Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 487
Filter
1.
IUBMB Life ; 69(12): 971-977, 2017 12.
Article in English | MEDLINE | ID: mdl-29083508

ABSTRACT

The innate immunity protein Tag7 (PGRP-S, PGLYRP1) is involved in antimicrobial and antitumor defense. As shown in our previous studies, Tag7 specifically interacts with the major heat shock protein Hsp70 to form a stable Tag7-Hsp70 complex with cytotoxic activity against tumor cells. A stable complex of Tag7 with the calcium-binding protein Mts1 (S100A4) stimulates migration of lymphocytes. Moreover, Tag7 can activate cytotoxic lymphocytes that recognize and kill HLA-negative tumor cells. Here, we have shown that Tag 7 treatment of human peripheral blood mononuclear cells (PBMCs) results in activation of different cytotoxic lymphocyte populations-natural killer (NK) cells and CD8+ NKG2D+ T lymphocytes-that kill Moloney murine leukemia virus (MMLV) infected SC-1 cells using different mechanisms of cell death induction. This mechanism in NK cells is based on the release of granzymes, which activate apoptosis in target cells, while CD8+ NKG2D+ T lymphocytes recognize the noncanonical MicA antigen on the surface of virus-containing cells and kill them via the FasL-Fas interaction, triggering the apoptotic or necroptotic cell death pathway. Preliminary incubation of PBMCs with virus-infected cells and following incubation with Tag7 results in activation of lymphocytes with a different phenotype. These lymphocytes change the spectrum of target cells and the mechanism of cell death induction, and their interaction with target cells is not species-specific. © 2017 IUBMB Life, 69(12):971-977, 2017.


Subject(s)
Cytokines/immunology , Cytotoxicity, Immunologic/drug effects , Fas Ligand Protein/immunology , Killer Cells, Natural/immunology , T-Lymphocytes, Cytotoxic/immunology , fas Receptor/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Cell Line , Coculture Techniques , Cytokines/genetics , Cytokines/pharmacology , Fas Ligand Protein/genetics , Fibroblasts/immunology , Fibroblasts/virology , Gene Expression , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immunity, Innate , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/drug effects , Mice , Moloney murine leukemia virus/growth & development , Moloney murine leukemia virus/immunology , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Necrosis/genetics , Necrosis/immunology , Primary Cell Culture , Protein Binding , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , fas Receptor/genetics
2.
Proc Natl Acad Sci U S A ; 114(9): E1659-E1667, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28193872

ABSTRACT

T-cell memory is critical for long-term immunity. However, the factors involved in maintaining the persistence, function, and phenotype of the memory pool are undefined. Eomesodermin (Eomes) is required for the establishment of the memory pool. Here, we show that in T cells transitioning to memory, the expression of high levels of Eomes is not constitutive but rather requires a continuum of cell-intrinsic NFκB signaling. Failure to maintain NFκB signals after the peak of the response led to impaired Eomes expression and a defect in the maintenance of CD8 T-cell memory. Strikingly, we found that antigen receptor [T-cell receptor (TCR)] signaling regulates this process through expression of the NFκB-dependent kinase proviral integration site for Moloney murine leukemia virus-1 (PIM-1), which in turn regulates NFκB and Eomes. T cells defective in TCR-dependent NFκB signaling were impaired in late expression of Pim-1, Eomes, and CD8 memory. These defects were rescued when TCR-dependent NFκB signaling was restored. We also found that NFκB-Pim-1 signals were required at memory to maintain memory CD8 T-cell longevity, effector function, and Eomes expression. Hence, an NFκB-Pim-1-Eomes axis regulates Eomes levels to maintain memory fitness.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Moloney murine leukemia virus/immunology , NF-kappa B/immunology , T-Box Domain Proteins/immunology , Animals , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology
3.
PLoS One ; 10(5): e0126034, 2015.
Article in English | MEDLINE | ID: mdl-25946103

ABSTRACT

A variety of adjuvants fostering humoral immunity are known as of today. However, there is a lack of adjuvants or adjuvant strategies, which directly target T cellular effector functions and memory. We here determined whether systemically toxic cytokines such as IL-2 can be restricted to the site of antigen presentation and used as 'natural adjuvants'. Therefore, we devised antigen-presenting virus-like nanoparticles (VNP) co-expressing IL-2 attached to different membrane-anchors and assessed their potency to modulate CD8+ T cell responses in vitro and in vivo. Efficient targeting of IL-2 to lipid rafts and ultimately VNP was achieved by fusing IL-2 at its C-terminus to a minimal glycosylphosphatidylinositol (GPI)-anchor acceptor sequence. To identify optimal membrane-anchor dimensions we inserted one (1Ig), two (2Ig) or four (4Ig) immunoglobulin(Ig)-like domains of CD16b between IL-2 and the minimal GPI-anchor acceptor sequence of CD16b (GPI). We found that the 2IgGPI version was superior to all other evaluated IL-2 variants (IL-2v) in terms of its i) degree of targeting to lipid rafts and to the VNP surface, ii) biological activity, iii) co-stimulation of cognate T cells in the absence of bystander activation and iv) potency to induce differentiation and acquisition of CD8+ T cell effector functions in vitro and in vivo. In contrast, the GPI version rather favored memory precursor cell formation. These results exemplify novel beneficial features of membrane-bound IL-2, which in addition to its mere T cell stimulatory capacity include the induction of differential effector and memory functions in CD8+ T lymphocytes.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interleukin-2/immunology , Vaccines, Virus-Like Particle/immunology , Animals , Antigen Presentation , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Cell Line , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , HEK293 Cells , Humans , Immunologic Memory , In Vitro Techniques , Interleukin-2/genetics , Lymphocyte Activation , Membrane Microdomains/immunology , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Transgenic , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/immunology , Nanoparticles , Protein Structure, Tertiary , Receptors, IgG/chemistry , Receptors, IgG/genetics , Receptors, IgG/immunology , Vaccines, Virus-Like Particle/genetics
4.
J Virol ; 88(17): 10066-77, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24965455

ABSTRACT

UNLABELLED: We developed a Moloney mouse leukemia virus (MLV)-based retroviral replicating vector (RRV), Toca 511, which has displayed tumor specificity in resected brain tumor material and blood in clinical trials. Here, we investigated the interaction between Toca 511 and human host cells, and we show that RRVs do not induce type I interferon (IFN) responses in cultured human tumor cells or cultured human primary cells. However, exogenous type I IFN inhibited RRV replication in tumor cells and induced IFN-regulated genes, albeit at a lower level than in primary cells. Unexpectedly, RRVs did not induce IFN-α production upon incubation in vitro with human plasmacytoid dendritic cells (pDCs), whereas lentivirus vector and heat-treated RRVs did. Coincubation of RRVs with heat-treated RRVs or with lentivirus vector suppressed IFN-α production in pDCs, suggesting that native RRV has a dominant inhibitory effect on type I IFN induction. This effect is sensitive to trypsin treatment. In addition, heat treatment inactivated that activity but exposed an immune-stimulatory activity. The immune-stimulating component is sensitive to deglycosidases, trypsin, and phospholipase C treatment. Experiments with retroviral nonreplicating vectors and virus-like particles demonstrated that the immunosuppressive activity is not associated with the amphotropic envelope or the glyco-Gag protein. In summary, our data provide evidence that RRVs do not directly trigger type I IFN responses in IFN-responsive tumor cells. Moreover, RRVs appear to carry a heat-labile component that actively suppresses activation of cellular innate immune responses in pDCs. Inhibition of IFN induction by RRVs and the reduced response to IFN should facilitate tumor-specific infection in vivo. IMPORTANCE: RRVs have a convincing preference for replicating in tumor cells in animal models, and we observed similar preferences in the initial treatment of human glioblastoma patients. This study investigates the basis for the interaction between RRV and human host cells (tumor versus nontumor) in vitro. We found that RRVs do not trigger an IFN-α/ß response in tumor cells, but the cells are capable of responding to type I IFNs and of producing them when stimulated with known agonists. Surprisingly, the data show that RRVs can actively inhibit induction of cellular innate immunity and that this inhibitory activity is heat labile and trypsin sensitive and not attributable to the envelope protein. These data partially explain the observed in vivo tumor specificity.


Subject(s)
Interferon Type I/metabolism , Moloney murine leukemia virus/immunology , Moloney murine leukemia virus/physiology , Neoplasms/immunology , Virus Replication , Cells, Cultured , Humans , Moloney murine leukemia virus/genetics
5.
J Immunol ; 187(9): 4695-704, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21964024

ABSTRACT

Dysregulation of the T cell-dependent Ab response can lead to numerous immunological disorders, ranging from systemic lupus erythematosus to B cell lymphomas. Cellular processes governed by MHC class II proteins play a major role in this response and its dysregulation. The extent to which processes controlled by the diverse family of MHC class I proteins impact such autoimmune and neoplastic disorders, however, is less clear. In this study, we genetically dissect the contributions of individual MHC class I family members and the pathological processes under their control in the systemic lupus erythematosus-like disease of BXSB.Yaa mice and B cell lymphomagenesis of SJL mice. This study reveals a powerful repressive regulatory axis comprised of MHC class I-dependent CD8(+) T cells and NK cells. These results indicate that the predominant role of the MHC class I protein family in such immunological disorders is to protect from more aggressive diseases.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Histocompatibility Antigens Class I/physiology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/prevention & control , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/prevention & control , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , H-2 Antigens/genetics , Histocompatibility Antigen H-2D , Histocompatibility Antigens Class I/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Lupus Erythematosus, Systemic/mortality , Lymphoma, B-Cell/mortality , Mice , Mice, Inbred Strains , Moloney murine leukemia virus/immunology , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics
7.
J Immunol ; 187(9): 4431-9, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21940675

ABSTRACT

Variant peptide vaccines are used clinically to expand T cells that cross-react with tumor-associated Ags (TAA). To investigate the effects of elevated endogenous TAA expression on variant peptide-induced responses, we used the GP70 TAA model. Although young BALB/c mice display T cell tolerance to the TAA GP70(423-431) (AH1), expression of GP70 and suppression of AH1-specific responses increases with age. We hypothesized that as TAA expression increases, the AH1 cross-reactivity of variant peptide-elicited T cell responses diminishes. Controlling for immunosenescence, we showed that elevated GP70 expression suppressed AH1 cross-reactive responses elicited by two AH1 peptide variants. A variant that elicited almost exclusively AH1 cross-reactive T cells in young mice elicited few or no T cells in aging mice with Ab-detectable GP70 expression. In contrast, a variant that elicited a less AH1 cross-reactive T cell response in young mice successfully expanded AH1 cross-reactive T cells in all aging mice tested. However, these T cells bound the AH1/MHC complex with a relatively short half-life and responded poorly to ex vivo stimulation with the AH1 peptide. Variant peptide vaccine responses were also suppressed when AH1 peptide is administered tolerogenically to young mice before vaccination. Analyses of variant-specific precursor T cells from naive mice with Ab-detectable GP70 expression determined that these T cells expressed PD-1 and had downregulated IL-7Rα expression, suggesting they were anergic or undergoing deletion. Although variant peptide vaccines were less effective as TAA expression increases, data presented in this article also suggest that complementary immunotherapies may induce the expansion of T cells with functional TAA recognition.


Subject(s)
Antigenic Variation/immunology , Cancer Vaccines/immunology , Gene Expression Regulation, Neoplastic/immunology , Moloney murine leukemia virus/immunology , Up-Regulation/immunology , Vaccines, Subunit/immunology , Viral Envelope Proteins/biosynthesis , Aging/immunology , Animals , Cancer Vaccines/antagonists & inhibitors , Cells, Cultured , Down-Regulation/immunology , Epitopes, T-Lymphocyte/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Up-Regulation/genetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/antagonists & inhibitors , Viral Envelope Proteins/deficiency , Viral Envelope Proteins/genetics
8.
Immunol Lett ; 138(1): 79-85, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21453727

ABSTRACT

By exploring induction and persistence of virus-specific memory CD8(+) T cells in the BM of Moloney-murine sarcoma/leukemia virus-immune mice, we observed that the amount of activated CD8(+)CD62L(-) cells increased more rapidly and persisted for a longer period than in peripheral organs. Among the CD8(+)CD62L(-) subset, the few cells, specific for M-MuLV encoded antigens, expressing TCRVß5 rearrangements increased in an explosive manner doubling the percentage of TCRVß5(+) subset so that as a final result more than 10% of CD8(+) lymphocytes became potential virus-specific cytotoxic effectors. The numerical expansion of Vß5(+) cells started and persisted in the same proportion among both CD8(+)CD62L(-) and CD8(+)CD62L(+) subsets. In these subsets the analysis of CD44 phenotype, to distinguish effector (TEM) and central (TCM) memory, evidenced a twofold increase of Vß5(+) TEM percentage and fourfold increase of Vß5(+) TCM. In parallel, the non virus-specific Vß5(-) counterpart, also numerically increased due to the CD8(+) expansion, was partially reduced as TEM percentage and doubled as TCM percentage. We conclude that the immune response to M-MuLV encoded antigens in BM generate not only a large number of virus-specific memory cells but also the re-shaping of the entire memory T cell repertoire.


Subject(s)
Antigens, Viral/immunology , Bone Marrow/immunology , Moloney murine leukemia virus/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Epitopes/immunology , Immunization , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology
9.
Retrovirology ; 8: 20, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21414229

ABSTRACT

BACKGROUND: The involvement of xenotropic murine leukemia virus-related virus (XMRV) in prostate cancer (PC) and chronic fatigue syndrome (CFS) is disputed as its reported prevalence ranges from 0% to 25% in PC cases and from 0% to more than 80% in CFS cases. To evaluate the risk of XMRV infection during blood transfusion in Japan, we screened three populations--healthy donors (n = 500), patients with PC (n = 67), and patients with CFS (n = 100)--for antibodies against XMRV proteins in freshly collected blood samples. We also examined blood samples of viral antibody-positive patients with PC and all (both antibody-positive and antibody-negative) patients with CFS for XMRV DNA. RESULTS: Antibody screening by immunoblot analysis showed that a fraction of the cases (1.6-3.0%) possessed anti-Gag antibodies regardless of their gender or disease condition. Most of these antibodies were highly specific to XMRV Gag capsid protein, but none of the individuals in the three tested populations retained strong antibody responses to multiple XMRV proteins. In the viral antibody-positive PC patients, we occasionally detected XMRV genes in plasma and peripheral blood mononuclear cells but failed to isolate an infectious or full-length XMRV. Further, all CFS patients tested negative for XMRV DNA in peripheral blood mononuclear cells. CONCLUSION: Our data show no solid evidence of XMRV infection in any of the three populations tested, implying that there is no association between the onset of PC or CFS and XMRV infection in Japan. However, the lack of adequate human specimens as a positive control in Ab screening and the limited sample size do not allow us to draw a firm conclusion.


Subject(s)
Antibodies, Viral/blood , Fatigue Syndrome, Chronic/virology , Prostatic Neoplasms/virology , RNA, Viral/blood , Retroviridae Infections/complications , Xenotropic murine leukemia virus-related virus/isolation & purification , Animals , Blood Donors , Cell Line , Female , Humans , Immunoblotting , Japan , Male , Mice , Moloney murine leukemia virus/immunology , Retroviridae Infections/virology , Reverse Transcriptase Polymerase Chain Reaction , Transfusion Reaction , Xenotropic murine leukemia virus-related virus/genetics , Xenotropic murine leukemia virus-related virus/immunology
10.
J Immunol ; 184(6): 3072-8, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20154210

ABSTRACT

Chlamydophila pneumoniae infection of the vascular wall as well as activation of the transcription factor IFN regulatory factor (IRF)3 have been linked to development of chronic vascular lesions and atherosclerosis. The innate immune system detects invading pathogens by use of pattern recognition receptors, some of which are able to stimulate IRF3/7 activation and subsequent type I IFN production (e. g., IFN-beta). In this study, we show that infection of human endothelial cells with C. pneumoniae-induced production of IFN-beta, a cytokine that so far has been mainly associated with antiviral immunity. Moreover, C. pneumoniae infection led to IRF3 and IRF7 nuclear translocation in HUVECs and RNA interference experiments showed that IRF3 and IRF7 as well as the mitochondrial antiviral signaling (MAVS) were essential for IFN-beta induction. Finally, C. pneumoniae replication was enhanced in endothelial cells in which IRF3, IRF7, or MAVS expression was inhibited by small interfering RNA and attenuated by IFN-beta treatment. In conclusion, C. pneumoniae infection of endothelial cells activates an MAVS-, IRF3-, and IRF7-dependent signaling, which controls bacterial growth and might modulate development of vascular lesions.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Chlamydophila pneumoniae/growth & development , Chlamydophila pneumoniae/immunology , Endothelium, Vascular/immunology , Interferon Regulatory Factor-3/physiology , Interferon Regulatory Factor-7/physiology , Interferon-beta/physiology , Mitochondrial Proteins/physiology , RNA Interference/physiology , Cells, Cultured , Down-Regulation/immunology , Endothelium, Vascular/microbiology , Endothelium, Vascular/virology , Humans , Immunity, Innate , Interferon-beta/biosynthesis , Interferon-beta/genetics , Leukemia, Experimental/immunology , Leukemia, Experimental/microbiology , Leukemia, Experimental/virology , Moloney murine leukemia virus/immunology , RNA, Viral/antagonists & inhibitors , Retroviridae Infections/immunology , Retroviridae Infections/microbiology , Retroviridae Infections/virology , Signal Transduction/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/microbiology , Tumor Virus Infections/virology
11.
J Immunol ; 181(9): 6595-603, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941250

ABSTRACT

Pancreatic carcinoma is a very aggressive disease with dismal prognosis. Although evidences for tumor-specific T cell immunity exist, factors related to tumor microenvironment and the presence of immunosuppressive cytokines in patients' sera have been related to its aggressive behavior. Carcinoembryonic Ag (CEA) is overexpressed in 80-90% of pancreatic carcinomas and contains epitopes recognized by CD4(+) T cells. The aim of this study was to evaluate the extent of cancer-immune surveillance and immune suppression in pancreatic carcinoma patients by comparing the anti-CEA and antiviral CD4(+) T cell immunity. CD4(+) T cells from 23 normal donors and 44 patients undergoing surgical resection were tested for recognition of peptides corresponding to CEA and viral naturally processed promiscuous epitopes by proliferation and cytokine release assays. Anti-CEA CD4(+) T cell immunity was present in a significantly higher number of normal donors than pancreatic cancer patients. Importantly, whereas CD4(+) T cells from normal donors produced mainly GM-CSF and IFN-gamma, CD4(+) T cells from the patients produced mainly IL-5, demonstrating a skew toward a Th2 type. On the contrary, the extent of antiviral CD4(+) T cell immunity was comparable between the two groups and showed a Th1 type. The immunohistochemical analysis of tumor-infiltrating lymphocytes showed a significantly higher number of GATA-3(+) compared with T-bet(+) lymphoid cells, supporting a Th2 skew also at the tumor site. Collectively, these results demonstrate that Th2-immune deviation in pancreatic cancer is not generalized but tumor related and suggests that the skew might be possibly due to factor(s) present at the tumor site.


Subject(s)
Carcinoembryonic Antigen/immunology , Epitopes, T-Lymphocyte/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/virology , Th1 Cells/immunology , Th2 Cells/immunology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma/virology , Adult , Aged , Aged, 80 and over , Antibodies, Viral/biosynthesis , Antibodies, Viral/physiology , Carcinoembryonic Antigen/blood , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/virology , Epitopes, T-Lymphocyte/biosynthesis , Epitopes, T-Lymphocyte/metabolism , Epstein-Barr Virus Nuclear Antigens/immunology , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Lymphocyte Activation/immunology , Male , Middle Aged , Moloney murine leukemia virus/immunology , Pancreatic Neoplasms/pathology , Th1 Cells/pathology , Th1 Cells/virology , Th2 Cells/pathology , Th2 Cells/virology , Tumor Cells, Cultured , Viral Proteins/immunology
12.
J Leukoc Biol ; 84(2): 380-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18483203

ABSTRACT

The TNF family member, a proliferation-inducing ligand (APRIL), has been suggested to act as a costimulatory molecule in T cell responses. However, studies addressing this role in vivo are largely lacking. Here, we evaluated the effects of APRIL on physiological T cell responses in vivo. Although receptors for APRIL are expressed on a subset of T cells, neither TCR transgenic (Tg) T cell responses nor endogenous TCR responses were affected by Tg APRIL expression in vivo. Moreover, APRIL did not significantly enhance the induction of T cell lymphomas upon Moloney murine leukemia virus (MLV) infection. This clearly contrasts current belief and indicates that APRIL does not serve a major role in T cell immunity or lymphomagenesis. However, we did observe a strong increase in erythroleukemia formation after MLV inoculation of APRIL Tg mice. Strikingly, this erythroleukemia-facilitating property of APRIL was confirmed using the erythroleukemogenic Friend-MLV. Erythroleukemia in APRIL Tg mice was characterized by low hematocrits and grossly enlarged spleens with an increased percentage of erythroid precursors. Altogether, these results unveil new proerythroleukemogenic properties of APRIL.


Subject(s)
Leukemia, Erythroblastic, Acute/immunology , Leukemia, Erythroblastic, Acute/virology , Lymphoma, T-Cell/physiopathology , T-Lymphocytes/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/physiology , Animals , Autoimmunity , Flow Cytometry , Hematocrit , Heterozygote , Homozygote , Lymphoma, T-Cell/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Moloney murine leukemia virus/immunology , Moloney murine leukemia virus/pathogenicity , Ovalbumin/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Stem Cells/physiology , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
13.
Virology ; 366(2): 234-44, 2007 Sep 30.
Article in English | MEDLINE | ID: mdl-17543365

ABSTRACT

The retroviral restriction factors, TRIM5alpha and TRIMCyp, consist of RING and B-box 2 domains separated by a coiled coil from carboxy-terminal domains. These carboxy-terminal domains (the B30.2(SPRY) domain in TRIM5alpha and the cyclophilin A domain in TRIMCyp) recognize the retroviral capsid. Here we show that some B-box 2 changes in TRIM5alpha, but not in TRIMCyp, resulted in decreased human immunodeficiency virus (HIV-1) capsid binding. The phenotypic effects of these B-box 2 changes on the restriction of retroviral infection depended on the potency of restriction and the affinity of the TRIM5alpha interaction with the viral capsid, two properties specified by the B30.2(SPRY) domain. Thus, some alterations in the TRIM5alpha B-box 2 domain apparently affect the orientation or conformation of the B30.2(SPRY) domain, influencing capsid recognition.


Subject(s)
Capsid Proteins/metabolism , Carrier Proteins/metabolism , HIV-1/immunology , Moloney murine leukemia virus/immunology , Proteins/metabolism , Simian Immunodeficiency Virus/immunology , Animals , Antiviral Restriction Factors , Carrier Proteins/chemistry , Carrier Proteins/genetics , HeLa Cells , Humans , Protein Binding , Protein Structure, Tertiary , Proteins/chemistry , Proteins/genetics , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
14.
J Leukoc Biol ; 81(2): 528-38, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17062606

ABSTRACT

Hypoxia, a local decrease in oxygen tension, occurring in many pathological processes, modifies macrophage (Mphi) gene expression and function. Here, we provide the first evidence that hypoxia inhibits transgene expression driven by the Moloney murine leukemia virus-long terminal repeats (MoMLV-LTR) in IFN-gamma-activated Mphi. Hypoxia silenced the expression of several MoMLV-LTR-driven genes, including v-myc, enhanced green fluorescence protein, and env, and was effective in different mouse Mphi cell lines and on distinct MoMLV backbone-based viruses. Down-regulation of MoMLV mRNA occurred at the transcriptional level and was associated with decreased retrovirus production, as determined by titration experiments, suggesting that hypoxia may control MoMLV retroviral spread through the suppression of LTR activity. In contrast, genes driven by the CMV or the SV40 promoter were up-regulated or unchanged by hypoxia, indicating a selective inhibitory activity on the MoMLV promoter. It is interesting that hypoxia was ineffective in suppressing MoMLV-LTR-controlled gene expression in T or fibroblast cell lines, suggesting a Mphi lineage-selective action. Finally, we found that MoMLV-mediated gene expression in Mphi was also inhibited by picolinic acid, a tryptophan catabolite with hypoxia-like activity and Mphi-activating properties, suggesting a pathophysiological role of this molecule in viral resistance and its possible use as an antiviral agent.


Subject(s)
Cell Hypoxia/immunology , Gene Expression Regulation, Viral/genetics , Macrophages/immunology , Macrophages/virology , Moloney murine leukemia virus/genetics , Animals , Cell Line , Gene Expression Regulation, Viral/drug effects , Gene Expression Regulation, Viral/immunology , Genetic Vectors/genetics , Macrophages/drug effects , Mice , Mice, Inbred BALB C , Moloney murine leukemia virus/drug effects , Moloney murine leukemia virus/immunology , Picolinic Acids/pharmacology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Terminal Repeat Sequences/drug effects , Terminal Repeat Sequences/genetics , Terminal Repeat Sequences/immunology
16.
J Virol ; 80(5): 2539-47, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16474160

ABSTRACT

The membrane-proximal region of the human immunodeficiency virus type 1 (HIV-1) transmembrane protein (TM) is critical for envelope (Env)-mediated membrane fusion and contains the target for broadly reactive neutralizing antibody 2F5. It has been proposed that 2F5 neutralization might involve interaction of its long, hydrophobic, complementarity-determining region (CDR) H3, with adjacent viral membrane. Using Moloney murine leukemia virus (MLV) as a tool, we examined the effect of epitope position on 2F5 neutralization. When the 2F5 epitope was inserted in the proline-rich region of MLV Env surface protein (SU), 2F5 blocked cell fusion and virus infection, whereas MLV with a hemagglutinin (HA) epitope at the same position was not neutralized by anti-HA, even though the antibodies bound their respective Envs on the surface of infected cells and viruses equally well. When the 2F5 epitope was inserted in the MLV Env TM at a position comparable to its natural position in HIV-1 TM, 2F5 antibody blocked Env-mediated cell fusion. Epitope position had subtle effects on neutralization by 2F5: the antibody concentration for 50% inhibition of cell fusion was more than 10-fold lower when the 2F5 epitope was in SU than in TM, and inhibition was less complete at high concentrations of antibody; we discuss possible explanations for these effects of epitope position. Since membrane proximity was not required for neutralization by 2F5 antibody, we speculate that the CDR H3 of 2F5 contributes to neutralization by destabilizing an adjacent protein rather than by inserting into an adjacent membrane.


Subject(s)
Antibodies, Monoclonal/immunology , Epitopes/immunology , HIV Antibodies/immunology , HIV Antigens/immunology , HIV-1/immunology , Antigen-Antibody Reactions , Cell Fusion , Cell Line , Complementarity Determining Regions/immunology , HeLa Cells , Humans , Membrane Fusion , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/immunology , Neutralization Tests
17.
Proc Natl Acad Sci U S A ; 102(48): 17412-7, 2005 Nov 29.
Article in English | MEDLINE | ID: mdl-16293690

ABSTRACT

Current methodologies that monitor immune responses rely on invasive techniques that sample tissues at a given point in time. New technologies are needed to elucidate the temporal patterns of immune responses and the spatial distribution of immune cells on a whole-body scale. We describe a noninvasive, quantitative, and tomographic approach to visualize a primary anti-tumor immune response by using positron emission tomography (PET). Bone marrow chimeric mice were generated by engraftment of hematopoietic stem and progenitor cells transduced with a trifusion reporter gene encoding synthetic Renilla luciferase (hRluc), EGFP, and Herpes virus thymidine kinase (sr39TK). Mice were challenged with the Moloney murine sarcoma and leukemia virus complex (M-MSV/M-MuLV), and the induced immune response was monitored by using PET. Hematopoietic cells were visualized by using 9-[4-[(18)F]fluoro-3-(hydroxymethyl)butyl]guanine ([(18)F]FHBG), a radioactive substrate specific for the sr39TK PET reporter protein. Immune cell localization and expansion were seen at the tumor and draining lymph nodes (DLNs). 2-[(18)F]fluoro-2-deoxy-D-glucose ([(18)F]FDG), which is sequestered in metabolically active cells, was used to follow tumor growth and regression. Elevated glucose metabolism was also seen in activated lymphocytes in the DLNs by using the [(18)F]FDG probe. When M-MSV/M-MuLV-challenged mice were treated with the immunosuppressive drug dexamethasone, activation and expansion of immune cell populations in the DLNs could no longer be detected with PET imaging. The method we describe can be used to kinetically measure the induction and therapeutic modulations of cell-mediated immune responses.


Subject(s)
Moloney murine leukemia virus/immunology , Moloney murine sarcoma virus/immunology , Retroviridae Infections/immunology , Sarcoma, Experimental/immunology , Tumor Virus Infections/immunology , Animals , Bone Marrow Transplantation , Dexamethasone/therapeutic use , Flow Cytometry , Fluorodeoxyglucose F18 , Green Fluorescent Proteins , Guanine/analogs & derivatives , Hematopoietic Stem Cells/diagnostic imaging , Immunohistochemistry , Luciferases , Lymph Nodes/diagnostic imaging , Mice , Mice, Inbred C57BL , Mice, SCID , Positron-Emission Tomography/methods , Sarcoma, Experimental/diagnostic imaging , Sarcoma, Experimental/drug therapy , Thymidine Kinase
18.
Cell Immunol ; 235(1): 12-20, 2005 May.
Article in English | MEDLINE | ID: mdl-16098495

ABSTRACT

Our previous studies indicated that antigen-specific tolerance could be achieved by the injection of LPS-activated B-cell blasts that were retrovirally gene-transferred with an IgG-antigen fusion construct. This system was shown to be effective for tolerance induction with a variety of inserted antigens ranging in size from a single peptide to a large multi-epitope protein in a variety of mouse strains. Moreover, it was shown to be effective in four animal models for human disease. To optimize the existing protocol, establish the role of the IgG H chain scaffold, and provide baseline for potential clinical applications, we examined the effects of different B-cell activators, including lipopolysaccharide (LPS), anti-CD40, CpG oligodeoxynucleotide (CpG-ODN), and anti-IgM plus IL-4, on B-cell proliferation, GFP transduction efficiency, and tolerance induction in vivo. The results show that all activators except CpG-ODN have similar effects on retroviral gene transfer and peptide-IgG-induced tolerance. Furthermore, dose-response analyses showed that T-cell tolerance could be induced with 10(5) peptide-IgG LPS B-cell blasts, but that 10(6) transduced B-cells were needed for humoral unresponsiveness. Transduced anti-IgM-induced blasts were tolerogenic at 10(6) cells, but no dose of transduced CpG blasts was tolerogenic. Finally, to examine the role of IgG scaffold, a retroviral construct encoding lambda repressor p1-102 and signal peptide of murine IgG heavy chain was engineered to allow secretion of the p1-102 domain in the same manner as that of p1-102-IgG fusion protein. The results demonstrate that not only is IgG scaffold important in tolerance induction and maintenance of the long-lasting immune hyporesponsiveness, but assembly of the IgG heterodimer may be required for the efficacy of this system.


Subject(s)
Adoptive Transfer , B-Lymphocytes/transplantation , Genetic Therapy , Immune Tolerance/genetics , Immunoglobulin G/genetics , Immunoglobulin Heavy Chains/genetics , Animals , B-Lymphocytes/immunology , Genetic Vectors , Immunoglobulin G/physiology , Immunoglobulin Heavy Chains/physiology , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/immunology , Peptides/genetics , Peptides/immunology , Transduction, Genetic
19.
J Immunol ; 174(9): 5398-406, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15843538

ABSTRACT

The CD8(+) T cell response to Moloney-murine leukemia virus (M-MuLV)-induced Ags is almost entirely dominated by the exclusive expansion of lymphocytes that use preferential TCRVbeta chain rearrangements. In mice lacking T cells expressing these TCRVbeta, we demonstrate that alternative TCRVbeta can substitute for the lack of the dominant TCRVbeta in the H-2-restricted M-MuLV Ag recognition. We show that, at least for the H-2(b)-restricted response, the shift of TCR usage is not related to a variation of the immunodominant M-MuLV epitope recognition. After virus immunization, all the potentially M-MuLV-reactive lymphocytes are primed, but only the deletion of dominant Vbeta rescues the alternative Vbeta response. The mechanism of clonal T cell "immunodomination" that guides the preferential Vbeta expansion is likely the result of a proliferative advantage of T cells expressing dominant Vbeta, due to differences in TCR affinity and/or cosignal requirements. In this regard, a CD8 involvement is strictly required for the virus-specific cytotoxic activity of CTL expressing alternative, but not dominant, Vbeta gene rearrangements. The ability of T cells expressing alternative TCRVbeta rearrangements to mediate tumor protection was evaluated by a challenge with M-MuLV tumor cells. Although T cells expressing alternative Vbeta chains were activated and expanded, they were not able to control tumor growth in a long-lasting manner due to their incapacity of conversion and accumulation in the T central memory pool.


Subject(s)
Antigen Presentation/immunology , Gene Products, gag/immunology , Gene Products, gag/metabolism , Graft Rejection/immunology , Moloney murine leukemia virus/immunology , Sarcoma, Experimental/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Virus Infections/immunology , Animals , Antigen Presentation/genetics , Cell Line, Tumor , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Gene Products, gag/biosynthesis , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Graft Rejection/virology , H-2 Antigens/immunology , Histocompatibility Antigen H-2D , Immunity, Innate/genetics , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Sarcoma, Experimental/prevention & control , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/virology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/virology , Tumor Virus Infections/prevention & control
20.
J Virol ; 79(3): 1701-12, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15650195

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) Gag protein is a major target antigen for cytotoxic-T-lymphocyte-based vaccine strategies because of its high level of conservation. The murine model has been used extensively to evaluate potential HIV-1 vaccines. However, the biology of HIV-1 Gag is somewhat different in human and murine tissues. The ability of HIV-1 Gag to form virus-like particles (VLPs) in human cells is severely curtailed in murine cells. Hence, it is not known whether immunizing mice with expression vectors encoding HIV-1 Gag provides an accurate assessment of the immunogenicity of these candidate vaccines in primates. In this report, we made use of a chimeric Moloney murine leukemia virus (MMLV)-HIV-1 Gag in which the p17 matrix domain of HIV-1 was replaced with the p15 matrix and p12 domains from MMLV. Murine cells expressing this construct released significant amounts of VLPs. The construct preserved H-2d-restricted antigenic determinants in the remaining portion of HIV-1 Gag, allowing immunogenicity studies to be performed with mice. We demonstrated that immunizing mice with plasmid DNA or adenoviral vectors encoding this chimeric Gag did not significantly increase the HIV-1 Gag-specific cellular or humoral immune response when compared to immunization with a myristoylation-incompetent version of the construct. Thus, the release of VLPs formed in vivo may not play a major role in the immunogenicity of vectors expressing HIV-1 Gag constructs.


Subject(s)
AIDS Vaccines/immunology , Gene Products, gag/immunology , HIV-1/immunology , Moloney murine leukemia virus/immunology , Vaccines, DNA/immunology , Virion/immunology , AIDS Vaccines/administration & dosage , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Cell Line , Female , Gene Products, gag/genetics , Gene Products, gag/metabolism , Genetic Vectors , HIV Antibodies/blood , HIV-1/genetics , HIV-1/metabolism , Humans , Immunization , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Vaccines, DNA/administration & dosage , Virion/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...