Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 12(9): e0184915, 2017.
Article in English | MEDLINE | ID: mdl-28910387

ABSTRACT

Dendritic cells (DC) are a promising cell type for cancer vaccines due to their high immunostimulatory capacity. However, improper maturation of DC prior to treatment may account for the limited efficacy of DC vaccine clinical trials. Latent Membrane Protein-1 (LMP1) of Epstein-Barr virus was examined for its ability to mature and activate DC as a gene-based molecular adjuvant for DC vaccines. DC were transduced with an adenovirus 5 vector (Ad5) expressing LMP1 under the control of a Tet-inducible promoter. Ad5-LMP1 was found to mature and activate both human and mouse DC. LMP1 enhanced in vitro migration of DC toward CCL19, as well as in vivo migration of DC to the inguinal lymph nodes of mice following intradermal injection. LMP1-transduced DC increased T cell proliferation in a Pmel-1 adoptive transfer model and enhanced survival in B16-F10 melanoma models. LMP1-DC also enhanced protection in a vaccinia-Gag viral challenge assay. LMP1 induced high levels of IL-12p70 secretion in mouse DC when compared to standard maturation protocols. Importantly, LMP1-transduced human DC retained the capacity to secrete IL-12p70 and TNF in response to DC restimulation. In contrast, DC matured with Monocyte Conditioned Media-Mimic cocktail (Mimic) were impaired in IL-12p70 secretion following restimulation. Overall, LMP1 matured and activated DC, induced migration to the lymph node, and generated high levels of IL-12p70 in a murine model. We propose LMP1 as a promising molecular adjuvant for DC vaccines.


Subject(s)
Dendritic Cells/transplantation , Herpesvirus 4, Human/metabolism , Interleukin-12/metabolism , Lymph Nodes/immunology , Melanoma, Experimental/therapy , Viral Matrix Proteins/genetics , Animals , Cancer Vaccines/immunology , Cell Movement , Chemokine CCL19/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Dependovirus/genetics , Dependovirus/physiology , Female , HEK293 Cells , Humans , Injections, Intradermal , Melanoma, Experimental/immunology , Mice , Viral Matrix Proteins/immunology
2.
Anticancer Res ; 34(11): 6431-41, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25368243

ABSTRACT

BACKGROUND/AIM: Interleukin-2 (IL2) transgenic Ewing sarcoma cells reduce tumor growth in vivo and in vitro. In the present study we analyzed the expression of immune suppressive indoleamine-2,3-dioxygenase (IDO) in this model. MATERIALS AND METHODS: Expression of IDO was analyzed by polymerase chain reaction. The impact of the cluster of differentiation 137 (CD137)/CD137 ligand (CD137L) co-stimulatory system on expression of IDO and different cytokines was analyzed both in vivo and in vitro. RESULTS: Tumors that developed in vivo in the presence of IL2 transgenic tumor cells expressed IDO. The presence of CD137L transgenic tumor cells led to down-regulation of IDO. Further in-vitro analysis of this phenomenon indicated that IDO was expressed in tumor cells as a consequence of interferon-gamma produced by lymphocytes in response to IL2. Depending on the concentration of IL2, stimulation of CD137 increased or reduced cytokine production in lymphocytes. CONCLUSION: Our data indicate that the CD137/CD137L pathway can modulate the immune response against Ewing sarcoma cells.


Subject(s)
Bone Neoplasms/therapy , Immunotherapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/pharmacology , Interleukin-2/physiology , Sarcoma, Ewing/therapy , Animals , Bone Neoplasms/enzymology , Bone Neoplasms/immunology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Flow Cytometry , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Lymphocytes/enzymology , Lymphocytes/immunology , Lymphocytes/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , OX40 Ligand/genetics , OX40 Ligand/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Ewing/enzymology , Sarcoma, Ewing/immunology , Tumor Cells, Cultured , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Xenograft Model Antitumor Assays
3.
Retrovirology ; 8: 39, 2011 May 18.
Article in English | MEDLINE | ID: mdl-21592361

ABSTRACT

BACKGROUND: Molecular adjuvants are a promising method to enhance virus-specific immune responses and protect against HIV-1 infection. Immune activation by ligands for receptors such as CD40 can induce dendritic cell activation and maturation. Here we explore the incorporation of two CD40 mimics, Epstein Barr Virus gene LMP1 or an LMP1-CD40 chimera, into a strain of SIV that was engineered to be limited to a single cycle of infection. RESULTS: Full length LMP1 or the chimeric protein LMP1-CD40 was cloned into the nef-locus of single-cycle SIV. Human and Macaque monocyte derived macrophages and DC were infected with these viruses. Infected cells were analyzed for activation surface markers by flow cytometry. Cells were also analyzed for secretion of pro-inflammatory cytokines IL-1ß, IL-6, IL-8, IL-12p70 and TNF by cytometric bead array. CONCLUSIONS: Overall, single-cycle SIV expressing LMP1 and LMP1-CD40 produced a broad and potent T(H)1-biased immune response in human as well as rhesus macaque macrophages and DC when compared with control virus. Single-cycle SIV-LMP1 also enhanced antigen presentation by lentiviral vector vaccines, suggesting that LMP1-mediated immune activation may enhance lentiviral vector vaccines against HIV-1.


Subject(s)
Adjuvants, Immunologic/pharmacology , Drug Carriers , Genetic Vectors , Lentivirus/genetics , SAIDS Vaccines/immunology , Viral Matrix Proteins/pharmacology , Adjuvants, Immunologic/genetics , Animals , Antigen Presentation , Cytokines/metabolism , Humans , Macaca mulatta , Monocytes/virology , Vaccines, Synthetic/immunology , Viral Matrix Proteins/genetics
4.
J Leukoc Biol ; 90(2): 389-98, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21586676

ABSTRACT

HIV-1 does not significantly activate cellular immunity, which has made it difficult to use attenuated forms of HIV-1 as a vaccine. In contrast, EBV induces robust T cell responses in most infected individuals, perhaps as this virus contains LMP1, a viral mimic of CD40, which is a key activating molecule for DCs and macrophages. Consequently, studies were conducted using LMP1 and LMP1-CD40, a related construct formed by replacing the intracellular signaling domain of LMP1 with that of CD40. Upon electroporation into DCs, LMP1 and LMP1-CD40 mRNAs were sufficient to up-regulate costimulatory molecules and proinflammatory cytokines, indicating that these molecules can function in isolation as adjuvant-like molecules. As a first step toward an improved HIV vaccine, LMP1 and LMP1-CD40 were introduced into a HIV-1 construct to produce virions encoding these proteins. Transduction of DCs and macrophages with these viruses induced morphological changes and up-regulated costimulatory molecules and cytokine production by these cells. HIV-LMP1 enhanced the antigen-presenting function of DCs, as measured in an in vitro immunization assay. Taken together, these data show that LMP1 and LMP1-CD40 are portable gene cassettes with strong adjuvant properties that can be introduced into viruses such as HIV, which by themselves, are insufficient to induce protective cellular immunity.


Subject(s)
AIDS Vaccines/immunology , Dendritic Cells/immunology , Herpesvirus 4, Human/immunology , Viral Matrix Proteins/immunology , Adjuvants, Immunologic/therapeutic use , Antigen Presentation , CD40 Antigens/therapeutic use , HIV/genetics , HIV/immunology , Herpesvirus 4, Human/chemistry , Herpesvirus 4, Human/genetics , Humans , Molecular Mimicry/immunology , Transduction, Genetic , Viral Matrix Proteins/therapeutic use
5.
Vaccine ; 29(11): 2110-9, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21241732

ABSTRACT

BACKGROUND: Dendritic cell (DC) therapy is a promising technology for the treatment of HIV infected individuals. HIV-1 Gag- and Nef RNA-loaded DC have previously been shown to induce immune responses ex vivo following coculture with autologous lymphocytes. However, polyfunctionality and memory responses following coculture have not been evaluated. In addition, little is known regarding whether specific HIV-1 proteome components, such as highly conserved regions of the HIV-1, could enhance clinical responses following DC therapy. METHODOLOGY AND PRINCIPAL FINDINGS: To determine the breadth of the immune responses to antigen loaded DC, we analyzed polyfunctional T cell response ex vivo to Gag RNA loaded DC. Blood samples were used to generate monocyte derived DC, which were then matured and cocultured with autologous lymphocytes. We found that cytokine-matured DC loaded with Gag RNA was able to induce Gag-specific IFN-γ and IL-2 responses after a 12-day coculture. We characterized these responses by polyfunctional intracellular cytokine staining and evaluation of T cell memory phenotypes. Central memory CD8+ T cells were induced ex vivo after DC coculture from each of 3 patients, and the effector memory pool was increased by DC coculture from 2 patients. We also observed a decrease in the terminal effector and intermediate CD8+ T cell pool and an increase in the naïve/other population. There was a reduction in terminal effector and intermediate CD4+ T cells, and a corresponding increase in naïve/other CD4+ T cells. Finally, we evaluated conserved regions of Gag as a novel DC therapy immunogen and found that a conserved element (CE) p24 Gag antigen elicited IFN-γ and IL-2 responses comparable to those induced by a full-length Gag antigen. CONCLUSIONS: We showed that RNA-loaded DC therapy induced a polyfunctional T cell response ex vivo, supporting the use of such DC-therapy for HIV infection. However, the central and effector memory phenotypes of T cells did not appear to be enhanced during coculture with Gag RNA-loaded DC. Furthermore, comparable antigen-specific responses were induced in HIV infected individuals using full-length Gag or only conserved elements of the Gag p24 protein. This indicates that immune responses can be focused onto the conserved elements of Gag in the absence of other Gag components.


Subject(s)
AIDS Vaccines/immunology , Dendritic Cells/immunology , HIV Infections/therapy , gag Gene Products, Human Immunodeficiency Virus/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Core Protein p24/immunology , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , Humans , Immunologic Memory , Interferon-gamma/immunology , Interleukin-2/immunology , RNA, Viral/genetics
6.
J Immunol ; 184(9): 4770-8, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20351189

ABSTRACT

Agonistic anti-CD137 mAbs either positively or negatively regulate T cell function. When administered at the beginning of lymphocytic choriomeningitis virus Armstrong infection anti-CD137 induced immunosuppression and T cell deletion, and in the case of influenza infection led to increased mortality. In contrast, 72 h delay in anti-CD137 treatment led to an enhanced virus-specific CD8 T cell response and rapid viral clearance. Virus-specific CD8 T cells in anti-CD137-injected mice rapidly upregulate Fas expression, and although necessary, was insufficient to induce CD8 T cell deletion. Strikingly, CD137 signaling in T cells was found to be insufficient to induce suppression or deletion. Rather, immunosuppression and T cell deletion was only observed if CD137 signals were provided to T cells and dendritic cells (DCs). In vitro CD137 crosslinking in DCs led to phosphorylation of Stat3, and importantly, anti-CD137 treatment of lymphocytic choriomeningitis virus Armstrong infected Stat3 conditional knock-out mice induced neither immune suppression or T cell deletion. Taken together, these data suggest that CD137 signaling in DCs can regulate CD8 T cell survival through a Stat3 and Fas-mediated pathway.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lymphocyte Activation/immunology , STAT3 Transcription Factor/physiology , Tumor Necrosis Factor Receptor Superfamily, Member 9/physiology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/virology , Cell Death/genetics , Cell Death/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Dendritic Cells/virology , Epitopes, T-Lymphocyte/immunology , Fas Ligand Protein , Female , Lymphocyte Activation/genetics , Lymphocyte Depletion , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/deficiency , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , fas Receptor/physiology
7.
J Clin Invest ; 117(10): 3029-41, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17853940

ABSTRACT

CD137 is expressed on activated T cells and ligands to this costimulatory molecule have clinical potential for amplifying CD8 T cell immunity to tumors and viruses, while suppressing CD4 autoimmune T cell responses. To understand the basis for this dichotomy in T cell function, CD4 and CD8 antiviral immunity was measured in lymphocytic choriomeningitis virus (LCMV) Armstrong- or A/PR8/34 influenza-infected mice injected with anti-CD137 mAbs. We found that the timing of administration of anti-CD137 mAbs profoundly altered the nature of the antiviral immune response during acute infection. Antiviral immunity progressed normally for the first 72 hours when the mAb was administered early in infection before undergoing complete collapse by day 8 postinfection. Anti-CD137-injected LCMV-infected mice became tolerant to, and persistently infected with, LCMV Armstrong. Elevated levels of IL-10 early in the response was key to the loss of CD4(+) T cells, whereas CD8(+) T cell deletion was dependent on a prolonged TNF-alpha response, IL-10, and upregulation of Fas. Blocking IL-10 function rescued CD4 antiviral immunity but not CD8(+) T cell deletion. Anti-CD137 treatment given beyond 72 hours after infection significantly enhanced antiviral immunity. Mice treated with anti-CD137 mAb 1 day before infection with A/PR8/34 virus experienced 80% mortality compared with 40% mortality of controls. When treatment was delayed until day 1 postinfection, 100% of the infected mice survived. These data show that anti-CD137 mAbs can induce T cell activation-induced cell death or enhance antiviral immunity depending on the timing of treatment, which may be important for vaccine development.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunosuppression Therapy , Influenza, Human/immunology , Lymphocytic Choriomeningitis/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Humans , Interleukin-10/metabolism , Mice , Mice, Mutant Strains , Time Factors , Tumor Necrosis Factor Receptor Superfamily, Member 9/analysis , Tumor Necrosis Factor-alpha/metabolism
8.
J Immunol ; 178(7): 4194-213, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17371976

ABSTRACT

CD137-mediated signals costimulate T cells and protect them from activation-induced apoptosis; they induce curative antitumor immunity and enhance antiviral immune responses in mice. In contrast, anti-CD137 agonistic mAbs can suppress T-dependent humoral immunity and reverse the course of established autoimmune disease. These results have provided a rationale for assessing the therapeutic potential of CD137 ligands in human clinical trials. In this study, we report that a single 200-mug injection of anti-CD137 given to otherwise naive BALB/c or C57BL/6 mice led to the development of a series of immunological anomalies. These included splenomegaly, lymphadenopathy, hepatomegaly, multifocal hepatitis, anemia, altered trafficking of B cells and CD8 T cells, loss of NK cells, and a 10-fold increase in bone marrow (BM) cells bearing the phenotype of hemopoietic stem cells. These events were dependent on CD8 T cells, TNF-alpha, IFN-gamma, and type I IFNs. BM cells up-regulated Fas, and there was a significant increase in the number of CD8+ T cells that correlated with a loss of CD19+ and Ab-secreting cells in the BM. TCR Valphabeta usage was random and polyclonal among liver-infiltrating CD8 T cells, and multifocal CD8+ T cell infiltrates were resolved upon termination of anti-CD137 treatment. Anti-CD137-treated mice developed lymphopenia, thrombocytopenia, and anemia, and had lowered levels of hemoglobin and increased numbers of reticulocytes.


Subject(s)
Cytokines/metabolism , Hematologic Diseases/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Anemia/immunology , Animals , Antibodies, Monoclonal/toxicity , Cell Movement , Cell Proliferation , Female , Hematopoiesis/immunology , Liver/immunology , Liver/pathology , Lung/immunology , Lung/pathology , Lymph Nodes/cytology , Lymphocyte Activation , Lymphocytes/immunology , Lymphopenia/immunology , Mice , Mice, Inbred Strains , Receptors, Antigen, T-Cell/immunology , Spleen/immunology , Spleen/pathology , Thrombocytopenia/immunology
9.
Immunol Res ; 29(1-3): 197-208, 2004.
Article in English | MEDLINE | ID: mdl-15181282

ABSTRACT

CD137 (4-1BB), is an inducible T-cell costimulatory receptor and a member of the tumor necrosis factor receptor (TNFR) superfamily. It is expressed on activated T cells and activated natural killer (NK) cells, but is constitutively expressed on a population of splenic dendritic cells (DCs). The natural counter receptor for CD137 is 4-1BB ligand, a member of the TNF superfamily that is weakly expressed on naïve or resting B cells, macrophages, and DCs. Upon activation, the level of 4-1BBL expression increases on these cells. In T cells CD137-induced signals lead to the recruitment of TRAF family members and activation of several kinases, including ASK-1, MKK, MAPK3/ MAPK4, p38, and JNK/SAPK. Kinase activation is then followed by the activation and nuclear translocation of several transcription factors, including ATF-2, Jun, and NF-kappaB. CD137-mediated T-cell costimulation as measured by enhanced proliferation and cytokine production can be induced by anti-CD137 monoclonal antibodies (MAbs) or by employing immobilized 4-1BB ligand. In addition to augmenting suboptimal TCR-induced proliferation, CD137-mediated signaling protects T cells, and in particular, CD8+ T cells from activation-induced cell death (AICD). Although studies with CD137-deficient or 4-1BBL-deficient mice failed to demonstrate any loss of essential immunological function, or other noteworthy deficits, we have found that 4-1BBL-deficient mice failed to generate a strong antiviral immune response following lymphocytic choriomeningitis virus (LCMV) peptide vaccination. We further found that although compromised, the immune response to LCMV vaccination in these mice could be fully restored by injecting them with anti-CD137 MAbs at the time of vaccination. Finally, we have found that injecting normal mice with anti-CD137 MAbs had profound effects on their ability to develop immune responses to allo- and autoantigens. The results of these studies discussed in this article provide a rationale for assessing the potential use of anti-CD137 MAbs for therapeutic purposes.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Neoplasms/drug therapy , Receptors, Nerve Growth Factor/antagonists & inhibitors , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Antigens, CD , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Clonal Anergy/immunology , Disease Models, Animal , Humans , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred Strains , Neoplasm Transplantation/immunology , Neoplasms/immunology , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9
SELECTION OF CITATIONS
SEARCH DETAIL
...