytes/immunology
3.
J Immunol ; 203(9): 2425-2442, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31562209

ABSTRACT

The BTLA-HVEM checkpoint axis plays extensive roles in immunomodulation and diseases, including cancer and autoimmune disorders. However, the functions of this checkpoint axis in hepatitis remain limited. In this study, we explored the regulatory role of the Btla-Hvem axis in a ConA-induced hepatitis model in zebrafish. Results showed that Btla and Hvem were differentially expressed on intrahepatic Cd8+ T cells and hepatocytes. Knockdown of Btla or Hvem significantly promoted hepatic inflammation. Btla was highly expressed in Cd8+ T cells in healthy liver but was downregulated in inflamed liver, as evidenced by a disparate proportion of Cd8+Btla+ and Cd8+Btla- T cells in individuals without or with ConA stimulation. Cd8+Btla+ T cells showed minimal cytotoxicity to hepatocytes, whereas Cd8+Btla- T cells were strongly reactive. The depletion of Cd8+Btla- T cells reduced hepatitis, whereas their transfer enhanced hepatic inflammation. These observations indicate that Btla endowed Cd8+Btla+ T cells with self-tolerance, thereby preventing them from attacking hepatocytes. Btla downregulation deprived this tolerization. Mechanistically, Btla-Hvem interaction contributed to Cd8+Btla+ T cell tolerization, which was impaired by Hvem knockdown but rescued by soluble Hvem protein administration. Notably, Light was markedly upregulated on Cd8+Btla- T cells, accompanied by the transition of Cd8+Btla+Light- to Cd8+Btla-Light+ T cells during hepatitis, which could be modulated by Cd4+ T cells. Light blockade attenuated hepatitis, thereby suggesting the positive role of Light in hepatic inflammation. These findings provide insights into a previously unrecognized Btla-Hvem-Light regulatory network in hepatic homeostasis and inflammation, thus adding a new potential therapeutic intervention for hepatitis.


Subject(s)
Concanavalin A/pharmacology , Hepatitis/immunology , Homeostasis , Inflammation/etiology , Liver/immunology , Receptors, Immunologic/physiology , Receptors, Tumor Necrosis Factor, Member 14/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Drosophila Proteins/physiology , HEK293 Cells , Humans , Vesicular Transport Proteins/physiology , Zebrafish
4.
Med Sci Monit ; 24: 387-396, 2018 Jan 20.
Article in English | MEDLINE | ID: mdl-29352109

ABSTRACT

BACKGROUND Acute rejection is a common predisposing cause of allograft dysfunction in kidney transplantation. Recently, the B and T lymphocyte attenuator (BTLA)/herpes virus entry mediator (HVEM)/lymphotoxin (LIGHT)/CD160 pathway was found to be potentially involved in the regulation of T cell activation. This could mean that this pathway is involved in graft rejection in kidney transplantation; the present study aimed to explore this possibility. MATERIAL AND METHODS The expression of BTLA, HVEM, LIGHT and CD160 on peripheral CD4+, CD8+ and CD19+ lymphocytes were analyzed by flow cytometry in recipients with biopsy-proven acute rejection (BPAR) or stable allograft function, as well as in healthy volunteers. Moreover, we performed HE staining and immunohistochemical staining to assess the expression of BTLA and HVEM in kidney samples from recipients with BPAR and patients who underwent the surgery of radical nephrectomy. RESULTS We observed the significantly lower expression of BTLA on CD4+ T cells in recipients from the BPAR group than in recipients from the stable group. The expression of BTLA on CD8+ T cells among recipients both from the BPAR and stable group was statistically increased than that in the healthy volunteers. A significant difference in the expression of CD160 in the stable group was found when compared with the BPAR group or control group. Moreover, there was no significance in the expression of HVEM, LIGHT or CD160 on other subtypes of T cells between the 3 groups or in the expression of BTLA on CD4+ T cells between the BPAR and control group. CONCLUSIONS The findings indicate that the BTLA/HVEM pathway does be involved in pathogenesis of acute rejection following kidney transplantation, as well as the induction of transplant tolerance. This pathway may therefore be a useful target for therapy against acute rejection after kidney transplantation.


Subject(s)
Graft Rejection/immunology , Receptors, Immunologic/metabolism , Receptors, Immunologic/physiology , Adult , Antigens, CD/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biopsy , Female , Flow Cytometry , GPI-Linked Proteins/metabolism , Graft Rejection/metabolism , Humans , Kidney/metabolism , Kidney Transplantation/adverse effects , Lymphocyte Activation/physiology , Lymphotoxin-alpha/metabolism , Male , Middle Aged , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Receptors, Tumor Necrosis Factor, Member 14/physiology , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplant Recipients
5.
J BUON ; 22(1): 80-86, 2017.
Article in English | MEDLINE | ID: mdl-28365939

ABSTRACT

PURPOSE: To explore the expression of herpesvirus entry mediator (HVEM) in ovarian serous adenocarcinoma tissues and its relationship with clinicopathological features. METHODS: Paraffin-embedded specimens from 40 patients with ovarian serous adenocarcinoma who were subjected to surgical treatment were used for the determination of HVEM expression by immunohistochemistry (IHC). Then the relationship between the expression of HVEM and the patient clinicopathological features was analyzed. RESULTS: There were 29 cases (72.5%) of HVEM/tumor necrosis factor receptor (TNFR)SF14-positive and 11 cases (27.5%) of HVEM/TNFRSF14-negative. The positive rate of HVEM was significantly correlated with TNM staging, lymph node metastasis and recurrence (p<0.05), but not with age, grade of differentiation and distant metastasis (p>0.05). CONCLUSION: HVEM is highly expressed in ovarian serous adenocarcinoma tissues and correlated with the patient clinicopathological features, such as TNM staging, lymph node metastasis and recurrence. HVEM can provide a basis in the search for a new targeting treatment for ovarian serous adenocarcinoma.


Subject(s)
Cystadenocarcinoma, Serous/pathology , Ovarian Neoplasms/pathology , Receptors, Tumor Necrosis Factor, Member 14/physiology , Adult , Aged , Female , Humans , Middle Aged , Neoplasm Staging , Receptors, Tumor Necrosis Factor, Member 14/analysis
6.
J Gastroenterol Hepatol ; 32(8): 1520-1524, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27987232

ABSTRACT

BACKGROUND AND AIM: The herpes virus entry mediator (HVEM) network has become new directions in targeting novel checkpoint inhibitors for cancer therapy. However, the changes of membrane-bound HVEM (mHVEM) and soluble HVEM (sHVEM) in hepatocellular carcinoma (HCC) are not fully understood. This study aims to study the changes of mHVEM and sHVEM in HCC patients. METHODS: Serum samples were collected from 65 HCC patients, from which sHVEM levels were examined by enzyme-linked immunosorbent assay. Expressions of mHVEM on peripheral lymphocytes from 20 HCC patients were determined using flow cytometry, and associations between mHVEM on T and B cells were analyzed. RESULTS: The levels of mHVEM were downregulated on peripheral lymphocytes in HCC patients, with a strong positive correlation between mHVEM expression on T and B cells. In contrast, the levels of soluble HVEM were upregulated in the serum of HCC patients. Furthermore, we found that the increase in sHVEM level was correlated with advanced stages HCC. CONCLUSION: Our data demonstrated paradoxical changes of membrane and soluble HVEM in the peripheral blood of HCC patients for the first time. These data supported the notion that roles of HVEM are likely to be immunosuppressive rather than activating tumor immunity. Future studies are warranted to further explore the translational values of mHVEM and sHVEM in peripheral blood as diagnostic markers and therapeutic targets.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Receptors, Tumor Necrosis Factor, Member 14/blood , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Adult , Aged , B-Lymphocytes/metabolism , Biomarkers/blood , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/immunology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immune Tolerance , Liver Neoplasms/blood , Liver Neoplasms/diagnosis , Liver Neoplasms/immunology , Male , Middle Aged , Neoplasm Staging , Receptors, Tumor Necrosis Factor, Member 14/physiology , T-Lymphocytes/metabolism , Young Adult
7.
Anticancer Res ; 35(3): 1361-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25750286

ABSTRACT

BACKGROUND: Herpesvirus entry mediator (HVEM) has been recently suggested to play certain roles in cancer biology. We examined HVEM expression in human colorectal cancer (CRC) to reveal its clinical importance. MATERIALS AND METHODS: Immunohistochemical staining was carried-out in normal epithelium, benign and malignant lesions. RESULTS: While intense HVEM expression was not observed in normal epithelium and hyperplastic polyps, 24% of adenoma and more than half of CRCs had high HVEM expression. In 234 CRCs, HVEM expression was significantly associated with tumor status and pathological stage. Patients with high HVEM expression had a significantly poorer prognosis than those with low expression. Importantly, HVEM status had an independent prognostic value in CRC. Furthermore, HVEM status was inversely corrected with the presence of tumor-infiltrating T-cells. CONCLUSION: HVEM may play a critical role in tumor progression and immune evasion, and may also be a novel prognostic marker and potential therapeutic target in human CRC.


Subject(s)
Colorectal Neoplasms/immunology , Receptors, Tumor Necrosis Factor, Member 14/physiology , Adult , Aged , Aged, 80 and over , Colon/chemistry , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease Progression , Humans , Immunohistochemistry , Lymphocytes, Tumor-Infiltrating/immunology , Middle Aged , Prognosis , Receptors, Tumor Necrosis Factor, Member 14/analysis , Tumor Escape
8.
Mol Immunol ; 62(1): 96-103, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24980868

ABSTRACT

Signals mediated by members of the tumor necrosis factor receptor superfamily modulate a network of diverse processes including initiation of inflammatory responses and altering cell fate between pathways favoring survival and death. Although such pathways have been well-described for the TNF-α receptor, less is known about signaling induced by the TNF superfamily member LIGHT and how it is differentially altered by expression of its two receptors LTßR and HVEM in the same cell. We used cell lines with different relative expression of HVEM and LTßR to show that LIGHT-induced signals mediated by these receptors were associated with altered TRAF2 stability and RelA nuclear translocation. Production of the inflammatory chemokine CXCL10 was primarily mediated by LTßR. Higher expression of HVEM was associated with cell survival, while unopposed LTßR signaling favored pathways leading to apoptosis. Importantly, restoring HVEM expression in cells with low endogenous expression recapitulated the phenotype of cells with higher endogenous expression. Together, our data provide evidence that relative expression of HVEM and LTßR modulates canonical NF-κB and pro-apoptotic signals stimulated by LIGHT.


Subject(s)
Lymphocyte Activation , Lymphotoxin beta Receptor/physiology , Receptors, Tumor Necrosis Factor, Member 14/physiology , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/immunology , Down-Regulation/drug effects , Down-Regulation/immunology , Gene Expression/physiology , HEK293 Cells , HT29 Cells , HeLa Cells , Humans , Interferon-gamma/pharmacology , Ligands , Lymphocyte Activation/drug effects , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , U937 Cells
9.
Int J Biol Sci ; 8(10): 1420-30, 2012.
Article in English | MEDLINE | ID: mdl-23197939

ABSTRACT

T lymphocytes require signaling by the T cell receptor and by nonclonotypic cosignaling receptors. The costimulatory and inhibitory signals profoundly influence the course of immune responses by amplifying or reducing the transcriptional effects of T cell receptor triggering. The inhibitory receptors such as CTLA-4, PD-1, and BTLA have recently drawn much attention as potential targets for immunotherapies. This review focuses on the progress that has been made with the mentioned receptors in the field of immunotherapies for autoimmune diseases, malignancies, infectious diseases, and transplantation.


Subject(s)
Antigens, Differentiation/immunology , Immunotherapy/trends , Receptors, Immunologic/immunology , Abatacept , Animals , Antigens, Differentiation/chemistry , Antigens, Differentiation/genetics , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , B7-H1 Antigen/physiology , CD28 Antigens/immunology , CD28 Antigens/metabolism , CTLA-4 Antigen/chemistry , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Mice , Programmed Cell Death 1 Receptor , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Receptors, Tumor Necrosis Factor, Member 14/physiology , Signal Transduction/drug effects , Transplantation Immunology
10.
Proc Natl Acad Sci U S A ; 109(50): 20649-54, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23184983

ABSTRACT

Infection with herpes simplex virus type 1 (HSV-1) and HSV-2 is initiated by viral glycoprotein D (gD) binding to a receptor on the host cell. Two receptors, herpesvirus entry mediator (HVEM) and nectin-1, mediate entry in murine models of HSV-1 and HSV-2. HVEM is dispensable for HSV-2 infection of the vagina and brain, but is required for WT pathogenesis of HSV-1 infection of the cornea. By challenging WT and HVEM KO mice with multiple strains of HSV-1 and HSV-2, we demonstrate that without HVEM, all HSV-1 strains tested do not replicate well in the cornea and infection does not result in severe symptoms, as observed in WT mice. In contrast, all HSV-2 strains tested had no requirement for HVEM to replicate to WT levels in the cornea and still cause severe disease. These findings imply that HSV-2 does not require HVEM to cause disease regardless of route of entry, but HVEM must be present for HSV-1 to cause full pathogenesis in the eye. These findings uncover a unique role for HVEM in mediating HSV-1 infection in an area innervated by the trigeminal ganglion and may explain why the presence of HVEM can lead to severe inflammation in the cornea. Thus, the dependence on HVEM is a dividing point between HSV-1 and HSV-2 that evolved to infect areas innervated by different sensory ganglia.


Subject(s)
Herpesvirus 1, Human/pathogenicity , Herpesvirus 2, Human/pathogenicity , Keratitis, Herpetic/etiology , Receptors, Tumor Necrosis Factor, Member 14/physiology , Animals , Disease Models, Animal , Female , Herpes Genitalis/virology , Herpesvirus 1, Human/classification , Herpesvirus 2, Human/classification , Herpesvirus 2, Human/physiology , Host-Pathogen Interactions , Keratitis, Herpetic/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Member 14/deficiency , Receptors, Tumor Necrosis Factor, Member 14/genetics , Serotyping , Species Specificity , Virulence/physiology , Virus Replication
11.
J Immunol ; 189(1): 39-49, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22634623

ABSTRACT

Allogeneic stem cell transplantation (allo-SCT) can cure hematological malignancies by inducing alloreactive T cell responses targeting minor histocompatibility antigens (MiHA) expressed on malignant cells. Despite induction of robust MiHA-specific T cell responses and long-term persistence of alloreactive memory T cells specific for the tumor, often these T cells fail to respond efficiently to tumor relapse. Previously, we demonstrated the involvement of the coinhibitory receptor programmed death-1 (PD-1) in suppressing MiHA-specific CD8(+) T cell immunity. In this study, we investigated whether B and T lymphocyte attenuator (BTLA) plays a similar role in functional impairment of MiHA-specific T cells after allo-SCT. In addition to PD-1, we observed higher BTLA expression on MiHA-specific CD8(+) T cells compared with that of the total population of CD8(+) effector-memory T cells. In addition, BTLA's ligand, herpes virus entry mediator (HVEM), was found constitutively expressed by myeloid leukemia, B cell lymphoma, and multiple myeloma cells. Interference with the BTLA-HVEM pathway, using a BTLA blocking Ab, augmented proliferation of BTLA(+)PD-1(+) MiHA-specific CD8(+) T cells by HVEM-expressing dendritic cells. Notably, we demonstrated that blocking of BTLA or PD-1 enhanced ex vivo proliferation of MiHA-specific CD8(+) T cells in respectively 7 and 9 of 11 allo-SCT patients. Notably, in 3 of 11 patients, the effect of BTLA blockade was more prominent than that of PD-1 blockade. Furthermore, these expanded MiHA-specific CD8(+) T cells competently produced effector cytokines and degranulated upon Ag reencounter. Together, these results demonstrate that BTLA-HVEM interactions impair MiHA-specific T cell functionality, providing a rationale for interfering with BTLA signaling in post-stem cell transplantation therapies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Hematopoietic Stem Cell Transplantation , Receptors, Immunologic/physiology , Antibodies, Blocking/physiology , Antibodies, Blocking/therapeutic use , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Epitopes, T-Lymphocyte/metabolism , Gene Targeting/methods , Humans , Immunologic Memory , Minor Histocompatibility Antigens/metabolism , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/therapy , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/immunology , Receptors, Tumor Necrosis Factor, Member 14/physiology , Tumor Cells, Cultured
12.
J Clin Invest ; 121(12): 4810-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22080867

ABSTRACT

Erythropoiesis, the production of red blood cells, must be tightly controlled to ensure adequate oxygen delivery to tissues without causing thrombosis or stroke. Control of physiologic and pathologic erythropoiesis is dependent predominantly on erythropoietin (EPO), the expression of which is regulated by hypoxia-inducible factor (HIF) activity in response to low oxygen tension. Accumulating evidence indicates that oxygen-independent mediators, including inflammatory stimuli, cytokines, and growth factors, also upregulate HIF activity, but it is unclear whether these signals also result in EPO production and erythropoiesis in vivo. Here, we found that signaling through herpesvirus entry mediator (HVEM), a molecule of the TNF receptor superfamily, promoted HIF-1α activity in the kidney and subsequently facilitated renal Epo production and erythropoiesis in vivo under normoxic conditions. This Epo upregulation was mediated by increased production of NO by renal macrophages. Hvem-deficient mice displayed impaired Epo expression and aggravated anemia in response to erythropoietic stress. These data reveal that HVEM signaling functions to promote HIF-1α activity and Epo production, and thus to regulate erythropoiesis. Furthermore, our findings suggest that this molecular mechanism could represent a therapeutic target for Epo-responsive diseases, including anemia.


Subject(s)
Erythropoiesis/physiology , Erythropoietin/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Kidney/metabolism , Receptors, Tumor Necrosis Factor, Member 14/physiology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Hypoxia , Erythropoietin/genetics , Female , Gene Expression Profiling , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/physiology , Oxygen/physiology , Radiation Chimera , Receptors, Tumor Necrosis Factor, Member 14/agonists , Receptors, Tumor Necrosis Factor, Member 14/deficiency , Receptors, Tumor Necrosis Factor, Member 14/genetics , Receptors, Tumor Necrosis Factor, Member 14/immunology , Signal Transduction/physiology
13.
J Exp Med ; 208(4): 797-809, 2011 Apr 11.
Article in English | MEDLINE | ID: mdl-21402741

ABSTRACT

Memory T helper cells (Th cells) play an important role in host defense against pathogens but also contribute to the pathogenesis of inflammatory disorders. We found that a soluble decoy lymphotoxin ß receptor (LT-ßR)-Fc, which can block tumor necrosis factor (TNF)-related ligands LIGHT (TNFSF14) and LT-αß binding to the herpesvirus entry mediator (HVEM) and the LT-ßR, inhibited the accumulation of memory Th2 cells after antigen encounter and correspondingly reduced inflammatory responses in vivo. Showing that this was a function of the receptor for LIGHT, antigen-specific memory CD4 T cells deficient in HVEM were also unable to persist, despite having a normal immediate response to recall antigen. HVEM(-/-) memory Th2 cells displayed reduced activity of PKB (protein kinase B; Akt), and constitutively active Akt rescued their survival and restored strong inflammation after antigen rechallenge. This was not restricted to Th2 memory cells as HVEM-deficient Th1 memory cells were also impaired in surviving after encounter with recall antigen. Furthermore, the absence of LIGHT on T cells recapitulated the defect seen with the absence of HVEM, suggesting that activated T cells communicate through LIGHT-HVEM interactions. Collectively, our results demonstrate a critical role of HVEM signals in the persistence of large pools of memory CD4 T cells.


Subject(s)
Immunologic Memory , Receptors, Tumor Necrosis Factor, Member 14/physiology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Survival , Female , Inflammation/etiology , Lymphotoxin beta Receptor/pharmacology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes, Helper-Inducer/cytology , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology
14.
PLoS One ; 6(1): e16122, 2011 Jan 24.
Article in English | MEDLINE | ID: mdl-21283640

ABSTRACT

Herpes virus entry mediator (HVEM) is one of two principal receptors mediating herpes simplex virus (HSV) entry into murine and human cells. It functions naturally as an immune signaling co-receptor, and may participate in enhancing or repressing immune responses depending on the natural ligand used. To investigate whether engagement of HVEM by HSV affects the in vivo response to HSV infection, we generated recombinants of HSV-2(333) that expressed wild-type gD (HSV-2/gD) or mutant gD able to bind to nectin-1 (the other principal entry receptor) but not HVEM. Replication kinetics and yields of the recombinant strains on Vero cells were indistinguishable from those of wild-type HSV-2(333). After intravaginal inoculation with mutant or wild-type virus, adult female C57BL/6 mice developed vaginal lesions and mortality in similar proportions, and mucosal viral titers were similar or lower for mutant strains at different times. Relative to HSV-2/gD, percentages of HSV-specific CD8(+) T-cells were similar or only slightly reduced after infection with the mutant strain HSV-2/gD-Δ7-15, in all tissues up to 9 days after infection. Levels of HSV-specific CD4(+) T-cells five days after infection also did not differ after infection with either strain. Levels of the cytokine IL-6 and of the chemokines CXCL9, CXCL10, and CCL4 were significantly lower in vaginal washes one day after infection with HSV-2/gD compared with HSV-2/gD-Δ7-15. We conclude that the interaction of HSV gD with HVEM may alter early innate events in the murine immune response to infection, without significantly affecting acute mortality, morbidity, or initial T-cell responses after lethal challenge.


Subject(s)
Chemokines/biosynthesis , Herpesvirus 2, Human/physiology , Mucositis/virology , Receptors, Tumor Necrosis Factor, Member 14/physiology , Vaginal Diseases/virology , Animals , Chemokines/analysis , Female , Herpesvirus 2, Human/genetics , Immunity, Innate , Mice , Mice, Inbred C57BL , Mucositis/immunology , Mutation , T-Lymphocytes/immunology , Vaginal Diseases/immunology
15.
J Leukoc Biol ; 89(4): 517-23, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21106644

ABSTRACT

The HVEM, or TNFRSF14, is a membrane-bound receptor known to activate the NF-κB pathway, leading to the induction of proinflammatory and cell survival-promoting genes. HVEM binds several ligands that are capable of mediating costimulatory pathways, predominantly through its interaction with LIGHT (TNFSF14). However, it can also mediate coinhibitory effects, predominantly by interacting with IGSF members, BTLA or CD160. Therefore, it can function like a "molecular switch" for various activating or inhibitory functions. Furthermore, recent studies suggest the existence of bidirectional signaling with HVEM acting as a ligand for signaling through BTLA, which may act as a ligand in other contexts. Bidirectional signaling, together with new information indicating signaling in cis by cells that coexpress HVEM and its ligands, makes signaling within a HVEM-mediated network complicated, although potentially rich in biology. Accumulating in vivo evidence has shown that HVEM-mediated, coinhibitory signaling may be dominant over HVEM-mediated costimulatory signaling. In several disease models the absence of HVEM-BTLA signaling predominantly resulted in severe mucosal inflammation in the gut and lung, autoimmune-like disease, and impaired immunity during bacterial infection. Here, we will summarize the current view about how HVEM-BTLA signaling is involved in the regulation of mucosal inflammation, autoimmunity, and infection immunity.


Subject(s)
Autoimmunity , Infections , Inflammation , Receptors, Immunologic/physiology , Receptors, Tumor Necrosis Factor, Member 14/physiology , Signal Transduction/physiology , Humans
16.
J Exp Med ; 207(12): 2551-9, 2010 Nov 22.
Article in English | MEDLINE | ID: mdl-21078889

ABSTRACT

Graft-versus-host disease (GVHD) causes significant morbidity and mortality in allogeneic hematopoietic stem cell transplantation (aHSCT), preventing its broader application to non-life-threatening diseases. We show that a single administration of a nondepleting monoclonal antibody specific for the coinhibitory immunoglobulin receptor, B and T lymphocyte associated (BTLA), permanently prevented GVHD when administered at the time of aHSCT. Once GVHD was established, anti-BTLA treatment was unable to reverse disease, suggesting that its mechanism occurs early after aHSCT. Anti-BTLA treatment prevented GVHD independently of its ligand, the costimulatory tumor necrosis factor receptor herpesvirus entry mediator (HVEM), and required BTLA expression by donor-derived T cells. Furthermore, anti-BTLA treatment led to the relative inhibition of CD4(+) forkhead box P3(-) (Foxp3(-)) effector T cell (T eff cell) expansion compared with precommitted naturally occurring donor-derived CD4(+) Foxp3(+) regulatory T cell (T reg cell) and allowed for graft-versus-tumor (GVT) effects as well as robust responses to pathogens. These results suggest that BTLA agonism rebalances T cell expansion in lymphopenic hosts after aHSCT, thereby preventing GVHD without global immunosuppression. Thus, targeting BTLA with a monoclonal antibody at the initiation of aHSCT therapy might reduce limitations imposed by histocompatibility and allow broader application to treatment of non-life-threatening diseases.


Subject(s)
Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Immunosuppression Therapy , Receptors, Immunologic/physiology , Animals , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Listeriosis/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Immunologic/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Member 14/physiology , T-Lymphocytes, Regulatory/immunology
17.
J Immunol ; 185(3): 1949-58, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20592286

ABSTRACT

The TNF superfamily member homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for herpesvirus entry mediator (HVEM), a receptor expressed by T lymphocytes (LIGHT) [TNF superfamily (SF)-14], is a key cytokine that activates T cells and dendritic cells and is implicated as a mediator of inflammatory, metabolic, and malignant diseases. LIGHT engages the lymphotoxin-beta receptor (LTbetaR) and HVEM (TNFRSF14), but is competitively limited in activating these receptors by soluble decoy receptor-3 (DcR3; TNFRSF6B). Two variants in the human LIGHT alter the protein at E214K (rs344560) in the receptor-binding domain and S32L (rs2291667) in the cytosolic domain; however, the functional impact of these polymorphisms is unknown. A neutralizing Ab failed to bind the LIGHT-214K variant, indicating this position as a part of the receptor-binding region. Relative to the predominant reference variant S32/E214, the other variants showed altered avidity with LTbetaR and less with HVEM. Heterotrimers of the LIGHT variants decreased binding avidity to DcR3 and minimized the inhibitory effect of DcR3 toward LTbetaR-induced activation of NF-kappaB. In patients with immune-mediated inflammatory diseases, such as rheumatoid arthritis, DcR3 protein levels were significantly elevated. Immunohistochemistry revealed synoviocytes as a significant source of DcR3 production, and DcR3 hyperexpression is controlled by posttranscriptional mechanisms. The increased potential for LTbetaR signaling, coupled with increased bioavailability due to lower DcR3 avidity, provides a mechanism of how polymorphic variants in LIGHT could contribute to the pathogenesis of inflammatory diseases.


Subject(s)
Genetic Variation/immunology , Polymorphism, Single Nucleotide/immunology , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Amino Acid Sequence , Biological Availability , Coculture Techniques , HeLa Cells , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Models, Immunological , Molecular Sequence Data , Mutagenesis, Site-Directed , NF-kappa B/antagonists & inhibitors , Protein Binding/genetics , Protein Binding/immunology , Receptors, Tumor Necrosis Factor, Member 14/physiology , Receptors, Tumor Necrosis Factor, Member 6b/physiology , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology
18.
J Immunol ; 184(10): 5475-84, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20410485

ABSTRACT

Immune responses diminish with age resulting in an increased susceptibility of the elderly to infectious agents and an inability to mount protective immune responses to vaccines. Immunosenescence affects multiple aspects of the immune system, including CD8(+) T cells, which control viral infections and are assumed to prevent the development of cancers. In this study, we tested if CD8(+) T cell responses in aged mice could be enhanced through a vaccine that concomitantly expresses Ag and a molecule that blocks an immunoinhibitory pathway. Specifically, we tested a vaccine based on a replication-defective chimpanzee-derived adenovirus vector expressing the nucleoprotein (NP) of influenza A virus as a fusion protein with the HSV type 1 glycoprotein D, which through binding to the herpes virus entry mediator, blocks the immunoinhibitory herpes virus entry mediator B and T lymphocyte attenuator/CD160 pathways. Our results show that the vaccine expressing a fusion protein of NP and glycoprotein D induces significantly higher NP-specific CD8(+) T cell responses in young and aged mice compared with the vaccine expressing NP only.


Subject(s)
Aging/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Influenza A virus/immunology , Receptors, Tumor Necrosis Factor, Member 14/antagonists & inhibitors , Signal Transduction/immunology , Up-Regulation/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Line , Epitopes, T-Lymphocyte/genetics , Female , Genetic Vectors/immunology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Humans , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Mice , Mice, Inbred C57BL , Nucleocapsid Proteins , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Receptors, Immunologic/genetics , Receptors, Tumor Necrosis Factor, Member 14/physiology , Recombinant Fusion Proteins/immunology , Signal Transduction/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/antagonists & inhibitors , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology , Viral Core Proteins/genetics , Viral Core Proteins/immunology
19.
J Clin Invest ; 120(1): 157-67, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20038811

ABSTRACT

The function of antigen-specific CD8+ T cells, which may protect against both infectious and malignant diseases, can be impaired by ligation of their inhibitory receptors, which include CTL-associated protein 4 (CTLA-4) and programmed cell death 1 (PD-1). Recently, B and T lymphocyte attenuator (BTLA) was identified as a novel inhibitory receptor with structural and functional similarities to CTLA-4 and PD-1. BTLA triggering leads to decreased antimicrobial and autoimmune T cell responses in mice, but its functions in humans are largely unknown. Here we have demonstrated that as human viral antigen-specific CD8+ T cells differentiated from naive to effector cells, their surface expression of BTLA was gradually downregulated. In marked contrast, human melanoma tumor antigen-specific effector CD8+ T cells persistently expressed high levels of BTLA in vivo and remained susceptible to functional inhibition by its ligand herpes virus entry mediator (HVEM). Such persistence of BTLA expression was also found in tumor antigen-specific CD8+ T cells from melanoma patients with spontaneous antitumor immune responses and after conventional peptide vaccination. Remarkably, addition of CpG oligodeoxynucleotides to the vaccine formulation led to progressive downregulation of BTLA in vivo and consequent resistance to BTLA-HVEM-mediated inhibition. Thus, BTLA activation inhibits the function of human CD8+ cancer-specific T cells, and appropriate immunotherapy may partially overcome this inhibition.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Melanoma/therapy , Neoplasm Proteins/immunology , Receptors, Immunologic/physiology , Vaccination , Animals , Antigens, CD/physiology , Apoptosis Regulatory Proteins/physiology , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Humans , Interferon-gamma/biosynthesis , Lymphocyte Activation , MART-1 Antigen , Melanoma/immunology , Oligodeoxyribonucleotides/pharmacology , Programmed Cell Death 1 Receptor , Receptors, Tumor Necrosis Factor, Member 14/physiology
20.
J Virol ; 84(4): 2038-46, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20007280

ABSTRACT

Of the four required herpes simplex virus (HSV) entry glycoproteins, the precise role of gH-gL in fusion remains the most elusive. The heterodimer gH-gL has been proposed to mediate hemifusion after the interaction of another required glycoprotein, gD, with a receptor. To identify functional domains of HSV-1 gH, we generated 22 randomized linker-insertion mutants. Analyses of 22 gH mutants revealed that gH is relatively tolerant of insertion mutations, as 15 of 22 mutants permitted normal processing and transport of gH-gL to the cell surface. gH mutants that were not expressed well at the cell surface did not function in fusion or viral entry. The screening of gH mutants for function revealed the following: (i) for wild-type gH and some gH mutants, fusion with nectin-1-expressing target cells occurred more rapidly than with herpesvirus entry mediator (HVEM)-expressing target cells; (ii) some gH mutants reduced the rate of cell fusion without abrogating fusion completely, indicating that gH may play a role in governing the kinetics of fusion and may be responsible for a rate-limiting first stage in HSV-1 fusion; and (iii) only one gH mutant, located within the short cytoplasmic tail, completely abrogated function, indicating that the gH cytoplasmic tail is crucial for cell fusion and viral infectivity.


Subject(s)
Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/physiology , Virus Internalization , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Genetic Complementation Test , Herpesvirus 1, Human/pathogenicity , Humans , Kinetics , Mutagenesis, Insertional , Protein Structure, Tertiary , Receptors, Tumor Necrosis Factor, Member 14/genetics , Receptors, Tumor Necrosis Factor, Member 14/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Vero Cells , Viral Envelope Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...