Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Ther Oncolytics ; 25: 174-188, 2022 Jun 16.
Article in English | MEDLINE | ID: mdl-35592387

ABSTRACT

Clinical studies have demonstrated that local expression of the cytokine IL-12 drives interferon-gamma expression and recruits T cells to the tumor microenvironment, ultimately yielding durable systemic T cell responses. Interrogation of longitudinal biomarker data from our late-stage melanoma trials identified a significant on-treatment increase of intratumoral CXCR3 transcripts that was restricted to responding patients, underscoring the clinical relevance of tumor-infiltrating CXCR3+ immune cells. In this study, we sought to understand if the addition of DNA-encodable CXCL9 could augment the anti-tumor immune responses driven by intratumoral IL-12. We show that localized IL-12 and CXCL9 treatment reshapes the tumor microenvironment to promote dendritic cell licensing and CD8+ T cell activation. Additionally, this combination treatment results in a significant abscopal anti-tumor response and provides a concomitant benefit to anti-PD-1 therapies. Collectively, these data demonstrate that a functional tumoral CXCR3/CXCL9 axis is critical for IL-12 anti-tumor efficacy. Furthermore, restoring or amplifying the CXCL9 gradient in the tumors via intratumoral electroporation of plasmid CXCL9 can not only result in efficient trafficking of cytotoxic CD8+ T cells into the tumor but can also reshape the microenvironment to promote systemic immune response.

2.
Mol Cancer Res ; 20(6): 983-995, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35302641

ABSTRACT

Intratumoral delivery of plasmid IL12 via electroporation (IT-tavo-EP) induces localized expression of IL12 leading to regression of treated and distant tumors with durable responses and minimal toxicity. A key driver in amplifying this local therapy into a systemic response is the magnitude and composition of immune infiltrate in the treated tumor. While intratumoral IL12 typically increases the density of CD3+ tumor-infiltrating lymphocytes (TIL), this infiltrate is composed of a broad range of T-cell subsets, including activated tumor-specific T cells, less functional bystander T cells, as well as suppressive T regulatory cells. To encourage a more favorable on-treatment tumor microenvironment (TME), we explored combining this IL12 therapy with an intratumoral polyclonal T-cell stimulator membrane-anchored anti-CD3 to productively engage a diverse subset of lymphocytes including the nonreactive and suppressive T cells. This study highlighted that combined intratumoral electroporation of IL12 and membrane-anchored anti-CD3 plasmids can enhance cytokine production, T-cell cytotoxicity, and proliferation while limiting the suppressive capacity within the TME. These collective antitumor effects not only improve regression of treated tumors but drive systemic immunity with control of nontreated contralateral tumors in vivo. Moreover, combination of IL12 and anti-CD3 restored the function of TIL isolated from a patient with melanoma actively progressing on programmed cell death protein 1 (PD-1) checkpoint inhibitor therapy. IMPLICATIONS: This DNA-encodable polyclonal T-cell stimulator (membrane-anchored anti-CD3 plasmid) may represent a key addition to intratumoral IL12 therapies in the clinic.


Subject(s)
Interleukin-12 , Melanoma , Electroporation , Humans , Immunotherapy , Interleukin-12/genetics , Interleukin-12/metabolism , Melanoma/pathology , Plasmids/genetics , Tumor Microenvironment
3.
PLoS Pathog ; 16(12): e1009136, 2020 12.
Article in English | MEDLINE | ID: mdl-33370418

ABSTRACT

The level of CD40 expression on dendritic cells (DCs) plays a decisive role in disease protection during Leishmania donovani (LD) infection. However, current understanding of the molecular regulation of CD40 expression remains elusive. Using molecular, cellular and functional approaches, we identified a role for Runx1 and Runx3 transcription factors in the regulation of CD40 expression in DCs. In response to lipopolysaccharide (LPS), tumor necrosis factor alpha (TNFα) or antileishmanial drug sodium antimony gluconate (SAG), both Runx1 and Runx3 translocated to the nucleus, bound to the CD40 promoter and upregulated CD40 expression on DCs. These activities of Runx proteins were mediated by the upstream phosphatidylinositol 3-kinase (PI3K)-Akt pathway. Notably, LD infection attenuated LPS- or TNFα-induced CD40 expression in DCs by inhibiting PI3K-Akt-Runx axis via protein tyrosine phosphatase SHP-1. In contrast, CD40 expression induced by SAG was unaffected by LD infection, as SAG by blocking LD-induced SHP-1 activation potentiated PI3K-Akt signaling to drive Runx-mediated CD40 upregulation. Adoptive transfer experiments further showed that Runx1 and Runx3 play a pivotal role in eliciting antileishmanial immune response of SAG-treated DCs in vivo by promoting CD40-mediated type-1 T cell responses. Importantly, antimony-resistant LD suppressed SAG-induced CD40 upregulation on DCs by blocking the PI3K-Akt-Runx pathway through sustained SHP-1 activation. These findings unveil an immunoregulatory role for Runx proteins during LD infection.


Subject(s)
CD40 Antigens/immunology , Core Binding Factor alpha Subunits/immunology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Leishmaniasis, Visceral/immunology , Animals , CD40 Antigens/biosynthesis , Cricetinae , Humans , Leishmania donovani/immunology , Mice , Mice, Inbred BALB C
4.
Bioinformation ; 16(1): 13-16, 2020.
Article in English | MEDLINE | ID: mdl-32025155

ABSTRACT

Bioinformatics has evolved from providing basic solutions, such as sequence alignment, structure predictions, and phylogenetic analysis to an independent data-driven field. The unprecedented growth of genomic technologies and the enormous data have opened an avenue for bioinformaticians (Bioinformatics professionals) never been seen before in the history of mankind. The novel opportunity also requires creative solutions that need skills to deal with noisy, unstructured information to offer valuable biological insights. Currently, we are seeing only the tip of an iceberg and the future will revolve around big data sets in all forms of biological research. The emerging challenge is to unfold the hidden iceberg of data.

5.
J Exp Med ; 216(9): 2128-2149, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31227543

ABSTRACT

High numbers of tissue-resident memory T (TRM) cells are associated with better clinical outcomes in cancer patients. However, the molecular characteristics that drive their efficient immune response to tumors are poorly understood. Here, single-cell and bulk transcriptomic analysis of TRM and non-TRM cells present in tumor and normal lung tissue from patients with lung cancer revealed that PD-1-expressing TRM cells in tumors were clonally expanded and enriched for transcripts linked to cell proliferation and cytotoxicity when compared with PD-1-expressing non-TRM cells. This feature was more prominent in the TRM cell subset coexpressing PD-1 and TIM-3, and it was validated by functional assays ex vivo and also reflected in their chromatin accessibility profile. This PD-1+TIM-3+ TRM cell subset was enriched in responders to PD-1 inhibitors and in tumors with a greater magnitude of CTL responses. These data highlight that not all CTLs expressing PD-1 are dysfunctional; on the contrary, TRM cells with PD-1 expression were enriched for features suggestive of superior functionality.


Subject(s)
Gene Expression Profiling , Immunologic Memory/genetics , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Single-Cell Analysis , T-Lymphocytes/immunology , Transcriptome/genetics , Cell Proliferation , Clone Cells , Cytotoxicity, Immunologic/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Lung/metabolism , Lung/pathology , Lymphocyte Subsets/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic
6.
J Clin Invest ; 129(3): 1193-1210, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30620725

ABSTRACT

Genetic variants at the PTPN2 locus, which encodes the tyrosine phosphatase PTPN2, cause reduced gene expression and are linked to rheumatoid arthritis (RA) and other autoimmune diseases. PTPN2 inhibits signaling through the T cell and cytokine receptors, and loss of PTPN2 promotes T cell expansion and CD4- and CD8-driven autoimmunity. However, it remains unknown whether loss of PTPN2 in FoxP3+ regulatory T cells (Tregs) plays a role in autoimmunity. Here we aimed to model human autoimmune-predisposing PTPN2 variants, the presence of which results in a partial loss of PTPN2 expression, in mouse models of RA. We identified that reduced expression of Ptpn2 enhanced the severity of autoimmune arthritis in the T cell-dependent SKG mouse model and demonstrated that this phenotype was mediated through a Treg-intrinsic mechanism. Mechanistically, we found that through dephosphorylation of STAT3, PTPN2 inhibits IL-6-driven pathogenic loss of FoxP3 after Tregs have acquired RORγt expression, at a stage when chromatin accessibility for STAT3-targeted IL-17-associated transcription factors is maximized. We conclude that PTPN2 promotes FoxP3 stability in mouse RORγt+ Tregs and that loss of function of PTPN2 in Tregs contributes to the association between PTPN2 and autoimmunity.


Subject(s)
Arthritis, Rheumatoid/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Disease Models, Animal , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , T-Lymphocytes, Regulatory/pathology
7.
Cytokine ; 110: 39-43, 2018 10.
Article in English | MEDLINE | ID: mdl-29702464

ABSTRACT

Trafficking of dendritic cells (DCs) from peripheral tissues to draining lymph nodes is a prerequisite for induction of adaptive immunity. An immunosuppressive cytokine transforming growth factor (TGF)-ß1, however, inhibits migration of DCs by downregulating the expression of chemokine receptor CCR-7. Whether TGF-ß1 engages any other receptor to mediate this inhibitory effect is currently unknown. In this article, we report that TGF-ß1 attenuated the lymph node homing ability of mouse DCs by reducing C-type lectin receptor-2 (CLEC-2) expression. Notably, TGF-ß1 inhibited CLEC-2 expression in DCs via c-Src. DCs silenced for c-Src were resistant to TGF-ß1-induced inhibition of CLEC-2 expression. Furthermore, silencing of c-Src substantially improved the lymph node homing capacity of TGF-ß1-treated DCs by restoring CLEC-2 expression. These results document a critical role for c-Src and CLEC-2 in TGF-ß1-mediated impairment of DC migration and define a previously unknown mechanism by which TGF-ß1 attenuates the lymph node homing ability of DCs.


Subject(s)
Dendritic Cells/metabolism , Down-Regulation/physiology , Lectins, C-Type/metabolism , Lymph Nodes/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Gene Expression/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , src Homology Domains/physiology
8.
Biochim Biophys Acta Gen Subj ; 1862(4): 895-906, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29288771

ABSTRACT

BACKGROUND: Elevated levels of inflammatory molecules are key players in muscle wasting/atrophy leading to human morbidity. TNFα is a well-known pro-inflammatory cytokine implicated in the pathogenesis of muscle wasting under diverse clinical settings. S-allyl cysteine (SAC), an active component of garlic (Allium sativum), has established anti-oxidant and anti-inflammatory effects in various cell types. However, the impact of SAC on skeletal muscle pathology remains unexplored. Owing to the known anti-inflammatory properties of SAC, we investigated whether pre-treatment with SAC has a protective role in TNFα-induced atrophy in cultured myotubes. METHODS AND RESULTS: C2C12 myotubes were treated with TNFα (100ng/ml) in the presence or absence of SAC (0.01mM). TNFα treatment induced atrophy in myotubes by up-regulating various proteolytic systems i.e. cathepsin L, calpain, ubiquitin-proteasome E3-ligases (MuRF1/atrogin1), caspase 3 and autophagy (Beclin1/LC3B). TNFα also induced the activation of NFκB by stimulating the degradation of IκBα (inhibitor of NFκB), in myotubes. The alterations in proteolytic systems likely contribute to the degradation of muscle-specific proteins and reduce the myotube length, diameter and fusion index. The SAC supplementation significantly impedes TNFα-induced protein loss and protects myotube morphology by suppressing protein catabolic systems and endogenous level of inflammatory molecules namely TNFα, IL-6, IL-1ß, TNF-like weak inducer of apoptosis (TWEAK), fibroblast growth factor-inducible 14 (Fn14) and Nox. CONCLUSION AND GENERAL SIGNIFICANCE: Our findings reveal anti-atrophic role for SAC, as it prevents alterations in protein metabolism and protects myotubes by regulating the level of inflammatory molecules and multiple proteolytic systems responsible for muscle atrophy.


Subject(s)
Cysteine/analogs & derivatives , Inflammation Mediators/metabolism , Muscle, Skeletal/drug effects , Muscular Atrophy/prevention & control , Tumor Necrosis Factor-alpha/pharmacology , Animals , Autophagy/drug effects , Autophagy/genetics , Cell Line , Cysteine/pharmacology , Cytokines/genetics , Cytokines/metabolism , Gene Expression/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Proteolysis/drug effects , TWEAK Receptor/genetics , TWEAK Receptor/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
J Immunol ; 197(5): 1650-62, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27439518

ABSTRACT

The enhanced expression of T cell Ig and mucin protein-3 (TIM-3) on tumor-associated dendritic cells (DCs) attenuates antitumor effects of DNA vaccines. To identify a potential target (or targets) for reducing TIM-3 expression on tumor-associated DCs, we explored the molecular mechanisms regulating TIM-3 expression. In this study, we have identified a novel signaling pathway (c-Src→Bruton's tyrosine kinase→transcription factors Ets1, Ets2, USF1, and USF2) necessary for TIM-3 upregulation on DCs. Both IL-10 and TGF-ß, which are produced in the tumor microenvironment, upregulated TIM-3 expression on DCs via this pathway. Suppressed expression of c-Src or downstream Bruton's tyrosine kinase, Ets1, Ets2, USF1, or USF2 blocked IL-10- and TGF-ß-induced TIM-3 upregulation on DCs. Notably, in vivo knockdown of c-Src in mice reduced TIM-3 expression on tumor-associated DCs. Furthermore, adoptive transfer of c-Src-silenced DCs in mouse tumors enhanced the in vivo antitumor effects of immunostimulatory CpG DNA; however, TIM-3 overexpression in c-Src-silenced DCs blocked this effect. Collectively, our data reveal the molecular mechanism regulating TIM-3 expression in DCs and identify c-Src as a target for improving the efficacy of nucleic acid-mediated anticancer therapy.


Subject(s)
Dendritic Cells/immunology , Genes, src , Hepatitis A Virus Cellular Receptor 2/metabolism , Neoplasms/immunology , T-Lymphocytes/immunology , src-Family Kinases/metabolism , Adoptive Transfer , Agammaglobulinaemia Tyrosine Kinase , Animals , CSK Tyrosine-Protein Kinase , Cell Differentiation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hepatitis A Virus Cellular Receptor 2/genetics , Interleukin-10/immunology , Interleukin-10/metabolism , Mice , Neoplasms/metabolism , Oligodeoxyribonucleotides/immunology , Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Up-Regulation
10.
J Immunol ; 193(7): 3417-25, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25172495

ABSTRACT

The receptor T cell Ig and mucin protein-3 (TIM-3) has emerged as an important regulator of innate immune responses. However, whether TIM-3-induced signaling promotes or inhibits the activation and maturation of dendritic cells (DCs) still remains uncertain. In addition, the TIM-3 signaling events involved in this immunoregulatory function are yet to be established. In this article, we report that TIM-3 crosslinking by anti-TIM-3 Ab inhibited DC activation and maturation by blocking the NF-κB pathway. After Ab-mediated crosslinking, TIM-3 became tyrosine phosphorylated, which then sequentially bound and activated the nonreceptor tyrosine kinases Bruton's tyrosine kinase (Btk) and c-Src. Activation of Btk-c-Src signaling in turn triggered the secretion of some inhibitory factor (or factors) from DCs that inhibited the NF-κB pathway and subsequent activation and maturation of DCs. Silencing of Btk or c-Src abrogated the inhibitory effects of TIM-3 on DCs. These results demonstrate an essential role for Btk-c-Src signaling in TIM-3-induced DC suppression. Thus, in addition to demonstrating an inhibitory role for TIM-3 signaling in DC activation, we define the molecular mechanism by which TIM-3 mediates this effect.


Subject(s)
Dendritic Cells/immunology , Protein-Tyrosine Kinases/immunology , Receptors, Virus/immunology , Signal Transduction/immunology , src-Family Kinases/immunology , Agammaglobulinaemia Tyrosine Kinase , Animals , CSK Tyrosine-Protein Kinase , Dendritic Cells/cytology , Gene Silencing , Hepatitis A Virus Cellular Receptor 2 , Mice , Mice, Inbred BALB C , NF-kappa B/genetics , NF-kappa B/immunology , Protein-Tyrosine Kinases/genetics , Receptors, Virus/genetics , Signal Transduction/genetics , src-Family Kinases/genetics
11.
Microbiol Immunol ; 57(5): 374-85, 2013 May.
Article in English | MEDLINE | ID: mdl-23668610

ABSTRACT

The coat protein of cardamom mosaic virus (CdMV), a member of the genus Macluravirus, assembles into virus-like particles when expressed in an Escherichia coli expression system. The N and C-termini of the coat protein were engineered with the Kennedy peptide and the 2F5 and 4E10 epitopes of gp41 of HIV. The chimeric proteins reacted with sera from HIV positive persons and also stimulated secretion of cytokines by peripheral blood mononuclear cells from these persons. Thus, a system based on the coat protein of CdMV can be used to display HIV-1 antigens.


Subject(s)
Capsid Proteins/immunology , Cytokines/metabolism , Drug Carriers , Epitopes/immunology , HIV Antibodies/blood , HIV Envelope Protein gp41/immunology , Leukocytes, Mononuclear/immunology , Capsid Proteins/genetics , Epitopes/genetics , HIV Envelope Protein gp41/genetics , Potyviridae/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...