Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Leukoc Biol ; 72(3): 546-53, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12223523

ABSTRACT

The ligand for the receptor tyrosine kinase fms-like tyrosine kinase 3 (Flt3L) is a growth factor for hematopoietic progenitors and induces expansion of the two distinct lineages of dendritic cells (DC) that have been described in humans. These two lineages, DC1 and DC2, have been described according to their ability to induce naive T cell differentiation to T helper cell type 1 (Th1) and Th2 effector cells, respectively. The immunoregulatory potential of DC1 and DC2 depends on their state of maturation and activation, which can be mediated by several molecules. Because monocyte-derived DC1 produce interleukin-12 (IL-12) when stimulated with CD40 ligand (CD40L), we hypothesized that similar results would be obtained with DC1 mobilized by Flt3L. Unexpectedly, we found that immature DC expanded in vivo by Flt3L treatment could not be stimulated to produce IL-12 in vitro using CD40L and/or interferon-gamma (IFN-gamma) alone. Instead, we found that Flt3L-mobilized DC from cancer patients require a sequence of specific signals for maturation, which included initial treatment with granulocyte macrophage-colony stimulating factor followed by a combination of maturation signals such as CD40L and IFN-gamma. Flt3L-mobilized DC matured in this manner possessed greater T cell-stimulatory function than nonmatured DC. The ability to generate phenotypically mature, IL-12-producing DC1 from peripheral blood mononuclear cells mobilized by Flt3L will have important implications for the development of effective cancer immunotherapy strategies.


Subject(s)
Antigens, CD , Cell Adhesion Molecules , Dendritic Cells/drug effects , Interleukin-12/biosynthesis , Lectins, C-Type , Membrane Proteins/pharmacology , Signal Transduction/drug effects , Breast Neoplasms/pathology , CD27 Ligand , CD40 Ligand/pharmacology , Cell Lineage , Colonic Neoplasms/pathology , Dendritic Cells/classification , Dendritic Cells/cytology , Female , Gene Expression Regulation/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunoglobulins/biosynthesis , Immunoglobulins/genetics , Interferon-gamma/pharmacology , Interleukin-12/genetics , Lectins/biosynthesis , Lectins/genetics , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Receptors, Chemokine/biosynthesis , Receptors, Chemokine/genetics , Recombinant Proteins , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , CD83 Antigen
2.
Cancer Biol Ther ; 2(5): 579-86, 2003.
Article in English | MEDLINE | ID: mdl-14614331

ABSTRACT

Inadequate costimulation by solid tumors is generally believed to induce immune tolerance during primary tumor growth. We looked for tumor-specific immunity vs. tolerance in patients with Ewing's sarcoma. Circulating T cells from patients with progressively growing Ewing's tumors displayed MHC restricted tumor-induced proliferation and robust tumor lysis. Tumor-reactive T cells reside within the memory CD3+CD8+ subset and are CD28-/4-1BB+. Autologous Ewing's tumors expressed 4-1BBL, and tumor-induced T cell proliferation and activation required costimulation by 4-1BBL. Stimulation of PBL with anti-CD3/4-1BBL, but not anti-CD3/anti-CD28 induced tumor lytic effectors. Similarly, in a xenograft model, anti-CD3/4-1BBL expanded T cells controlled primary growth and prevented metastasis of autologous tumors while nonactivated and anti-CD3/anti-CD28 activated CD8+ cells did not. These results question prevailing models of tumor induced tolerance accompanying progressive tumor growth; rather, we show coexistence of progressive tumor growth and anti-tumor immunity, with costimulation provided by the tumor itself. They further demonstrate a potential new therapeutic role for 4-1BBL mediated costimulation in expanding tumor reactive CTLs for use in the adoptive immunotherapy of cancer.


Subject(s)
Bone Neoplasms/immunology , Lymphocyte Activation , Sarcoma, Ewing/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/metabolism , 4-1BB Ligand , Adolescent , Adult , Animals , Bone Neoplasms/prevention & control , CD28 Antigens/metabolism , CD3 Complex/metabolism , CD8 Antigens/metabolism , Dendritic Cells/immunology , Female , Humans , Ligands , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Mice , Mice, SCID , Receptors, Antigen, T-Cell/metabolism , Sarcoma, Ewing/prevention & control
3.
Immunol Lett ; 81(1): 13-24, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11841841

ABSTRACT

We have constructed a DNA plasmid vaccine encoding the C-terminal 42-kDa region of the merozoite surface protein 1 (pMSP1(42)) from the 3D7 strain of Plasmodium falciparum (Pf3D7). This plasmid expressed recombinant MSP1(42) after in vitro transfection in mouse VM92 cells. Rhesus monkeys immunized with pMSP1(42) produced antibodies reactive with Pf3D7 infected erythrocytes by IFAT, and by ELISA against yeast produced MSP1(19) (yMSP1(19)). Immunization also induced antigen specific T cell responses as measured by interferon-gamma production, and by classical CTL chromium release assays. In addition, immunization with pMSP1(42) primed animals for an enhanced antibody response to a subsequent boost with the recombinant yMSP1(19). We also evaluated Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) as an adjuvant for pMSP1(42.) We tested both rhesus GM-CSF expressed from a DNA plasmid, and E. coli produced recombinant human GM-CSF. Plasmids encoding rhesus GM-CSF (prhGM-CSF) and human GM-CSF (phuGM-CSF) were constructed; these plasmids expressed bio-active recombinant GMCSF. Co-immunization with a mixture of prhGM-CSF and pMSP1(42) induced higher specific antibody responses after the first dose of plasmid, but after three doses of DNA monkeys immunized with or without prhGM-CSF had the same final antibody titers and T cell responses. In comparison, rhuGM-CSF protein did not lead to accelerated antibody production after the first DNA dose. However, antibody titers were maintained at a slightly higher level in monkeys receiving GM-CSF protein, and they had a higher response to boosting with recombinant MSP1(19). The GM-CSF plasmid or protein appears to be less potent as an adjuvant in rhesus monkeys than each is in mice, and more work is needed to determine if GM-CSF can be a useful adjuvant in DNA vaccination of primates.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Malaria Vaccines , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Vaccines, DNA , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Protozoan/immunology , Cytotoxicity, Immunologic , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interferon-gamma/immunology , Macaca mulatta , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Merozoite Surface Protein 1/genetics , Merozoite Surface Protein 1/immunology , Plasmids , Plasmodium falciparum/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology
4.
J Immunol ; 176(6): 3753-9, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16517744

ABSTRACT

Virus-specific CD4+ T cell help and CD8+ cytotoxic T cell responses are critical for maintenance of effective immunity in chronic viral infections. The importance of CD4+ T cells has been documented in HIV infection. To investigate whether a stronger CD4+ T cell response can be induced by modifications to enhance the T1 epitope, the first CD4+ T cell epitope discovered in HIV-1-gp120, we developed a T1-specific CD4+ T cell line from a healthy volunteer immunized with a canarypox vector expressing gp120 and boosted with recombinant gp120. This T1-specific CD4+ T cell line was restricted to DR13, which is common in U.S. Caucasians and African-Americans and very frequent in Africans. Peptides with certain amino acid substitutions in key positions induced enhanced specific CD4+ T cell proliferative responses at lower peptide concentration than the original epitope. This relatively conserved CD4 epitope improved by the epitope enhancement strategy could be a component of a more effective second generation vaccine construct for HIV infection.


Subject(s)
AIDS Vaccines/chemistry , AIDS Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , HIV-1/immunology , Alanine/analogs & derivatives , Alanine/immunology , Amino Acid Sequence , Amino Acid Substitution , CD5 Antigens/immunology , Cell Proliferation , Cells, Cultured , HIV-1/chemistry , Humans , Interferon-gamma/biosynthesis , Molecular Sequence Data
5.
Oncology ; 70(4): 245-54, 2006.
Article in English | MEDLINE | ID: mdl-17047396

ABSTRACT

Flt3 ligand (Flt3L) therapy that expands dendritic cells in vivo in combination with local tumor radiotherapy (RT) significantly improved survival and induced a long-term tumor-specific immune response in a murine model of Lewis lung carcinoma (3LL). The irradiated tumor cells were able to significantly restimulate the splenocytes of the RT + Flt3L cohort in vitro. The restimulated splenocytes demonstrated increased cytotoxic response, lymphocytic proliferation and elevated levels of Th type I cytokines (IL-2, IL-12, IFN-gamma and TNF-alpha). The combination therapy of RT + Flt3L induced a long-term protective immunity in the disease-free animals. The protective effect was further enhanced when the disease-free animals were vaccinated with irradiated tumor cells. The vaccinated animals had significantly greater protection compared to the nonvaccinated group against subsequent challenge with 3LL cells. Taken together, these results indicate that the release of tumor antigens by irradiated dying tumors and concomitant administration of Flt3L was able to facilitate the generation of a tumor-specific long-term immune response against a poorly immunogenic tumor. This effect was further boosted by vaccination with irradiated tumor cells.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antineoplastic Agents/therapeutic use , Cancer Vaccines , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/therapy , Membrane Proteins/therapeutic use , Animals , Carcinoma, Lewis Lung/pathology , Combined Modality Therapy , Cytokines/blood , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology
6.
Blood ; 103(7): 2699-704, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14684414

ABSTRACT

The CD8+ cell noncytotoxic anti-HIV response (CNAR) is associated with a long-term healthy clinical state in HIV-infected individuals. Over time CNAR is reduced concomitant with progression to disease. In studies to evaluate whether the interaction between CD8+ cells and dendritic cells (DCs) could increase CNAR, CD8+ cells from individuals who showed a decrease in this antiviral activity were cocultured with monocyte-derived dendritic cells matured with CD40 ligand. After coculture with these mature DCs, the CD8+ cells showed an increase in CNAR greater than that observed with CD8+ cells costimulated with CD3/CD28 antibodies. This antiviral response appeared to be mediated primarily by production of interleukin-15 (IL-15) by the mature DCs. Purified IL-15 also enhanced CNAR, whereas IL-12 showed no substantial effect. These studies provide another potential approach by which the immune system in HIV infection could be restored by cytokine therapy, particularly IL-15 administration.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , HIV Infections/immunology , Interleukin-15/immunology , CD3 Complex/immunology , CD8 Antigens/immunology , Cytokines/immunology , Cytokines/therapeutic use , Disease Progression , Humans , Interleukin-15/therapeutic use , Reference Values
7.
J Immunol ; 172(6): 3725-35, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15004177

ABSTRACT

Dendritic cells (DCs) reside in tissues, where they function as sentinels, providing an essential link between innate and adaptive immunity. Increasing the numbers of DCs in vivo augments T cell responses, and can cause dramatic CTL-dependent tumor regression. To determine whether greater DC numbers promoted T cell-mediated protection in the context of host defense against intracellular bacteria, we treated mice with Flt3 ligand (Flt3-L) to increase DCs in vivo and challenged them with Listeria monocytogenes. Unexpectedly, after primary challenge with Listeria, the overall control of Listeria infection was impaired in Flt3-L-treated mice, which had greater bacterial burden and mortality than controls. Similar results were obtained when DC numbers were increased by treatment with polyethylene glycol-conjugated GM-CSF rather than Flt3-L and in mice infected with Mycobacterium tuberculosis. Impaired protection was not due to dysfunctional T cell responses, as Flt3-L-treated mice had a greater frequency and absolute number of Ag-specific CD8+ T cells, which produced IFN-gamma, exhibited cytolytic activity, and transferred protection. The increased Listeria burden in Flt3-L-treated mice was preferentially associated with DCs, which were unable to kill Listeria and more resistant to CTL lysis compared with macrophages in vitro. Although we cannot exclude the possibility that other potential effects, in addition to increased numbers of DCs, are shared by Flt3-L and polyethylene glycol-conjugated GM-CSF and contributed to the increase in susceptibility observed in treated mice, these results support the notion that DC numbers must be properly controlled within physiological limits to optimize host defense to intracellular bacterial pathogens.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Epitopes, T-Lymphocyte/immunology , Intracellular Fluid/immunology , Intracellular Fluid/microbiology , Adjuvants, Immunologic/administration & dosage , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Count , Cell Division/immunology , Cells, Cultured , Cytotoxicity, Immunologic , Dendritic Cells/pathology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Humans , Immunity, Cellular , Immunity, Innate , Immunosuppressive Agents/administration & dosage , Injections, Intraperitoneal , Ligands , Listeria monocytogenes/growth & development , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/microbiology , Listeriosis/pathology , Membrane Proteins/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Polyethylene Glycols/administration & dosage , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/pathology
8.
Cytokine ; 23(3): 53-63, 2003 Aug 07.
Article in English | MEDLINE | ID: mdl-12906868

ABSTRACT

CD40 ligand is a cell surface molecule on CD4(+) T cells that interacts with its receptor, CD40, on antigen presenting cells to mediate humoral and cellular immune responses. Our previous studies demonstrated that a trimeric soluble form of CD40L (CD40LT) activates macrophages to produce beta-chemokines and decrease CCR5 and CD4 cell surface expression, thus inducing resistance to HIV-1 infection. However, the mechanism(s) by which CD40LT mediates these effects in primary macrophages remains unclear. In this report, we demonstrate that CD40LT induces synthesis of beta-chemokines through the activation of MAPK signaling pathways. Treatment of macrophages with CD40LT results in a rapid activation of p38 and ERK1/2 mitogen-activated protein kinases. Inhibitors of these MAPKs blocked beta-chemokine production, while protein kinase A and C inhibitors had little or no effect. We also provide evidence that CD40LT stimulates beta-chemokine production directly, as well as indirectly via a TNF-alpha-dependent mechanism. At the early time points, CD40LT directly stimulated beta-chemokine production, whereas at later time points the effect was mediated to some extent by TNF-alpha. In conclusion, our results suggest that CD40-CD40L interactions are important for the activation of monocyte-derived macrophage antiviral response affecting both viral replication and the recruitment of immune cells.


Subject(s)
CD40 Ligand/metabolism , Chemokines, CC/metabolism , Macrophages/metabolism , Mitogen-Activated Protein Kinases/metabolism , Gene Expression Regulation , Humans , Signal Transduction/physiology , Transcription, Genetic , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases
9.
Med Sci Monit ; 8(6): BR208-11, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12070425

ABSTRACT

BACKGROUND: Patients who recover from acute trypanosomiasis may succumb to chronic Chagas' disease later in life due to age related immunosuppression, immune system disorders such as AIDS, or during periods of immunosuppressive therapy for organ transplantation. In this study, effects of immunomodulators with diverse properties were examined on the course of an acute and lethal Trypanosoma cruzi infection. MATERIAL/METHODS: ICR (Swiss) mice inoculated with Tulahuen (lethal) strain of T. cruzi were treated with 6 different immunomodulators and the course of infection was studied. RESULTS: Tacrolimus (FK-506) and dexamethasone increased parasitemia in mice when compared to infected untreated animals. Mycophenolate mofetil (RS-61443) and recombinant interleukin-15 (IL-15) treatment decreased the number of parasites but had no effect on animal survival. In contrast, compound L-685-818 (tacrolimus analog) and CD40 ligand (CD40L), provided protection against lethal infection. Mice that survived initial infection were all protected against reinfection. CONCLUSIONS: This study demonstrates the dangers of immunosuppression with tacrolimus and dexamethasone in T. cruzi infected subjects. While mycophenolate did not exacerbate the infection, our data suggest potential therapeutic applications for L-685-818 and CD40 ligand in trypanosomiasis.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Immunosuppressive Agents/therapeutic use , Tacrolimus/analogs & derivatives , Trypanosoma cruzi/isolation & purification , Trypanosomiasis/drug therapy , Animals , CD40 Ligand/therapeutic use , Dexamethasone/therapeutic use , Interleukin-15/therapeutic use , Mice , Mice, Inbred ICR , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/therapeutic use , Tacrolimus/therapeutic use , Trypanosomiasis/parasitology
10.
Infect Immun ; 71(3): 1209-16, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12595434

ABSTRACT

Bacterially induced bone infections often result in significant local inflammatory responses which are coupled with loss of bone. However, the mechanisms necessary for the protective host response, or those responsible for pathogen-induced bone loss, are not clear. Recent evidence demonstrates that bacterially infected osteoblasts secrete chemokines and cytokines, suggesting that these cells may have an unappreciated role in supporting localized inflammation. In this study, mouse and human osteoblasts were investigated for their ability to express functional CD40 upon exposure to two important pathogens of bone, Staphylococcus aureus and Salmonella enterica serovar Dublin. Bacterial infection of cultured mouse or human osteoblasts resulted in increased CD40 mRNA and CD40 protein expression induced by either pathogen. Importantly, CD40 expression by osteoblasts was functional, as assessed by ligation of this molecule with recombinant, soluble CD154. CD40 activity was assessed by induction of interleukin-6 and granulocyte-macrophage colony-stimulating factor in osteoblasts following ligation. Cocultures of activated CD4(+) T lymphocytes and osteoblasts could interact via CD40 and CD154, since an antibody against CD40 could block macrophage inflammatory protein-1alpha secretion. Taken together, these studies conclusively demonstrate that infected osteoblasts can upregulate expression of functional CD40 molecules which mediate cytokine secretion. This surprising result further supports the notion that bone-forming osteoblasts can directly interact with CD154-expressing cells (i.e., T lymphocytes) and can contribute to the host response during bone infection.


Subject(s)
CD40 Antigens/analysis , Osteoblasts/metabolism , Osteoblasts/microbiology , Salmonella enterica/pathogenicity , Staphylococcus aureus/pathogenicity , Animals , CD4-Positive T-Lymphocytes/metabolism , CD40 Antigens/genetics , CD40 Antigens/physiology , CD40 Ligand/physiology , Chemokine CCL4 , Cytokines/biosynthesis , Flow Cytometry , Gene Expression Regulation , Humans , Immunohistochemistry , Macrophage Inflammatory Proteins/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Osteoblasts/chemistry , RNA, Messenger
11.
J Immunol ; 170(4): 1797-805, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12574344

ABSTRACT

Members of the TNF superfamily have been shown to be instrumental in enhancing cell-mediated immune responses, primarily through their interactions with dendritic cells (DCs). We systematically evaluated the ability of three TNF superfamily molecules, CD40 ligand (CD40L), receptor activator of NF-kappaB ligand (RANKL), and TNF-alpha, to expand ex vivo EBV-specific CTL responses in healthy human individuals and ex vivo HIV-1-specific CTL responses in HIV-1-infected individuals. In both groups of individuals, we found that all three TNF family molecules could expand CTL responses, albeit at differing degrees. CD40L treatment alone was better than RANKL or TNF-alpha alone to mature DCs and to expand CTL. In healthy volunteers, TNF-alpha or RANKL could cooperate with CD40L to maximize the ability of DCs to expand virus-specific CTL responses. In HIV-1 infection, cooperative effects between TNF-alpha or RANKL in combination with CD40L were variable. TNF-alpha and RANKL cooperated with CD40L via differing mechanisms, i.e., TNF-alpha enhanced IL-12 production, whereas RANKL enhanced survival of CD40L-stimulated DCs. These findings demonstrate that optimal maturation of DCs requires multiple signals by TNF superfamily members that include CD40L. In HIV-1 infection, DCs may only require CD40L to maximally expand CTL. Finally, CTL responses were higher in CD4(+) T cell-containing conditions even in the presence of TNF family molecules, suggesting that CD4(+) T cells can provide help to CD8(+) T cells independently of CD40L, RANKL, or TNF-alpha.


Subject(s)
CD40 Ligand/physiology , CD8-Positive T-Lymphocytes/immunology , Carrier Proteins/physiology , Dendritic Cells/immunology , Glycoproteins/metabolism , HIV Infections/immunology , Immunologic Memory , Membrane Glycoproteins/physiology , NF-kappa B/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Necrosis Factor-alpha/physiology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/immunology , Cell Division/immunology , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/pharmacology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Drug Combinations , Epitopes, T-Lymphocyte/immunology , HIV Infections/virology , HIV-1/immunology , Herpesvirus 4, Human/immunology , Humans , Immunophenotyping , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Interleukin-15/biosynthesis , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Lymphocyte Depletion , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Osteoprotegerin , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptors, Tumor Necrosis Factor , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology
12.
J Immunol ; 171(11): 5842-52, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14634094

ABSTRACT

While studying Ag-pulsed syngeneic dendritic cell (DC) immunization, we discovered that surprisingly, unpulsed DCs induced protection against tumor lung metastases resulting from i.v. injection of a syngeneic BALB/c colon carcinoma CT26 or a syngeneic C57BL/6 lung carcinoma LL/2. Splenocytes or immature splenic DCs did not protect. The protection was mediated by NK cells, in that it was abrogated by treatment with anti-asialo-GM1 but not anti-CD8, and was induced by CD1(-/-) DCs unable to stimulate NKT cells, but did not occur in beige mice lacking NK cells. Protection correlated with increased NK activity, and increased infiltration of NK but not CD8(+) cells in lungs of tumor-bearing mice. Protection depended on the presence of costimulatory molecules CD80, CD86, and CD40 on the DCs, but surprisingly did not require DCs that could make IL-12 or IL-15. Unexpectedly, protection sensitive to anti-asialo-GM1 and increased NK activity were still present 14 mo after DC injection. As NK cells lack memory, we found by depletion that CD4(+) not CD8(+) T cells were required for induction of the NK antitumor response. The role of DCs and CD4(+) T cells provides a novel mechanism for NK cell induction and innate immunity against cancer that may have potential in preventing clinical metastases.


Subject(s)
Colonic Neoplasms/immunology , Colonic Neoplasms/therapy , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Dendritic Cells/transplantation , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, CD/physiology , B7-1 Antigen/biosynthesis , B7-1 Antigen/genetics , B7-1 Antigen/physiology , B7-2 Antigen , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Cell Line, Tumor , Colonic Neoplasms/genetics , Cytotoxicity, Immunologic/genetics , Female , Immunity, Innate , Immunotherapy, Adoptive/methods , Injections, Intravenous , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Mice, SCID , Neoplasm Transplantation
13.
Blood ; 99(9): 3360-6, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11964304

ABSTRACT

Human herpesvirus 8 (HHV-8; Kaposi sarcoma-associated herpesvirus)-specific cytotoxic T-lymphocyte (CTL) and interferon-gamma (IFN-gamma) responses to proteins produced during the lytic cycle of HHV-8 replication are mediated by HLA class I-restricted, CD8(+) T cells. We have characterized the fine specificity of the CD8(+) T-cell response to 25 peptides derived from 5 HHV-8 lytic cycle proteins based on a prediction model for HLA A*0201 binding motifs. One of the 25 HLA A*0201 peptides derived from the glycoprotein B (gB) homolog of Epstein-Barr virus (gB(492-500); LMWYELSKI; single-letter amino acid codes) bound to HLA A*0201 and stimulated IFN-gamma responses in CD8(+) T cells from HHV-8(+), HLA A*0201 persons, but not HHV-8-seronegative or non-HLA A*0201 persons. The peptide also induced IFN-gamma and CTL reactivity to naturally processed gB protein. The peptide was a major immunogenic epitope of HHV-8 as indicated by induction of IFN-gamma responses in peripheral blood mononuclear cells from 5 of 5 HHV-8 seropositive, HLA A*0201 persons when gB(492-500) was presented by autologous dendritic cells. T-cell reactivity to gB(492-500) was not related to detectable HHV-8 DNA in the blood. These data show that CD8(+) T cells recognize an HLA A*0201-restricted epitope for HHV-8 lytic cycle protein gB, particularly when presented by dendritic cells. This epitope may be important in control of HHV-8 infection by CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A Antigens/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Antigen Presentation/immunology , Case-Control Studies , Dendritic Cells/immunology , HLA-A2 Antigen , Herpesvirus 8, Human/chemistry , Herpesvirus 8, Human/immunology , Humans , Immunodominant Epitopes , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Peptide Fragments/immunology , Viral Envelope Proteins/chemistry
14.
J Infect Dis ; 187(2): 315-9, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12552458

ABSTRACT

Proteins may serve as ideal CD8(+) T cell immunogens for human immunodeficiency virus type 1 (HIV-1) if they can be delivered to and processed through the human leukocyte antigen class I pathway. This study shows that human blood monocyte-derived dendritic cells loaded with liposome-complexed HIV-1 proteins and matured with CD40 ligand can prime CD8(+) T cells to HIV-1 in vitro. Whole HIV-1 protein in liposome may be an effective immunogen for HIV-1 vaccine protocols.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , HIV Antigens/immunology , HIV-1/immunology , Lymphocyte Activation , AIDS Vaccines/administration & dosage , AIDS Vaccines/immunology , CD40 Ligand , Cell Membrane , Cells, Cultured , HIV Antigens/administration & dosage , Humans , Liposomes , Phosphatidylethanolamines
15.
Br J Haematol ; 120(2): 243-50, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12542481

ABSTRACT

Idiotypic structures of immunoglobulins from malignant B cells constitute tumour-specific antigens, though the function of immunoglobulin-specific CD8+ T cells in disease control and rejection remains unclear. We have studied five cases of B chronic lymphocytic leukaemia patients affected with indolent (three patients) or aggressive (two patients) disease. We showed that CD8+ T cells with major histocompatibility complex class I-restricted cytotoxicity against autologous tumour B cells could be generated following repeated stimulations with idiotype-pulsed dendritic cells in vitro. CD8+ T-cell lines were able to upregulate CD69 expression and to release interferon (IFN)-gamma upon contact with the autologous B cells, though cytolytic activity was only substantiated for patients with indolent disease. The failure of cytolytic activity in patients with aggressive disease may be explained by a skewed maturation of memory CD8 cells.


Subject(s)
CD8 Antigens/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , T-Lymphocytes, Cytotoxic/immunology , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , B-Lymphocytes/immunology , Histocompatibility Antigens Class I , Humans , Immunoglobulin M/administration & dosage , Immunoglobulin M/immunology , Immunologic Memory , Interferon-gamma/immunology , Lectins, C-Type , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphocyte Activation
16.
J Virol ; 76(6): 3007-14, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11861866

ABSTRACT

T-cell responses to X4 strains of human immunodeficiency virus type 1 (HIV-1) are considered important in controlling progression of HIV-1 infection. We investigated the ability of dendritic cells (DC) and various forms of HIV-1 X4 antigen to induce anti-HIV-1 T-cell responses in autologous peripheral blood mononuclear cells from HIV-1-infected persons. Immature DC loaded with HIV-1 IIIB-infected, autologous, apoptotic CD8(-) cells and matured with CD40 ligand induced gamma interferon production in autologous CD8(+) and CD4(+) T cells. In contrast, mature DC loaded with HIV-1 IIIB-infected, necrotic cells or directly infected with cell-free HIV-1 IIIB were poorly immunogenic. Thus, HIV-1-infected cells undergoing apoptosis serve as a rich source of X4 antigen for CD8(+) and CD4(+) T cells by DC. This may be an important mechanism of HIV-1 immunogenicity and provides a strategy for immunotherapy of HIV-1-infected patients on combination antiretroviral therapy.


Subject(s)
Apoptosis , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , HIV-1/immunology , Leukocytes, Mononuclear/virology , HIV Antigens/immunology , HIV-1/classification , Humans , Leukocytes, Mononuclear/physiology
17.
Proc Natl Acad Sci U S A ; 99(20): 13020-5, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12232042

ABSTRACT

Although a role for CD4(+) helper cells in CD8(+) cytotoxic T lymphocyte (CTL) induction by vaccines is widely recognized, much less is known about a counterbalancing role of CD4(+) T cells in down-modulating this response, or about ways to optimize vaccine responses through abrogation of this negative regulatory mechanism. Here, we discovered a synergistic enhancement of vaccine-mediated CTL induction and protection by the relief of suppression through depletion of regulatory CD4(+) cells, including CD4(+) NKT cells, or blockade of IL-13 made by these cells, combined with the cytokine granulocyte/macrophage colony-stimulating factor and the costimulatory molecule CD40L. Indeed, in the absence of helper epitopes, granulocyte/macrophage colony-stimulating factor and the helper-mimetic molecule CD40L are not sufficient to replace help to induce CTL without abrogation of CD4(+) T cell-mediated suppression, suggesting a role for T cell help in overcoming suppression. The increased CTL induction translated to striking protection against viral infection by a vaccine by using this synergistic combined approach. These results argue for a push-pull approach to maximize vaccine efficacy, especially for HIV and cancer.


Subject(s)
CD40 Ligand/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-13/antagonists & inhibitors , Analysis of Variance , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Dimerization , Epitopes , Female , Interleukin-13/metabolism , Mice , Mice, Inbred BALB C , T-Lymphocytes, Cytotoxic/metabolism , Vaccines, Synthetic/immunology , Vaccinia virus/genetics
18.
Vaccine ; 20 Suppl 4: A8-A22, 2002 Dec 19.
Article in English | MEDLINE | ID: mdl-12477423

ABSTRACT

Dendritic cells (DCs) can induce tumor- or pathogen-specific T cell responses in humans. We comprehensively compared the clinically available DC maturation stimuli for their ability to promote uniformly mature DCs that elicit higher levels of T cell responses. We compared the standard maturation stimulus, autologous monocyte-conditioned medium (MCM), with a synthetic double stranded RNA (poly I:C), soluble CD40 ligand trimer, and a defined cocktail of cytokines (TNF-alpha, IL-1 beta, IL-6) and PGE(2) to promote mature phenotype and function in human monocyte-derived DCs. The cocktail was the most efficient despite the lack of induction of IL-12p70. While these results support the use of the MCM-mimic cocktail in clinical DC immunotherapy trials, the roles of it's individual constituents remain to be completely defined.


Subject(s)
Cytokines/pharmacology , Dendritic Cells/cytology , Dinoprostone/pharmacology , Immunotherapy , Monocytes/cytology , Cell Division/immunology , Culture Media, Conditioned , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Interferon-gamma/metabolism , Interleukin-12/metabolism , Lymphocyte Culture Test, Mixed
19.
J Immunol ; 170(6): 3180-6, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12626576

ABSTRACT

We investigated the effect of recombinant CD40 ligand trimer (CD40LT) on the functional capacity of peripheral blood CD8(+) T cells from healthy tuberculin reactors that were cultured with Mycobacterium tuberculosis-infected autologous monocytes. CD40LT enhanced the capacity of M. tuberculosis-responsive CD8(+) T cells to produce IFN-gamma by increasing the number of IFN-gamma-producing CD8(+) T cells and the amount of IFN-gamma produced per cell. CD40LT-induced IFN-gamma production was dependent on production of IL-12 and IL-18, but did not require IL-15. CD40LT up-regulated expression of the transcription factors phosphorylated CREB and c-Jun, both of which have been previously shown to stimulate IFN-gamma mRNA transcription by binding to the IFN-gamma promoter. CD40LT also enhanced the capacity of CD8(+) T cells to lyse M. tuberculosis-infected monocytes, and increased CTL activity was associated with higher expression of perforin and granulysin, but not of Fas ligand. We conclude that CD40LT can enhance CD8(+) T cell effector function in response to M. tuberculosis.


Subject(s)
Adjuvants, Immunologic/physiology , CD40 Ligand/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Mycobacterium tuberculosis/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytotoxicity, Immunologic , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Monocytes/immunology , Monocytes/microbiology , Monokines/physiology , Phosphorylation , Promoter Regions, Genetic/immunology , Recombinant Proteins/pharmacology , Transcription Factors/biosynthesis , Transcription Factors/metabolism , Transcription Factors/physiology , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL