Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.902
Filter
2.
Cell Rep ; 37(6): 109991, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758311

ABSTRACT

The existence of a dysfunctional CD8+ T cell state in cancer is well established. However, the degree to which CD8+ T cell fates are influenced by the context in which they encounter cognate tumor antigen is less clear. We previously demonstrated that CD8+ T cells reactive to a model leukemia antigen were deleted by antigen cross-presenting type 1 conventional dendritic cells (cDC1s). Here, through a study of T cell receptor (TCR) transgenic CD8+ T cells (TCRTg101) reactive to a native C1498 leukemia cell antigen, we uncover a different mode of T cell tolerance in which TCRTg101 undergo progressive expansion and differentiation into an exhausted state. Antigen encounter by TCRTg101 requires leukemia cell major histocompatibility complex (MHC)-I expression and is independent of DCs, implying that leukemia cells directly mediate the exhausted TCRTg101 phenotype. Collectively, our data reveal that leukemia antigens are presented to CD8+ T cells via discrete pathways, leading to distinct tolerant states.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immune Tolerance , Leukemia, Experimental/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Cells, Cultured , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic
3.
Aging (Albany NY) ; 13(5): 7199-7210, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33653969

ABSTRACT

Chimeric antigen receptor (CAR) T cells target specific tumor antigens and lyse tumor cells in an MHC-independent manner. However, the efficacy of CAR-T cell and other cancer immunotherapies is limited by the expression of immune-checkpoint molecules such as programmed death-ligand 1 (PD-L1) on tumor cells, which binds to PD-1 receptors on T cells leading to T cell inactivation and immune escape. Here, we incorporated a PD-L1-targeted single-chain variable fragment (scFv) fusion protein sequence into a CAR vector to generate human anti-PD-L1-CAR-T cells (aPDL1-CART cells) targeting the PD-L1 antigen. Unlike control T cells, aPDL1-CART cells significantly halted the expansion and reduced the viability of co-cultured leukemia cells (Raji, CD46, and K562) overexpressing PD-L1, and this effect was paralleled by increased secretion of IL-2 and IFN-γ. The antitumor efficacy of aPDL1-CART cells was also evaluated in vivo by co-injecting control T cells or aPDL1-CART cells along with PDL1-CA46 cells to generate subcutaneous xenografts in NCG mice. Whereas large tumors developed in mice inoculated with PDL1-CA46 cells alone or together with control T cells, no tumor formation was detected in xenografts containing aPDL1-CART cells. Our data suggest that immune checkpoint-targeted CAR-T cells may be useful for controlling and eradicating immune-refractory hematological malignancies.


Subject(s)
B7-H1 Antigen/immunology , Immunotherapy, Adoptive/methods , Leukemia, Experimental/therapy , Animals , Cell Line, Tumor , Humans , Interferon-gamma/metabolism , Interleukin-2/metabolism , Leukemia, Experimental/immunology , Mice , Neoplasm Transplantation
4.
Leukemia ; 35(8): 2274-2284, 2021 08.
Article in English | MEDLINE | ID: mdl-33526858

ABSTRACT

Despite significant progress over the last few decades in the treatment of acute myeloid leukemia (AML), there still remains a major unmet medical need for this disease. Immunotherapy approaches for redirecting pan CD3+ T cells to target leukemia blasts have shown limited efficacy in clinical trials and often accompanied with severe toxicity in AML patients. We designed an alternative engager molecule (Anti-TRGV9/anti-CD123), a bispecific antibody that can simultaneously bind to the Vγ9 chain of the Vγ9Vδ2+ γδ T cell receptor and to AML target antigen, CD123, to selectively recruit Vγ9+ γδ T cells rather than pan T cells to target AML blasts. Our results suggest that prototypic bispecific antibodies (a) selectively activate Vγ9+ γδ T cells as judged by CD69 and CD25 surface expression, and intracellular Granzyme B expression, (b) selectively recruit Vγ9+ γδ T cells into cell-cell conjugate formation of γδ T cells with tumor cells indicating selective and effective engagement of effector and target tumor cells, and (c) mediate γδ T cell cytotoxicity (in vitro and in vivo) against tumor antigen-expressing cells. Collectively, these findings suggest that selectively redirecting Vγ9+ γδ T cells to target AML blasts has a potential for immunotherapy for AML patients and favors further exploration of this concept.


Subject(s)
Antibodies, Bispecific/immunology , Antineoplastic Agents, Immunological/pharmacology , Immunotherapy/methods , Leukemia, Experimental/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Receptors, Antigen, T-Cell, gamma-delta/immunology , Animals , Cytotoxicity, Immunologic , Humans , Leukemia, Experimental/immunology , Leukemia, Experimental/pathology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Leukemia ; 35(8): 2243-2257, 2021 08.
Article in English | MEDLINE | ID: mdl-33414484

ABSTRACT

Targeted T cell therapy is highly effective in disease settings where tumor antigens are uniformly expressed on malignant cells and where off-tumor on-target-associated toxicity is manageable. Although acute myeloid leukemia (AML) has in principle been shown to be a T cell-sensitive disease by the graft-versus-leukemia activity of allogeneic stem cell transplantation, T cell therapy has so far failed in this setting. This is largely due to the lack of target structures both sufficiently selective and uniformly expressed on AML, causing unacceptable myeloid cell toxicity. To address this, we developed a modular and controllable MHC-unrestricted adoptive T cell therapy platform tailored to AML. This platform combines synthetic agonistic receptor (SAR) -transduced T cells with AML-targeting tandem single chain variable fragment (scFv) constructs. Construct exchange allows SAR T cells to be redirected toward alternative targets, a process enabled by the short half-life and controllability of these antibody fragments. Combining SAR-transduced T cells with the scFv constructs resulted in selective killing of CD33+ and CD123+ AML cell lines, as well as of patient-derived AML blasts. Durable responses and persistence of SAR-transduced T cells could also be demonstrated in AML xenograft models. Together these results warrant further translation of this novel platform for AML treatment.


Subject(s)
Immunotherapy, Adoptive/methods , Leukemia, Experimental/therapy , Leukemia, Myeloid, Acute/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/transplantation , Animals , Female , Humans , Leukemia, Experimental/immunology , Leukemia, Experimental/pathology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , T-Lymphocytes/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
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
8.
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
9.
mBio ; 10(1)2019 01 22.
Article in English | MEDLINE | ID: mdl-30670616

ABSTRACT

Friend virus (FV) is a naturally occurring mouse retrovirus that infects dividing cells of the hematopoietic lineage, including antigen-presenting cells (APCs). The infection of APCs by viruses often induces their dysfunction, and it has been shown that FV infection reduces the ability of dendritic cells (DCs) to prime critical CD8+ T cell responses. Nonetheless, mice mount vigorous CD8+ T cell responses, so we investigated whether B cells might serve as alternative APCs during FV infection. Direct ex vivo analysis of B cells from FV-infected mice revealed that infected but not uninfected B cells upregulated expression of the costimulatory molecules CD80, CD86, and CD40, as well as major histocompatibility complex class II (MHC-II) molecules. Furthermore, in vitro studies showed that, compared to uninfected B cells from the same mice, the FV-infected B cells had significantly enhanced APC function, as measured by their capacity to prime CD8+ T cell activation and proliferation. Thus, in contrast to DCs, infection of B cells with FV enhanced their APC capacity and ability to stimulate the CD8+ T cell responses essential for virus control. FV infections also induce the activation and expansion of regulatory T cells (Tregs), so it was of interest to determine the impact of Tregs on B cell activation. The upregulation of costimulatory molecule expression and APC function of B cells was even more strongly enhanced by in vivo depletion of regulatory T cells than infection. Thus, Tregs exert potent homeostatic suppression of B cell activation that is partially overcome by FV infection.IMPORTANCE The primary role of B cells in immunity is considered the production of pathogen-specific antibodies, but another, less-well-studied, function of B cells is to present foreign antigens to T cells to stimulate their activation and proliferation. Dendritic cells (DCs) are considered the most important antigen-presenting cells (APCs) for CD8+ T cells, but DCs lose APC function when infected with Friend virus (FV), a model retrovirus of mice. Interestingly, B cells were better able to stimulate CD8+ T cell responses when they were infected with FV. We also found that the activation status of B cells under homeostatic conditions was potently modulated by regulatory T cells. This study illustrates an important link between B cell and T cell responses and illustrates an additional mechanism by which regulatory T cells suppress critical T cell responses during viral infections.


Subject(s)
Antigen Presentation , B-Lymphocytes/immunology , Friend murine leukemia virus/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B-Lymphocytes/chemistry , B7-1 Antigen/analysis , B7-2 Antigen/analysis , CD40 Antigens/analysis , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Histocompatibility Antigens Class II/analysis , Leukemia, Experimental/immunology , Leukemia, Experimental/virology , Lymphocyte Activation , Mice , Retroviridae Infections/immunology , Retroviridae Infections/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/virology
10.
JCI Insight ; 3(9)2018 05 03.
Article in English | MEDLINE | ID: mdl-29720574

ABSTRACT

Using a mouse retroviral model, we have shown that mAb-based immunotherapy can induce life-long endogenous protective immunity (vaccine-like effects). This observation has potentially important consequences for treating life-threatening human viral infections. Here, we investigated the role of neutrophils in this effect. Neutrophils are innate immunity effector cells with well-established microbe-killing activities that are rapidly mobilized upon infection. They are also emerging as orchestrators of innate and adaptive immunities. However, their immunomodulatory activity during antiviral mAb immunotherapies has never been studied. Our data reveal that neutrophils have an essential role in immunotherapy-induced immune protection of infected mice. Unexpectedly, neutrophils have a limited effect in controlling viral propagation upon passive immunotherapy administration, which is mostly mediated by NK cells. Instead, neutrophils operate as essential inducers of a potent host humoral antiviral response. Thus, neutrophils play an unexpected key role in protective immunity induction by antiviral mAbs. Our work opens approaches to improve antiviral immunotherapies, as it suggests that preserving neutrophil functions and counts might be required for achieving mAb-induced protective immunity.


Subject(s)
Antibodies, Monoclonal/immunology , Killer Cells, Natural/immunology , Leukemia Virus, Murine , Leukemia, Experimental/immunology , Neutrophils/immunology , Retroviridae Infections/immunology , Tumor Virus Infections/immunology , Virus Replication , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Humoral , Immunity, Innate , Immunotherapy , Leukemia, Erythroblastic, Acute/immunology , Mice
11.
PLoS One ; 13(4): e0195402, 2018.
Article in English | MEDLINE | ID: mdl-29614127

ABSTRACT

Recent vaccine studies with experimental antigens have shown that regulatory T cells (Tregs) constrain the magnitude of B cell responses. This homeostatic Treg-mediated suppression is thought to reduce the potential of germinal center (GC) responses to generate autoreactive antibodies. However, essentially opposite results were observed in live influenza infections where Tregs promoted B cell and antibody responses. Thus, it remains unclear whether Tregs dampen or enhance B cell responses, especially during live viral infections. Here, we use mice infected with Friend retrovirus (FV), which induces a robust expansion of Tregs. Depletion of Tregs led to elevated activation, proliferation, and class switching of B cells. In addition, Treg depletion enhanced the production of virus-specific and virus-neutralizing antibodies and reduced FV viremia. Thus, in contrast to influenza infection, Tregs either directly or indirectly suppress B cells during mouse retroviral infection indicating that the ultimate effect of Tregs on B cell responses is specific to the particular infectious agent.


Subject(s)
Antibodies, Viral/metabolism , Friend murine leukemia virus/immunology , Leukemia, Experimental/immunology , Retroviridae Infections/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Virus Infections/immunology , Animals , B-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunoglobulin G/metabolism , Mice, Transgenic , Spleen/immunology
12.
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
13.
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
14.
Sci Rep ; 7(1): 7785, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28798348

ABSTRACT

CD4+ helper T cells and cytotoxic CD8+ T cells are key players for adaptive immune responses against acute infections with retroviruses. Similar to textbook knowledge the most important function of CD4+ T cells during an acute retrovirus infection seems to be their helper function for other immune cells. Whereas there was no direct anti-viral activity of CD4+ T cells during acute Friend Virus (FV) infection, they were absolutely required for the control of chronic infection. During chronic FV infection a population of activated FV-specific CD4+ T cells did not express cytotoxic molecules, but Fas Ligand that can induce Fas-induced apoptosis in target cells. Using an MHC II-restricted in vivo CTL assay we demonstrated that FV-specific CD4+ T cells indeed mediated cytotoxic effects against FV epitope peptide loaded targets. CD4 + CTL killing was also detected in FV-infected granzyme B knockout mice confirming that the exocytosis pathway was not involved. However, killing could be blocked by antibodies against FasL, which identified the Fas/FasL pathway as critical cytotoxic mechanism during chronic FV infection. Interestingly, targeting the co-stimulatory receptor CD137 with an agonistic antibody enhanced CD4+ T cell cytotoxicity. This immunotherapy may be an interesting new approach for the treatment of chronic viral infections.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Fas Ligand Protein/immunology , Leukemia, Experimental/immunology , Retroviridae Infections/immunology , Tumor Virus Infections/immunology , Animals , Apoptosis , Cells, Cultured , Female , Friend murine leukemia virus/immunology , Mice , Mice, Inbred C57BL
15.
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
16.
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
17.
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
18.
Oncotarget ; 7(21): 30804-19, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27127171

ABSTRACT

The induction of long-lived effector CD8+ T cells is key to the development of efficient cancer vaccines. In this study, we demonstrated that a Toll-like receptor 2 (TLR2) agonist-fused antigen increased antigen presentation via TLR2 signaling and induced effector memory-like CD8+ T cells against cancer after immunization. The N-terminus of ovalbumin (OVA) was biologically fused with a bacterial lipid moiety TLR2 agonist to produce a recombinant lipidated ovalbumin (rlipo-OVA). We demonstrated that rlipo-OVA activated bone marrow-derived dendritic cells (BM-DCs) maturation and increased antigen presentation by major histocompatibility complex (MHC) class I via TLR2. After immunization, rlipo-OVA skewed the immune response towards T helper (Th) 1 and induced OVA-specific cytotoxic T lymphocyte (CTL) responses. Moreover, immunization with rlipo-OVA induced higher numbers of effector memory (CD44+CD62L-) CD8+ T cells compared with recombinant ovalbumin (rOVA) alone or rOVA mixed with the TLR2 agonist Pam3CSK4. Accordingly, the CD27+CD43+ effector memory CD8+ T cells expressed high levels of the long-lived CD127 marker. The administration of rlipo-OVA could inhibit tumor growth, but the anti-tumor effects were lost after the depletion of CD8 or CD127 cells in vivo. These findings suggested that the TLR2 agonist-fused antigen induced long-lived memory CD8+ T cells for efficient cancer therapy.


Subject(s)
Antigen Presentation/immunology , Cancer Vaccines/immunology , Leukemia, Experimental/therapy , Ovalbumin/immunology , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 2/agonists , Animals , Bone Marrow Cells/immunology , CD8 Antigens/metabolism , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Dendritic Cells/immunology , Female , Histocompatibility Antigens Class I/immunology , Interleukin-7 Receptor alpha Subunit/metabolism , Leukemia, Experimental/immunology , Lipopeptides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism
19.
J Leukoc Biol ; 100(1): 143-53, 2016 07.
Article in English | MEDLINE | ID: mdl-26792819

ABSTRACT

Leukemia dissemination (the spread of leukemia cells from the bone marrow) and relapse are associated with poor prognosis. Often, relapse occurs in peripheral organs, such as the CNS, which acts as a sanctuary site for leukemia cells to escape anti-cancer treatments. Similar to normal leukocyte migration, leukemia dissemination entails migration of cells from the blood circulation into tissues by extravasation. To extravasate, leukemia cells cross through vascular endothelial walls via a process called transendothelial migration, which requires cytoskeletal remodeling. However, the specific molecular players in leukemia extravasation are not fully known. We examined the role of myosin-IIA a cytoskeletal class II myosin motor protein, in leukemia progression and dissemination into the CNS by use of a mouse model of Bcr-Abl-driven B cell acute lymphoblastic leukemia. Small hairpin RNA-mediated depletion of myosin-IIA did not affect apoptosis or the growth rate of B cell acute lymphoblastic leukemia cells. However, in an in vivo leukemia transfer model, myosin-IIA depletion slowed leukemia progression and prolonged survival, in part, by reducing the ability of B cell acute lymphoblastic leukemia cells to engraft efficiently. Finally, myosin-IIA inhibition, either by small hairpin RNA depletion or chemical inhibition by blebbistatin, drastically reduced CNS infiltration of leukemia cells. The effects on leukemia cell entry into tissues were mostly a result of the requirement for myosin-IIA to enable leukemia cells to complete the transendothelial migration process during extravasation. Overall, our data implicate myosin-IIA as a key mediator of leukemia cell migration, making it a promising target to inhibit leukemia dissemination in vivo and potentially reduce leukemia relapses.


Subject(s)
Brain/immunology , Cell Movement , Disease Models, Animal , Leukemia, Experimental/immunology , Nonmuscle Myosin Type IIA/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Animals , Apoptosis , Brain/metabolism , Brain/pathology , Cell Adhesion , Cells, Cultured , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Male , Mice , Mice, Inbred C57BL , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...