Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.898
Filter
Add more filters

Publication year range
1.
Environ Toxicol ; 35(4): 457-467, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31793136

ABSTRACT

Cardamonin, a monomeric alkaloid, is isolated from Alpinia conchigera Griff and other natural plants. Recently, it has been focused on its anticancer activities, and no information showing its immune effects on leukemia mice was reported. In this study, we investigated the immune effects of cardamonin on WEHI-3 cell-generated leukemia mice. Forty BALB/c mice were randomly divided into four groups: Group I mice were normal animals and groups II-IV were leukemia. Group II mice, as a positive control, were administered with normal diet, and group III and IV mice were treated with 1 and 5 mg/kg of cardamonin, respectively, by intraperitoneal injection every 2 days for 14 days. The population of white blood cells, macrophage phagocytosis, and the proliferations of T and B cells were analyzed by flow cytometry. Another forty mice were also separated randomly into four groups for the determination of survival rate. Results showed that cardamonin did not affect body weight. Cardamonin decreased CD3, CD11b, and Mac-3 cell populations but increased CD19 number. Cardamonin enhanced phagocytic abilities of macrophages from the peripheral blood mononuclear cells of leukemia mice. Furthermore, cardamonin at 1 mg/kg treatment improved the survival rate of leukemia mice in vivo. Therefore, cardamonin could be applied for a leukemia therapeutic reagent at a defined dose.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Chalcones/pharmacology , Leukemia, Experimental/drug therapy , Leukemia, Experimental/immunology , Leukocytes, Mononuclear/drug effects , Animals , Antigens, CD19/blood , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Leukocytes, Mononuclear/immunology , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Phagocytosis/drug effects , Phagocytosis/immunology , Survival Rate
2.
Environ Toxicol ; 34(5): 659-665, 2019 May.
Article in English | MEDLINE | ID: mdl-30761740

ABSTRACT

Ouabain, a cardiotonic steroid, was used for the treatment of heart failure and atrial fibrillation and induces cancer cell apoptosis in many human cancer cells including human leukemia cells. However, there are no reports to show the effects on immune responses in a leukemia mouse model. In this study, WEHI-3 cell generated leukemia mice were developed and treated by oral ouabain at 0, 0.75, 1.5, and 3 mg/kg for 15 days. Results indicated that ouabain did not affect body appearance, but decreased liver and spleen weights, B- and T-cell proliferation at all three doses treatment and increased CD19 cells at 3.0 mg/kg treatment, decreased CD3, CD11b, and Mac-3 cells levels compared with positive control. Furthermore, ouabain increased the macrophage phagocytosis from peripheral blood mononuclear cell and peritoneal cavity at all three doses treatment and increased NK cell activities. Ouabain restored GOT, GPT and LDH levels in WEHI-3 leukemia mice in vivo.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Cytotoxicity, Immunologic/drug effects , Killer Cells, Natural/drug effects , Leukemia, Experimental/drug therapy , Lymphocyte Activation/drug effects , Ouabain/therapeutic use , Phagocytosis/drug effects , Animals , Cell Line, Tumor , Killer Cells, Natural/immunology , Leukemia, Experimental/immunology , Leukemia, Experimental/pathology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Phagocytosis/immunology
3.
Int J Cancer ; 142(5): 976-987, 2018 03 01.
Article in English | MEDLINE | ID: mdl-28975621

ABSTRACT

The antitumor effectiveness of cyclophosphamide (CTX) and other chemotherapeutics was shown to rely not only on direct cytotoxicity but also on immunogenic tumor cell death and systemic immunomodulatory mechanisms, including regulatory T cell (Treg) depletion, Th1 cell polarization, type I interferon (IFN) and proinflammatory cytokine production. IFN regulatory factor (IRF)-1 is a transcriptional regulator of IFNs and IFN-inducible genes, involved in the control of Th1 and Treg differentiation and in sterile inflammation. Aim of this study was to explore the role of IRF-1 in CTX-induced antitumor effects and related immune activities. This study shows for the first time that IRF-1 is important for the antitumor efficacy of CTX in mice. Moreover, experiments in tumor-bearing C57BL/6 mice showed that Irf1 gene expression in the spleen was transiently increased following CTX administration and correlated with the induction of Th1 cell expansion and of Il12p40 gene expression, which is the main Th1-driving cytokine. At the same time, CTX administration reduced both Foxp3 expression and Treg cell percentages. These effects were abrogated in Irf1-/- mice. Further experiments showed that the gene and/or protein expression of caspase-1, iNOS, IL-1ß, IL-6 and CXCL10 and the levels of nitric oxide were modulated following CTX in an IRF-1-direct- or -indirect-dependent manner, and highlighted the importance of caspase-1 in driving the sterile inflammatory response to CTX. Our data identify IRF-1 as important for the antitumor efficacy of CTX and for the regulation of many immunomodulatory activities of CTX, such as Th1 polarization, Treg depletion and inflammation.


Subject(s)
Cyclophosphamide/pharmacology , Inflammasomes/immunology , Interferon Regulatory Factor-1/physiology , Leukemia, Experimental/drug therapy , Retroviridae Infections/drug therapy , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Tumor Virus Infections/drug therapy , Animals , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Cytokines/metabolism , Female , Gene Expression Regulation/drug effects , Leukemia, Experimental/immunology , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Rauscher Virus/pathogenicity , Retroviridae Infections/immunology , Retroviridae Infections/metabolism , Retroviridae Infections/pathology , Tumor Cells, Cultured , Tumor Virus Infections/immunology , Tumor Virus Infections/metabolism , Tumor Virus Infections/pathology
4.
J Immunol ; 197(9): 3628-3638, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27647833

ABSTRACT

Elucidation of the immune requirements for control or elimination of retroviral infection remains an important aim. We studied the induction of adaptive immunity to neonatal infection with a murine retrovirus, under conditions leading to immunological tolerance. We found that the absence of either maternal or offspring adaptive immunity permitted efficient vertical transmission of the retrovirus. Maternal immunodeficiency allowed the retrovirus to induce central Th cell tolerance in the infected offspring. In turn, this compromised the offspring's ability to mount a protective Th cell-dependent B cell response. However, in contrast to T cells, offspring B cells were not centrally tolerized and retained their ability to respond to the infection when provided with T cell help. Thus, escape of retrovirus-specific B cells from deletional tolerance offers the opportunity to induce protective retroviral immunity by restoration of retrovirus-specific T cell help, suggesting similar T cell immunotherapies for persistent viral infections.


Subject(s)
Adoptive Transfer , B-Lymphocytes/immunology , Infectious Disease Transmission, Vertical/prevention & control , Leukemia Virus, Murine/immunology , Leukemia, Experimental/prevention & control , Retroviridae Infections/prevention & control , T-Lymphocytes/immunology , Tumor Virus Infections/prevention & control , Animals , Animals, Newborn , B-Lymphocytes/transplantation , B-Lymphocytes/virology , Cells, Cultured , Central Tolerance , Female , Leukemia, Experimental/immunology , Male , Maternal Exposure/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Retroviridae Infections/immunology , Retroviridae Infections/transmission , T-Lymphocytes/transplantation , T-Lymphocytes/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/transmission
5.
Retrovirology ; 14(1): 42, 2017 Aug 23.
Article in English | MEDLINE | ID: mdl-28835242

ABSTRACT

BACKGROUND: Myeloid-derived suppressor cells (MDSCs) can suppress T cell responses in several different diseases. Previously these suppressive cells were observed to expand in HIV patients and in a mouse retrovirus model, yet their suppressive effect on virus-specific CD8+ T cells in vitro and in vivo has not been characterized thus far. RESULTS: We used the Friend retrovirus (FV) model to demonstrate that MDSCs expand and become activated during the late phase of acute FV infection. Only the subpopulation of granulocytic MDSCs (gMDSCs) but not monocytic MDSC suppressed virus-specific CD8+ T cell proliferation and function in vitro. gMDSCs expressed arginase 1, high levels of the inhibitory ligand PD-L1 and the ATP dephosphorylating enzyme CD39 on the cell surface upon infection. All three molecules were involved in the suppressive effect of the gMDSCs in vitro. MDSC depletion experiments in FV-infected mice revealed that they restrict virus-specific CD8+ T cell responses and thus affect the immune control of chronic retroviruses in vivo. CONCLUSIONS: Our study demonstrates that MDSCs become activated and expand during the acute phase of retrovirus infection. Their suppressive activity on virus-specific CD8+ T cells may contribute to T cell dysfunction and the development of chronic infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Friend murine leukemia virus/immunology , Granulocytes/immunology , Myeloid-Derived Suppressor Cells/immunology , Retroviridae Infections/immunology , Animals , Antigens, Differentiation/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cell Proliferation , Granulocytes/metabolism , Granulocytes/pathology , Leukemia, Experimental/immunology , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Retroviridae Infections/metabolism , Retroviridae Infections/pathology , Tumor Virus Infections/immunology , Tumor Virus Infections/metabolism , Tumor Virus Infections/pathology
6.
J Virol ; 90(16): 7118-7130, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27226373

ABSTRACT

UNLABELLED: Injection of the LP-BM5 murine leukemia virus into mice causes murine AIDS, a disease characterized by many dysfunctions of immunocompetent cells. To establish whether the disease is characterized by glutathione imbalance, reduced glutathione (GSH) and cysteine were quantified in different organs. A marked redox imbalance, consisting of GSH and/or cysteine depletion, was found in the lymphoid organs, such as the spleen and lymph nodes. Moreover, a significant decrease in cysteine and GSH levels in the pancreas and brain, respectively, was measured at 5 weeks postinfection. The Th2 immune response was predominant at all times investigated, as revealed by the expression of Th1/Th2 cytokines. Furthermore, investigation of the activation status of peritoneal macrophages showed that the expression of genetic markers of alternative activation, namely, Fizz1, Ym1, and Arginase1, was induced. Conversely, expression of inducible nitric oxide synthase, a marker of classical activation of macrophages, was detected only when Th1 cytokines were expressed at high levels. In vitro studies revealed that during the very early phases of infection, GSH depletion and the downregulation of interleukin-12 (IL-12) p40 mRNA were correlated with the dose of LP-BM5 used to infect the macrophages. Treatment of LP-BM5-infected mice with N-(N-acetyl-l-cysteinyl)-S-acetylcysteamine (I-152), an N-acetyl-cysteine supplier, restored GSH/cysteine levels in the organs, reduced the expression of alternatively activated macrophage markers, and increased the level of gamma interferon production, while it decreased the levels of Th2 cytokines, such as IL-4 and IL-5. Our findings thus establish a link between GSH deficiency and Th1/Th2 disequilibrium in LP-BM5 infection and indicate that I-152 can be used to restore the GSH level and a balanced Th1/Th2 response in infected mice. IMPORTANCE: The first report of an association between Th2 polarization and alteration of the redox state in LP-BM5 infection is presented. Moreover, it provides evidence that LP-BM5 infection causes a decrease in the thiol content of peritoneal macrophages, which can influence IL-12 production. The restoration of GSH levels by GSH-replenishing molecules can represent a new therapeutic avenue to fight this retroviral infection, as it reestablishes the Th1/Th2 balance. Immunotherapy based on the use of pro-GSH molecules would permit LP-BM5 infection and probably all those viral infections characterized by GSH deficiency and a Th1/Th2 imbalance to be more effectively combated.


Subject(s)
Glutathione/deficiency , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/complications , Murine Acquired Immunodeficiency Syndrome/etiology , Retroviridae Infections/complications , Th2 Cells/immunology , Tumor Virus Infections/complications , Animals , Cells, Cultured , Cytokines/metabolism , Female , Leukemia, Experimental/immunology , Leukemia, Experimental/virology , Lymphocyte Activation , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/virology , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/metabolism , Murine Acquired Immunodeficiency Syndrome/pathology , Retroviridae Infections/immunology , Retroviridae Infections/virology , Spleen/immunology , Spleen/metabolism , Spleen/virology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/virology , Th2 Cells/metabolism , Th2 Cells/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/virology
7.
Blood ; 126(11): 1314-23, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26138686

ABSTRACT

MicroRNAs (miRs) play important roles in orchestrating many aspects of the immune response. The miR-17-92 cluster, which encodes 6 miRs including 17, 18a, 19a, 20a, 19b-1, and 92-1, is among the best characterized of these miRs. The miR-17-92 cluster has been shown to regulate a variety of immune responses including infection, tumor, and autoimmunity, but the role of this cluster in T-cell response to alloantigens has not been previously explored. By using major histocompatibility complex (MHC)-matched, -mismatched, and haploidentical murine models of allogeneic bone marrow transplantation (allo-BMT), we demonstrate that the expression of miR-17-92 on donor T cells is essential for the induction of graft-versus-host disease (GVHD), but dispensable for the graft-versus-leukemia (GVL) effect. The miR-17-92 plays a major role in promoting CD4 T-cell activation, proliferation, survival, and Th1 differentiation, while inhibiting Th2 and iTreg differentiation. Alternatively, miR-17-92 may promote migration of CD8 T cells to GVHD target organs, but has minimal impact on CD8 T-cell proliferation, survival, or cytolytic function, which could contribute to the preserved GVL effect mediated by T cells deficient for miR-17-92. Furthermore, we evaluated a translational approach and found that systemic administration of antagomir to block miR-17 or miR-19b in this cluster significantly inhibited alloreactive T-cell expansion and interferon-γ (IFNγ) production, and prolonged the survival in recipients afflicted with GVHD while preserving the GVL effect. Taken together, the current work provides a strong rationale and demonstrates the feasibility to target miR-17-92 for the control of GVHD while preserving GVL activity after allo-BMT.


Subject(s)
Graft vs Host Disease/immunology , Leukemia, Experimental/immunology , MicroRNAs/genetics , MicroRNAs/immunology , T-Lymphocytes/immunology , Allografts , Animals , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/genetics , Graft vs Leukemia Effect/genetics , Graft vs Leukemia Effect/immunology , Interferon-gamma/biosynthesis , Leukemia, Experimental/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , MicroRNAs/antagonists & inhibitors , Oligonucleotides/pharmacology
8.
Blood ; 125(10): 1578-88, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25564405

ABSTRACT

Chronic lymphocytic leukemia (CLL) is a common B-cell malignancy characterized by a highly variable course and outcome. The disease is believed to be driven by B-cell receptor (BCR) signals generated by external antigens and/or cell-autonomous BCR interactions, but direct in vivo evidence for this is still lacking. To further define the role of the BCR pathway in the development and progression of CLL, we evaluated the capacity of different types of antigen/BCR interactions to induce leukemia in the Eµ-TCL1 transgenic mouse model. We show that cell autonomous signaling capacity is a uniform characteristic of the leukemia-derived BCRs and represents a prerequisite for CLL development. Low-affinity BCR interactions with autoantigens generated during apoptosis are also positively selected, suggesting that they contribute to the pathogenesis of the disease. In contrast, high-affinity BCR interactions are not selected, regardless of antigen form or presentation. We also show that the capacity of the leukemic cells to respond to cognate antigen correlates inversely with time to leukemia development, suggesting that signals induced by external antigen increase the aggressiveness of the disease. Collectively, these findings provide in vivo evidence that the BCR pathway drives the development and can influence the clinical course of CLL.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/etiology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Amino Acid Sequence , Animals , Antigen Presentation , Autoantigens/genetics , Disease Progression , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Gene Rearrangement, B-Lymphocyte, Light Chain , Humans , Leukemia, Experimental/etiology , Leukemia, Experimental/genetics , Leukemia, Experimental/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Muramidase/genetics , Muramidase/immunology , Proto-Oncogene Proteins/genetics , Receptors, Antigen, B-Cell/genetics , Signal Transduction/immunology , snRNP Core Proteins/genetics , snRNP Core Proteins/immunology
9.
Retrovirology ; 13: 24, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27076190

ABSTRACT

BACKGROUND: Regulatory T cells (Tregs) have been shown to limit anti-viral immunity during chronic retroviral infection and to restrict vaccine-induced T cell responses. The objective of the study was to assess whether a combinational therapy of nanoparticle-based therapeutic vaccination and concomitant transient ablation of Tregs augments anti-viral immunity and improves virus control in chronically retrovirus-infected mice. Therefore, chronically Friend retrovirus (FV)-infected mice were immunized with calcium phosphate (CaP) nanoparticles functionalized with TLR9 ligand CpG and CD8(+) or CD4(+) T cell epitope peptides (GagL85-93 or Env gp70123-141) of FV. In addition, Tregs were ablated during the immunization process. Reactivation of CD4(+) and CD8(+) effector T cells was analysed and the viral loads were determined. RESULTS: Therapeutic vaccination of chronically FV-infected mice with functionalized CaP nanoparticles transiently reactivated cytotoxic CD8(+) T cells and significantly reduced the viral loads. Transient ablation of Tregs during nanoparticle-based therapeutic vaccination strongly enhanced anti-viral immunity and further decreased viral burden. CONCLUSION: Our data illustrate a crucial role for CD4(+) Foxp3(+) Tregs in the suppression of anti-viral T cell responses during therapeutic vaccination against chronic retroviral infection. Thus, the combination of transient Treg ablation and therapeutic nanoparticle-based vaccination confers robust and sustained anti-viral immunity.


Subject(s)
Leukemia, Experimental/therapy , Leukocyte Reduction Procedures , Nanoparticles/administration & dosage , Retroviridae Infections/therapy , T-Lymphocytes, Regulatory/immunology , Tumor Virus Infections/therapy , Viral Vaccines/administration & dosage , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Combined Modality Therapy/methods , Friend murine leukemia virus/immunology , Leukemia, Experimental/immunology , Mice, Inbred C57BL , Retroviridae Infections/immunology , Treatment Outcome , Tumor Virus Infections/immunology
10.
J Virol ; 89(2): 1468-73, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25378499

ABSTRACT

Toll-like receptor 7 and Myd88 are required for antiretroviral antibody and germinal center responses, but whether somatic hypermutation and class-switch recombination are required for antiretroviral immunity has not been examined. Mice deficient in activation-induced cytidine deaminase (AID) resisted Friend virus infection, produced virus-neutralizing antibodies, and controlled viremia. Passive transfer demonstrated that immune IgM from AID-deficient mice contributes to Friend virus control in the presence of virus-specific CD4+ T cells.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Friend murine leukemia virus/immunology , Leukemia, Experimental/immunology , Retroviridae Infections/immunology , Somatic Hypermutation, Immunoglobulin , Tumor Virus Infections/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cytidine Deaminase/deficiency , Immunization, Passive , Immunoglobulin Class Switching , Immunoglobulin M/immunology , Leukemia, Experimental/virology , Mice , Retroviridae Infections/virology , Tumor Virus Infections/virology
11.
J Immunol ; 193(6): 2952-60, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25098294

ABSTRACT

Vß5(+) regulatory T cells (Tregs), which are specific for a mouse endogenous retroviral superantigen, become activated and proliferate in response to Friend virus (FV) infection. We previously reported that FV-induced expansion of this Treg subset was dependent on CD8(+) T cells and TNF-α, but independent of IL-2. We now show that the inflammatory milieu associated with FV infection is not necessary for induction of Vß5(+) Treg expansion. Rather, it is the presence of activated CD8(+) T cells that is critical for their expansion. The data indicate that the mechanism involves signaling between the membrane-bound form of TNF-α on activated CD8(+) T cells and TNFR2 on Tregs. CD8(+) T cells expressing membrane-bound TNF-α but no soluble TNF-α remained competent to induce strong Vß5(+) Treg expansion in vivo. In addition, Vß5(+) Tregs expressing only TNFR2 but no TNFR1 were still responsive to expansion. Finally, treatment of naive mice with soluble TNF-α did not induce Vß5(+) Treg expansion, but treatment with a TNFR2-specific agonist did. These results reveal a new mechanism of intercellular communication between activated CD8(+) T cell effectors and Tregs that results in the activation and expansion of a Treg subset that subsequently suppresses CD8(+) T cell functions.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Carrier Proteins/genetics , Female , Friend murine leukemia virus/immunology , Leukemia, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II/agonists , Retroviridae Infections/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Virus Infections/immunology
12.
J Immunol ; 192(3): 1302-12, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24391208

ABSTRACT

Exposure to pathogen-associated molecular patterns (PAMPs) by vaccination or infection is known to have beneficial effects on neoplastic diseases, although the underlying molecular mechanisms are so far unclear. In this article, we report that Abs against (1→6)-ß-d-glucan, a typical microbial PAMP and a major target for high titer circulating natural Abs in healthy human subjects, cross-recognize a novel tumor-associated carbohydrate Ag on cancer cells. The (1→6)-ß-glucan cross-reactive moiety is immunologically dominant in tumor cells, as C57BL/6 mice harboring EL-4 solid tumors produced anti-(1→6)-ß-glucan Abs and the titer of which significantly correlated with enhanced survival and smaller tumor burden. Moreover, the (1→6)-ß-glucan-specific Abs exhibited potent tumoricidal activities in vitro. C57BL/6 mice immunized with Candida albicans produced protective immunity against inoculated EL-4 tumors, which was attributed to the formation of (1→6)-ß-glucan-specific Abs. Importantly, (1→6)-ß-glucan-specific Abs significantly prolonged the survival and reduced the tumor size in mice inoculated with EL-4 tumors. Our results demonstrate that the (1→6)-ß-glucan cross-reactive moiety represents a focal point between infection immunity and cancer surveillance, and natural Abs against this epitope may contribute to the first-line antitumor surveillance in humans. Our data also provide important explanation for the long-observed relationship between feverish infection and concurrent remission from cancer.


Subject(s)
Antibodies, Fungal/immunology , Antibodies, Neoplasm/immunology , Antigens, Fungal/immunology , Antigens, Neoplasm/immunology , Candida albicans/immunology , Immunodominant Epitopes/immunology , Immunologic Surveillance , Leukemia, Experimental/immunology , beta-Glucans/immunology , Adult , Animals , Antibody Specificity , Carbohydrate Conformation , Cells, Cultured , Cross Reactions , Dextrans/immunology , Female , Fungal Vaccines , Glycogen/immunology , Humans , Immunization , Male , Mice , Mice, Inbred C57BL , Middle Aged , Polysaccharides/immunology , Proteoglycans , beta-Glucans/isolation & purification
13.
Arch Toxicol ; 90(8): 1929-38, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26486797

ABSTRACT

PM2.5 (aerodynamic diameter ≤2.5 µm) has been a dominating and ubiquitous air pollutant and has become a global concern. Emerging evidences suggest a positive correlation between PM2.5 and leukemia, but the underlying molecular mechanisms remain unclear and need to be elucidated. Here, we assessed the impacts of PM2.5 on the progression and inflammation of human myeloid leukemia at lower environmental doses and explored the possible pathway. We showed that PM2.5 exposure significantly induced the leukemia cell growth and enhanced the release of inflammatory mediators in both in vitro and in vivo models. Additionally, NF-κB p65 and p-STAT3 were activated in PM2.5-treated leukemia cells, with a concomitant increase in both ROS formation and NADPH oxidase expressions. Strikingly, the supplement of inhibitors, including NAC (ROS), PDTC (NF-κB), or WP1066 (STAT3), contributed to a decline in leukemia cell growth. Furthermore, enhanced expressions of inflammatory cytokines were attenuated by the addition of NAC or PDTC, but not affected by WP1066. This study demonstrates that PM2.5 promotes leukemia progression, identifies a potential intervention target, and provides further understanding of the detrimental effect of PM2.5 exposure on human health.


Subject(s)
Air Pollutants/toxicity , Cell Proliferation/drug effects , Cytokines/metabolism , Leukemia, Experimental/immunology , Leukemia, Myeloid/immunology , Particulate Matter/toxicity , Animals , Cell Survival/drug effects , Cell Survival/immunology , Disease Progression , Dose-Response Relationship, Drug , HL-60 Cells , Humans , K562 Cells , Leukemia, Experimental/pathology , Leukemia, Myeloid/pathology , Mice, Nude , Neoplasm Transplantation , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Transcription Factor RelA/metabolism
14.
J Virol ; 88(23): 13892-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25231296

ABSTRACT

It is still unclear whether expanded and activated regulatory T cells (Tregs) in chronic viral infections can influence primary immune responses against superinfections with unrelated viruses. Expanded Tregs found in the spleens of chronically Friend virus (FV)-infected mice decreased murine cytomegalovirus (mCMV)-specific CD8(+) T cell responses during acute mCMV superinfection. This suppression of mCMV-specific T cell immunity was found only in organs with FV-induced Treg expansion. Surprisingly, acute mCMV infection itself did not expand or activate Tregs.


Subject(s)
Friend murine leukemia virus/immunology , Herpesviridae Infections/immunology , Immune Tolerance , Muromegalovirus/immunology , Retroviridae Infections/immunology , Superinfection/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Leukemia, Experimental/complications , Leukemia, Experimental/immunology , Male , Mice, Inbred C57BL , Retroviridae Infections/complications , Spleen/immunology , Tumor Virus Infections/complications , Tumor Virus Infections/immunology
15.
Blood ; 121(17): 3511-20, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23430112

ABSTRACT

Donor T cells play pivotal roles in graft-versus-host disease (GVHD) and graft-versus-leukemia (GVL) effects following bone marrow transplantation (BMT). DNAX accessory molecule 1 (DNAM-1) is a costimulatory and adhesion molecule, expressed mainly by natural killer cells and CD8(+) T cells at steady state to promote adhesion to ligand-expressing targets and enhance cytolysis. We have analyzed the role of this pathway in GVHD and GVL. The absence of DNAM-1 on the donor graft attenuated GVHD in major histocompatibility complex (MHC)-mismatched and MHC-matched BMT following conditioning with lethal and sublethal irradiation. In contrast, DNAM-1 was not critical for GVL effects against ligand (CD155) expressing and nonexpressing leukemia. The effects on GVHD following myeloablative conditioning were independent of CD8(+) T cells and dependent on CD4(+) T cells, and specifically donor FoxP3(+) regulatory T cells (Treg). The absence of DNAM-1 promoted the expansion and suppressive function of Treg after BMT. These findings provide support for therapeutic DNAM-1 inhibition to promote tolerance in relevant inflammatory-based diseases characterized by T-cell activation.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/physiology , Bone Marrow Transplantation , Graft vs Host Disease/prevention & control , Graft vs Leukemia Effect/immunology , Leukemia, Experimental/prevention & control , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Differentiation, T-Lymphocyte/chemistry , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Female , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Leukemia, Experimental/etiology , Leukemia, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Transplantation Conditioning , Tumor Cells, Cultured , Whole-Body Irradiation
16.
Environ Toxicol ; 30(11): 1343-53, 2015 Nov.
Article in English | MEDLINE | ID: mdl-24890016

ABSTRACT

Diallyl trisulfide (DATS), a chemopreventive dietary constituent and extracted from garlic, has been shown to against cultured many types of human cancer cell liens but the fate of apoptosis in murine leukemia cells in vitro and immune responses in leukemic mice remain elusive. Herein, we clarified the actions of DATS on growth inhibition of murine leukemia WEHI-3 cells in vitro and used WEHI-3 cells to generate leukemic mice in vivo, following to investigate the effects of DATS in animal model. In in vitro study, DATS induced apoptosis of WEHI-3 cells through the G0/G1 phase arrest and induction of caspase-3 activation. In in vivo study DATS decreased the weight of spleen of leukemia mice but did not affect the spleen weight of normal mice. DATS promoted the immune responses such as promotions of the macrophage phagocytosis and NK cell activities in WEHI-3 leukemic and normal mice. However, DATS only promotes NK cell activities in normal mice. DATS increases the surface markers of CD11b and Mac-3 in leukemia mice but only promoted CD3 in normal mice. In conclusion, the present study indicates that DATS induces cell death through induction of apoptosis in mice leukemia WHEI-3 cells. DATS also promotes immune responses in leukemia and normal mice in vivo.


Subject(s)
Allyl Compounds/pharmacology , Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Leukemia, Experimental/immunology , Leukemia, Experimental/prevention & control , Sulfides/pharmacology , Allyl Compounds/therapeutic use , Animals , Anticarcinogenic Agents/therapeutic use , Antigens, Differentiation/immunology , Caspase 3/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cytotoxicity, Immunologic/drug effects , Garlic/chemistry , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Phagocytosis/drug effects , Phagocytosis/immunology , Spleen/drug effects , Spleen/immunology , Sulfides/therapeutic use
17.
Retrovirology ; 11: 126, 2014 Dec 24.
Article in English | MEDLINE | ID: mdl-25539593

ABSTRACT

BACKGROUND: Pathogen recognition drives host defense towards viral infections. Specific groups rather than single members of the protein family of pattern recognition receptors (PRRs) such as membrane spanning Toll-like receptors (TLRs) and cytosolic helicases might mediate sensing of replication intermediates of a specific virus species. TLR7 mediates host sensing of retroviruses and could significantly influence retrovirus-specific antibody responses. However, the origin of efficient cell-mediated immunity towards retroviruses is unknown. Double-stranded RNA intermediates produced during retroviral replication are good candidates for immune stimulatory viral products. Thus, we considered TLR3 as primer of cell-mediated immunity against retroviruses in vivo. RESULTS: Infection of mice deficient in TLR3 (TLR3(-/-)) with Friend retrovirus (FV) complex revealed higher viral loads during acute retroviral infection compared to wild type mice. TLR3(-/-) mice exhibited significantly lower expression levels of type I interferons (IFNs) and IFN-stimulated genes like Pkr or Ifi44, as well as reduced numbers of activated myeloid dendritic cells (DCs) (CD86(+) and MHC-II(+)). DCs generated from FV-infected TLR3(-/-) mice were less capable of priming virus-specific CD8(+) T cell proliferation. Moreover, cytotoxicity of natural killer (NK) cells as well as CD8(+) T cells were reduced in vitro and in vivo, respectively, in FV-infected TLR3(-/-) mice. CONCLUSIONS: TLR3 mediates antiretroviral cytotoxic NK cell and CD8(+) T cell activity in vivo. Our findings qualify TLR3 as target of immune therapy against retroviral infections.


Subject(s)
Friend murine leukemia virus/immunology , Toll-Like Receptor 3/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Killer Cells, Natural/immunology , Leukemia, Experimental/immunology , Leukemia, Experimental/virology , Mice, Inbred C57BL , Mice, Knockout , Retroviridae Infections/immunology , Retroviridae Infections/virology , Toll-Like Receptor 3/deficiency , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Viral Load
18.
Blood ; 120(17): 3444-54, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22936657

ABSTRACT

BLT1 is a high-affinity receptor for leukotriene B4 (LTB4) that is a potent lipid chemoattractant for myeloid leukocytes. The role of LTB4/BLT1 axis in tumor immunology, including cytokine-based tumor vaccine, however, remains unknown. We here demonstrated that BLT1-deficient mice rejected subcutaneous tumor challenge of GM-CSF gene-transduced WEHI3B (WGM) leukemia cells (KO/WGM) and elicited robust antitumor responses against second tumor challenge with WEHI3B cells. During GM-CSF-induced tumor regression, the defective LTB4/BLT1 signaling significantly reduced tumor-infiltrating myeloid-derived suppressor cells, increased the maturation status of dendritic cells in tumor tissues, enhanced their CD4(+) T-cell stimulation capacity and migration rate of dendritic cells that had phagocytosed tumor-associated antigens into tumor-draining lymph nodes, suggesting a positive impact on GM-CSF-sensitized innate immunity. Furthermore, KO/WGM mice displayed activated adaptive immunity by attenuating regulatory CD4(+) T subsets and increasing numbers of Th17 and memory CD44(hi)CD4(+) T subsets, both of which elicited superior antitumor effects as evidenced by adoptive cell transfer. In vivo depletion assays also revealed that CD4(+) T cells were the main effectors of the persistent antitumor immunity. Our data collectively underscore a negative role of LTB4/BLT1 signaling in effective generation and maintenance of GM-CSF-induced antitumor memory CD4(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Leukemia, Experimental/immunology , Receptors, Leukotriene B4/immunology , Signal Transduction/immunology , Adaptive Immunity , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/pathology , Cell Differentiation , Cell Line, Tumor , Cell Movement , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Gene Expression Regulation, Neoplastic/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunity, Innate , Immunologic Memory , Leukemia, Experimental/genetics , Leukemia, Experimental/pathology , Leukotriene B4/immunology , Leukotriene B4/metabolism , Lymphocyte Activation , Male , Mice , Mice, Knockout , Receptors, Leukotriene B4/deficiency , Receptors, Leukotriene B4/genetics , Signal Transduction/genetics , Transduction, Genetic
19.
J Immunol ; 189(7): 3724-33, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22956581

ABSTRACT

Distal symmetrical polyneuropathy is the most common form of HIV infection-associated peripheral neuropathy and is often associated with pain. C57BL/6 (B6) mice infected with LP-BM5, a murine retroviral isolate, develop a severe immunodeficiency syndrome similar to that in humans infected with HIV-1, hence the term murine AIDS. We investigated the induction of peripheral neuropathy after LP-BM5 infection in B6 mice. Infected B6 mice, like HIV-infected humans, exhibited behavioral (increased sensitivity to mechanical and heat stimuli) and pathological (transient loss of intraepidermal nerve fibers) signs of peripheral neuropathy. The levels of viral gag RNA were significantly increased in all tissues tested, including spleen, paw skin, lumbar dorsal root ganglia, and lumbar spinal cord, postinfection (p.i.). Correlated with the development of peripheral neuropathy, the tissue levels of several cytokines, including IFN-γ, IL-1ß, IL-6, and IL-12, were significantly elevated p.i. These increases had cytokine-specific and tissue-specific profiles and kinetics. Further, treatment with the antiretroviral agent zidovudine either significantly reduced or completely reversed the aforementioned behavioral, pathologic, and cytokine changes p.i. These data suggest that LP-BM5 infection is a potential mouse model of HIV-associated distal symmetrical polyneuropathy that can be used for investigating the roles of various cytokines in infection-induced neuropathic pain. Further investigation of this model could give a better understanding of, and lead to more effective treatments for, HIV infection-associated painful peripheral neuropathy.


Subject(s)
Cytokines/biosynthesis , Leukemia Virus, Murine/immunology , Murine Acquired Immunodeficiency Syndrome/immunology , Murine Acquired Immunodeficiency Syndrome/metabolism , Peripheral Nervous System Diseases/immunology , Peripheral Nervous System Diseases/virology , Animals , Cytokines/genetics , Disease Models, Animal , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/virology , Leukemia Virus, Murine/isolation & purification , Leukemia, Experimental/immunology , Leukemia, Experimental/metabolism , Leukemia, Experimental/virology , Male , Mice , Mice, Inbred C57BL , Peripheral Nervous System Diseases/metabolism , RNA, Messenger/biosynthesis , Retroviridae Infections/immunology , Retroviridae Infections/metabolism
20.
J Immunol ; 189(5): 2521-9, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22821964

ABSTRACT

The immune system is tasked with defending against a myriad of microbial infections, and its response to a given infectious microbe may be strongly influenced by coinfection with another microbe. It was shown that infection of mice with lactate dehydrogenase-elevating virus (LDV) impairs early adaptive immune responses to Friend virus (FV) coinfection. To investigate the mechanism of this impairment, we examined LDV-induced innate immune responses and found LDV-specific induction of IFN-α and IFN-γ. LDV-induced IFN-α had little effect on FV infection or immune responses, but unexpectedly, LDV-induced IFN-γ production dampened Th1 adaptive immune responses and enhanced FV infection. Two distinct effects were identified. First, LDV-induced IFN-γ signaling indirectly modulated FV-specific CD8+ T cell responses. Second, intrinsic IFN-γ signaling in B cells promoted polyclonal B cell activation and enhanced early FV infection, despite promotion of germinal center formation and neutralizing Ab production. Results from this model reveal that IFN-γ production can have detrimental effects on early adaptive immune responses and virus control.


Subject(s)
Adaptive Immunity , Down-Regulation/immunology , Interferon-gamma/physiology , Leukemia Virus, Murine/immunology , Retroviridae Infections/immunology , Adaptive Immunity/genetics , Animals , Disease Models, Animal , Down-Regulation/genetics , Female , Friend murine leukemia virus/immunology , Friend murine leukemia virus/pathogenicity , Interferon-gamma/deficiency , Interferon-gamma/genetics , Lactate dehydrogenase-elevating virus/immunology , Lactate dehydrogenase-elevating virus/pathogenicity , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/genetics , Leukemia, Experimental/immunology , Leukemia, Experimental/virology , Mice , Mice, Congenic , Mice, Inbred A , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Retroviridae Infections/genetics , Retroviridae Infections/pathology , Spleen Focus-Forming Viruses/immunology , Spleen Focus-Forming Viruses/pathogenicity , Tumor Virus Infections/genetics , Tumor Virus Infections/immunology , Tumor Virus Infections/virology
SELECTION OF CITATIONS
SEARCH DETAIL