Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Heliyon ; 7(7): e07496, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34401556

ABSTRACT

Phagocytosis is a critical component of the innate immune response to viral infection, resulting in the clearance of infected cells while minimizing the exposure of uninfected cells. On the other hand, phagocytosis of HIV-infected T cells may cause phagocytes, such as macrophages and dendritic cells, to be infected, thus leading to HIV cell-to-cell transmission. V domain immunoglobulin suppressor of T cell activation (VISTA, gene Vsir, aliases Gi24, Dies-1, PD-1H, and DD1α) has been identified as an immune checkpoint molecule that possesses dual activities when expressed on APCs and T cells. Our study found that VISTA might play a significant role during the immune response to HIV infection via apoptosis upregulation and subsequent phagocytosis of infected CEM-SS T cells. HIV-induced apoptosis and monocytic cell engulfment were tested utilizing CEM-SS T cells as target cells and the monocytic cell line THP-1 as phagocytic cells. Cells were infected with a GFP-labeled HIV strain, NL4-3. HIV-infected CEM-SS T cells displayed greater apoptotic activity (approximately 18.0%) than mock-infected controls. Additionally, phagocytosis of HIV-infected CEM-SS T cells was increased approximately 4-fold. Expression of VISTA on infected CEM-SS T cells was detected in 16.7% of cells, which correlated with the increased phagocytosis observed. When an antagonistic antibody against VISTA was used, the number of phagocytosed cells was reduced by a factor of 2, which was replicated utilizing human stem cell-derived dendritic cells. Phagocytosis was also confirmed by the upregulation of IL-1ß expression, which was 5-fold higher in infected cells than in control cells. We also found that VISTA overexpression on both phagocytes and HIV-infected CEM-SS T cells facilitated phagocytosis. Our study suggests that VISTA may act as a direct ligand in the phagocytosis of HIV-infected T cells.

2.
Nat Metab ; 2(7): 635-647, 2020 07.
Article in English | MEDLINE | ID: mdl-32694789

ABSTRACT

T cells undergo metabolic rewiring to meet their bioenergetic, biosynthetic and redox demands following antigen stimulation. To fulfil these needs, effector T cells must adapt to fluctuations in environmental nutrient levels at sites of infection and inflammation. Here, we show that effector T cells can utilize inosine, as an alternative substrate, to support cell growth and function in the absence of glucose in vitro. T cells metabolize inosine into hypoxanthine and phosphorylated ribose by purine nucleoside phosphorylase. We demonstrate that the ribose subunit of inosine can enter into central metabolic pathways to provide ATP and biosynthetic precursors, and that cancer cells display diverse capacities to utilize inosine as a carbon source. Moreover, the supplementation with inosine enhances the anti-tumour efficacy of immune checkpoint blockade and adoptive T-cell transfer in solid tumours that are defective in metabolizing inosine, reflecting the capability of inosine to relieve tumour-imposed metabolic restrictions on T cells.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Carbon/metabolism , Glucose/deficiency , Inosine/metabolism , Adoptive Transfer , Animals , Cell Line, Tumor , HeLa Cells , Humans , Hypoxanthine/metabolism , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Nutrients , Purine-Nucleoside Phosphorylase/metabolism , Ribose/metabolism
3.
Front Oncol ; 9: 322, 2019.
Article in English | MEDLINE | ID: mdl-31114756

ABSTRACT

The adoptive transfer of T cells expressing chimeric antigen receptors (CARs) through genetic engineering is one of the most promising new therapies for treating cancer patients. A robust CAR T cell-mediated anti-tumor response requires the coordination of nutrient and energy supplies with CAR T cell expansion and function. However, the high metabolic demands of tumor cells compromise the function of CAR T cells by competing for nutrients within the tumor microenvironment (TME). To substantially improve clinical outcomes of CAR T immunotherapy while treating solid tumors, it is essential to metabolically prepare CAR T cells to overcome the metabolic barriers imposed by the TME. In this review, we discuss a potential metabolism toolbox to improve the metabolic fitness of CAR T cells and maximize the efficacy of CAR T therapy.

4.
Hum Gene Ther ; 29(5): 614-625, 2018 05.
Article in English | MEDLINE | ID: mdl-29334771

ABSTRACT

CD19-targeted chimeric antigen receptor (CAR) engineered T/natural killer (NK)-cell therapies can result in durable clinical responses in B-cell malignancies. However, CAR-based immunotherapies have been much less successful in solid cancers, in part due to "on-target off-tumor" toxicity related to expression of target tumor antigens on normal tissue. Based on preliminary observations of safety and clinical activity in proof-of-concept clinical trials, tumor antigen-specific messenger RNA (mRNA) CAR transfection into selected, activated, and expanded T/NK cells may permit prospective control of "on-target off-tumor" toxicity. To develop a commercial product for solid tumors, mesothelin was selected as an antigen target based on its association with poor prognosis and overexpression in multiple solid cancers. It was hypothesized that selecting, activating, and expanding cells ex vivo prior to mRNA CAR transfection would not be necessary, thus simplifying the complexity and cost of manufacturing. Now, the development of anti-human mesothelin mRNA CAR transfected peripheral blood lymphocytes (CARMA-hMeso) is reported, demonstrating the manufacture and cryopreservation of multiple cell aliquots for repeat administrations from a single human leukapheresis. A rapid, automated, closed system for cGMP-compliant transfection of mRNA CAR in up to 20 × 109 peripheral blood lymphocytes was developed. Here we show that CARMA-hMeso cells recognize and lyse tumor cells in a mesothelin-specific manner. Expression of CAR was detectable over approximately 7 days in vitro, with a progressive decline of CAR expression that appears to correlate with in vitro cell expansion. In a murine ovarian cancer model, a single intraperitoneal injection of CARMA-hMeso resulted in the dose-dependent inhibition of tumor growth and improved survival of mice. Furthermore, repeat weekly intraperitoneal administrations of the optimal CARMA-hMeso dose further prolonged disease control and survival. No significant off-target toxicities were observed. These data support further investigation of CARMA-hMeso as a potential treatment for ovarian cancer and other mesothelin-expressing cancers.


Subject(s)
GPI-Linked Proteins/immunology , Natural Killer T-Cells/transplantation , Ovarian Neoplasms/therapy , Receptors, Antigen, T-Cell/therapeutic use , Animals , Cell Line, Tumor , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/therapeutic use , Humans , Immunotherapy, Adoptive , Lymphocytes/immunology , Mesothelin , Mice , Natural Killer T-Cells/immunology , Ovarian Neoplasms/immunology , RNA, Messenger/genetics , RNA, Messenger/therapeutic use , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use , Xenograft Model Antitumor Assays
5.
EBioMedicine ; 25: 87-94, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29033368

ABSTRACT

A DNA vaccine encoding prM and E protein has been shown to induce protection against Zika virus (ZIKV) infection in mice and monkeys. However, its effectiveness in humans remains undefined. Moreover, identification of which immune cell types are specifically infected in humans is unclear. We show that human myeloid cells and B cells are primary targets of ZIKV in humanized mice. We also show that a DNA vaccine encoding full length prM and E protein protects humanized mice from ZIKV infection. Following administration of the DNA vaccine, humanized DRAG mice developed antibodies targeting ZIKV as measured by ELISA and neutralization assays. Moreover, following ZIKV challenge, vaccinated animals presented virtually no detectable virus in human cells and in serum, whereas unvaccinated animals displayed robust infection, as measured by qRT-PCR. Our results utilizing humanized mice show potential efficacy for a targeted DNA vaccine against ZIKV in humans.


Subject(s)
Antibodies, Neutralizing/administration & dosage , Vaccines, DNA/administration & dosage , Zika Virus Infection/pathology , Zika Virus/immunology , Animals , Antibodies, Neutralizing/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Mice, Transgenic , Vaccines, DNA/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Zika Virus/pathogenicity , Zika Virus Infection/immunology , Zika Virus Infection/virology
6.
JCI Insight ; 2(13)2017 Jul 06.
Article in English | MEDLINE | ID: mdl-28679955

ABSTRACT

A central issue for adoptive cellular immunotherapy is overcoming immunosuppressive signals to achieve tumor clearance. While γδ T cells are known to be potent cytolytic effectors that can kill a variety of cancers, it is not clear whether they are inhibited by suppressive ligands expressed in tumor microenvironments. Here, we have used a powerful preclinical model where EBV infection drives the de novo generation of human B cell lymphomas in vivo, and autologous T lymphocytes are held in check by PD-1/CTLA-4-mediated inhibition. We show that a single dose of adoptively transferred Vδ2+ T cells has potent antitumor effects, even in the absence of checkpoint blockade or activating compounds. Vδ2+ T cell immunotherapy given within the first 5 days of EBV infection almost completely prevented the outgrowth of tumors. Vδ2+ T cell immunotherapy given more than 3 weeks after infection (after neoplastic transformation is evident) resulted in a dramatic reduction in tumor burden. The immunotherapeutic Vδ2+ T cells maintained low cell surface expression of PD-1 in vivo, and their recruitment to tumors was followed by a decrease in B cells expressing PD-L1 and PD-L2 inhibitory ligands. These results suggest that adoptively transferred PD-1lo Vδ2+ T cells circumvent the tumor checkpoint environment in vivo.

7.
Hum Vaccin Immunother ; 13(7): 1548-1555, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28272967

ABSTRACT

Chimeric antigen receptor T cells are T cells genetically engineered with CAR constructs which mainly contain scFV and TCR zeta chain. With promising development in blood cancers, CAR T trials are also applied in solid cancers. However, the treatment effect in solid cancers is lower than expected. This review summarizes difference of CAR T applications in solid and blood cancers. Future challenges of CAR T cell treatment in solid cancer are also discussed using ovarian cancer as an example.


Subject(s)
Immunotherapy/methods , Ovarian Neoplasms/therapy , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Female , Humans
8.
Cell Rep ; 16(12): 3273-3285, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27653689

ABSTRACT

Invariant natural killer T (iNKT) cells are innate T lymphocytes that promote host defense against a variety of microbial pathogens. Whether microbial ligands are required for their protective effects remains unclear. Here, we show that iNKT cells stimulate human-monocyte-derived dendritic cells (DCs) to produce inflammatory mediators in a manner that does not require the presence of microbial compounds. Interleukin 2 (IL-2)-exposed iNKT cells selectively induced repeated cytoplasmic Ca(2+) fluxes in DCs that were dependent on signaling by the P2X7 purinergic receptor and mediated by ATP released during iNKT-DC interactions. Exposure to iNKT cells led to DC cyclooxygenase 2 (PTGS2) gene transcription, and release of PGE2 that was associated with vascular permeabilization in vivo. Additionally, soluble factors were released that induced neutrophil recruitment and activation and enhanced control of Candida albicans. These results suggest that sterile interactions between iNKT cells and monocyte-derived DCs lead to the production of non-redundant inflammatory mediators that promote neutrophil responses.


Subject(s)
Dendritic Cells/metabolism , Inflammation/immunology , Natural Killer T-Cells/immunology , Receptors, Purinergic P2X7/immunology , Animals , Dendritic Cells/immunology , Humans , Mice , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Natural Killer T-Cells/metabolism , Receptors, Purinergic P2X7/metabolism , Signal Transduction/immunology
9.
PLoS Pathog ; 12(5): e1005642, 2016 05.
Article in English | MEDLINE | ID: mdl-27186886

ABSTRACT

Epstein-Barr virus (EBV) infection causes B cell lymphomas in humanized mouse models and contributes to a variety of different types of human lymphomas. T cells directed against viral antigens play a critical role in controlling EBV infection, and EBV-positive lymphomas are particularly common in immunocompromised hosts. We previously showed that EBV induces B cell lymphomas with high frequency in a cord blood-humanized mouse model in which EBV-infected human cord blood is injected intraperitoneally into NOD/LtSz-scid/IL2Rγnull (NSG) mice. Since our former studies showed that it is possible for T cells to control the tumors in another NSG mouse model engrafted with both human fetal CD34+ cells and human thymus and liver, here we investigated whether monoclonal antibodies that block the T cell inhibitory receptors, PD-1 and CTLA-4, enhance the ability of cord blood T cells to control the outgrowth of EBV-induced lymphomas in the cord-blood humanized mouse model. We demonstrate that EBV-infected lymphoma cells in this model express both the PD-L1 and PD-L2 inhibitory ligands for the PD-1 receptor, and that T cells express the PD-1 and CTLA-4 receptors. Furthermore, we show that the combination of CTLA-4 and PD-1 blockade strikingly reduces the size of lymphomas induced by a lytic EBV strain (M81) in this model, and that this anti-tumor effect requires T cells. PD-1/CTLA-4 blockade markedly increases EBV-specific T cell responses, and is associated with enhanced tumor infiltration by CD4+ and CD8+ T cells. In addition, PD-1/CTLA-4 blockade decreases the number of both latently, and lytically, EBV-infected B cells. These results indicate that PD-1/CTLA-4 blockade enhances the ability of cord blood T cells to control outgrowth of EBV-induced lymphomas, and suggest that PD-1/CTLA-4 blockade might be useful for treating certain EBV-induced diseases in humans.


Subject(s)
Epstein-Barr Virus Infections/complications , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/virology , Programmed Cell Death 1 Receptor/metabolism , Animals , CTLA-4 Antigen/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Epstein-Barr Virus Infections/immunology , Fetal Blood , Flow Cytometry , Herpesvirus 4, Human , Humans , Lymphoma, B-Cell/metabolism , Mice , Mice, Inbred NOD , Mice, SCID
10.
J Clin Invest ; 125(1): 304-15, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25485679

ABSTRACT

Epstein-Barr virus (EBV) infection transforms B cells in vitro and is associated with human B cell lymphomas. The major EBV oncoprotein, latent membrane protein 1 (LMP1), mimics constitutively active CD40 and is essential for outgrowth of EBV-transformed B cells in vitro; however, EBV-positive diffuse large B cell lymphomas and Burkitt lymphomas often express little or no LMP1. Thus, EBV may contribute to the development and maintenance of human lymphomas even in the absence of LMP1. Here, we found that i.p. injection of human cord blood mononuclear cells infected with a LMP1-deficient EBV into immunodeficient mice induces B cell lymphomas. In this model, lymphoma development required the presence of CD4+ T cells in cord blood and was inhibited by CD40-blocking Abs. In contrast, LMP1-deficient EBV established persistent latency but did not induce lymphomas when directly injected into mice engrafted with human fetal CD34+ cells and human thymus. WT EBV induced lymphomas in both mouse models and did not require coinjected T cells in the cord blood model. Together, these results demonstrate that LMP1 is not essential for EBV-induced lymphomas in vivo and suggest that T cells supply signals that substitute for LMP1 in EBV-positive B cell lymphomagenesis.


Subject(s)
Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/genetics , Lymphoma/virology , Viral Matrix Proteins/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/metabolism , Carcinogenesis , Cell Proliferation , Epstein-Barr Virus Infections/immunology , Gene Expression , Gene Knockout Techniques , Herpesvirus 4, Human/immunology , Humans , Lymphoma/immunology , Lymphoma/pathology , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Signal Transduction , Tumor Cells, Cultured , Viral Matrix Proteins/metabolism , Virus Latency
SELECTION OF CITATIONS
SEARCH DETAIL
...