Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Annu Rev Med ; 68: 139-152, 2017 01 14.
Article in English | MEDLINE | ID: mdl-27860544

ABSTRACT

The field of cancer immunotherapy has been re-energized by the application of chimeric antigen receptor (CAR) T cell therapy in cancers. These CAR T cells are engineered to express synthetic receptors that redirect polyclonal T cells to surface antigens for subsequent tumor elimination. Many CARs are designed with elements that augment T cell persistence and activity. To date, CAR T cells have demonstrated tremendous success in eradicating hematologic malignancies (e.g., CD19 CARs in leukemias). However, this success has yet to be extrapolated to solid tumors, and the reasons for this are being actively investigated. We characterize some of the challenges that CAR T cells have to surmount in the solid tumor microenvironment and new approaches that are being considered to overcome these hurdles.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy, Adoptive/methods , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/therapeutic use , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Humans , Immune Tolerance , Immunity, Cellular , Lymphocytes, Tumor-Infiltrating/immunology , Receptors, Antigen, T-Cell/immunology
2.
EBioMedicine ; 13: 99-112, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27769803

ABSTRACT

Foxp3+ T-regulatory (Treg) cells are known to suppress protective host immune responses to a wide variety of solid tumors, but their therapeutic targeting is largely restricted to their transient depletion or "secondary" modulation, e.g. using anti-CTLA-4 monoclonal antibody. Our ongoing studies of the post-translational modifications that regulate Foxp3 demonstrated that the histone/protein acetyltransferase, Tip60, plays a dominant role in promoting acetylation, dimerization and function in Treg cells. We now show that the ubiquitin-specific protease, Usp7, controls Treg function largely by stabilizing the expression and promoting the multimerization of Tip60 and Foxp3. Genetic or pharmacologic targeting of Usp7 impairs Foxp3+ Treg suppressive functions, while conventional T cell responses remain intact. As a result, pharmacologic inhibitors of Usp7 can limit tumor growth in immunocompetent mice, and promote the efficacy of antitumor vaccines and immune checkpoint therapy with anti-PD1 monoclonal antibody in murine models. Hence, pharmacologic therapy with Usp7 inhibitors may have an important role in future cancer immunotherapy.


Subject(s)
Forkhead Transcription Factors/metabolism , Histone Acetyltransferases/metabolism , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Trans-Activators/metabolism , Ubiquitin-Specific Proteases/antagonists & inhibitors , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Cell Line, Tumor , Disease Models, Animal , Gene Expression , Immunity , Lymphocyte Activation/immunology , Lysine Acetyltransferase 5 , Mice , Mice, Knockout , Mice, Transgenic , Neoplasms/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Burden , Ubiquitin-Specific Peptidase 7 , Ubiquitin-Specific Proteases/genetics
3.
Mol Ther Oncolytics ; 3: 16006, 2016.
Article in English | MEDLINE | ID: mdl-27162934

ABSTRACT

Chimeric antigen receptor (CAR) T cells are engineered constructs composed of synthetic receptors that direct T cells to surface antigens for subsequent elimination. Many CAR constructs are also manufactured with elements that augment T-cell persistence and activity. To date, CAR T cells have demonstrated tremendous success in eradicating hematological malignancies (e.g., CD19 CARs in leukemias). This success is not yet extrapolated to solid tumors, and the reasons for this are being actively investigated. Here in this mini-review, we discuss some of the key hurdles encountered by CAR T cells in the solid tumor microenvironment.

4.
Cancer Immunol Res ; 4(6): 541-51, 2016 06.
Article in English | MEDLINE | ID: mdl-27045023

ABSTRACT

Antitumor treatments based on the infusion of T cells expressing chimeric antigen receptors (CAR T cells) are still relatively ineffective for solid tumors, due to the presence of immunosuppressive mediators [such as prostaglandin E2 (PGE2) and adenosine] and poor T-cell trafficking. PGE2 and adenosine activate protein kinase A (PKA), which then inhibits T-cell receptor (TCR) activation. This inhibition process requires PKA to localize to the immune synapse via binding to the membrane protein ezrin. We generated CAR T cells that expressed a small peptide called the "regulatory subunit I anchoring disruptor" (RIAD) that inhibits the association of PKA with ezrin, thus blunting the negative effects of PKA on TCR activation. After exposure to PGE2 or adenosine in vitro, CAR-RIAD T cells showed increased TCR signaling, released more cytokines, and showed enhanced killing of tumor cells compared with CAR T cells. When injected into tumor-bearing mice, the antitumor efficacy of murine and human CAR-RIAD T cells was enhanced compared with that of CAR T cells, due to resistance to tumor-induced hypofunction and increased T-cell infiltration of established tumors. Subsequent in vitro assays showed that both mouse and human CAR-RIAD cells migrated more efficiently than CAR cells did in response to the chemokine CXCL10 and also had better adhesion to various matrices. Thus, the intracellular addition of the RIAD peptide to adoptively transferred CAR T cells augments their efficacy by increasing their effector function and by improving trafficking into tumor sites. This treatment strategy, therefore, shows potential clinical application for treating solid tumors. Cancer Immunol Res; 4(6); 541-51. ©2016 AACR.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Immunotherapy, Adoptive/methods , Mesothelioma/therapy , Receptors, Antigen, T-Cell/immunology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytotoxicity, Immunologic , Female , Humans , Interferon-gamma/biosynthesis , Lymphocytes, Tumor-Infiltrating/immunology , Mesothelioma/enzymology , Mesothelioma/immunology , Mice, Inbred C57BL , Neoplasm Transplantation , Signal Transduction/immunology , Tumor Cells, Cultured
5.
Cancer Res ; 76(6): 1578-90, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26979791

ABSTRACT

Chimeric antigen receptor (CAR)-modified adoptive T-cell therapy has been successfully applied to the treatment of hematologic malignancies, but faces many challenges in solid tumors. One major obstacle is the immune-suppressive effects induced in both naturally occurring and genetically modified tumor-infiltrating lymphocytes (TIL) by inhibitory receptors (IR), namely PD1. We hypothesized that interfering with PD1 signaling would augment CAR T-cell activity against solid tumors. To address this possibility, we introduced a genetically engineered switch receptor construct, comprising the truncated extracellular domain of PD1 and the transmembrane and cytoplasmic signaling domains of CD28, into CAR T cells. We tested the effect of this supplement, "PD1CD28," on human CAR T cells targeting aggressive models of human solid tumors expressing relevant tumor antigens. Treatment of mice bearing large, established solid tumors with PD1CD28 CAR T cells led to significant regression in tumor volume due to enhanced CAR TIL infiltrate, decreased susceptibility to tumor-induced hypofunction, and attenuation of IR expression compared with treatments with CAR T cells alone or PD1 antibodies. Taken together, our findings suggest that the application of PD1CD28 to boost CAR T-cell activity is efficacious against solid tumors via a variety of mechanisms, prompting clinical investigation of this potentially promising treatment modality.


Subject(s)
Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/immunology , CD28 Antigens/immunology , Cell Line, Tumor , Female , Humans , Immunotherapy, Adoptive/methods , K562 Cells , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Tumor Burden/immunology , Xenograft Model Antitumor Assays/methods
6.
Clin Cancer Res ; 22(2): 436-47, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26324743

ABSTRACT

PURPOSE: Tumor-infiltrating lymphocytes (TILs) become hypofunctional, although the mechanisms are not clear. Our goal was to generate a model of human tumor-induced TIL hypofunction to study mechanisms and to test anti-human therapeutics. EXPERIMENTAL DESIGN: We transduced human T cells with a published, optimized T-cell receptor (TCR) that is directed to a peptide within the cancer testis antigen, NY-ESO-1. After demonstrating antigen-specific in vitro activity, these cells were used to target a human lung cancer line that expressed NY-ESO-1 in the appropriate HLA context growing in immunodeficient mice. The ability of anti-PD1 antibody to augment efficacy was tested. RESULTS: Injection of transgenic T cells had some antitumor activity, but did not eliminate the tumors. The injected T cells became profoundly hypofunctional accompanied by upregulation of PD1, Tim3, and Lag3 with coexpression of multiple inhibitory receptors in a high percentage of cells. This model allowed us to test reagents targeted specifically to human T cells. We found that injections of an anti-PD1 antibody in combination with T cells led to decreased TIL hypofunction and augmented the efficacy of the adoptively transferred T cells. CONCLUSIONS: This model offers a platform for preclinical testing of adjuvant immunotherapeutics targeted to human T cells prior to transition to the bedside. Because the model employs engineering of human T cells with a TCR clone instead of a CAR, it allows for study of the biology of tumor-reactive TILs that signal through an endogenous TCR. The lessons learned from TCR-engineered TILs can thus be applied to tumor-reactive TILs.


Subject(s)
Antibodies/immunology , Antigens, Neoplasm/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Membrane Proteins/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Adoptive Transfer/methods , Animals , Cell Line, Tumor , Cells, Cultured , HLA Antigens/immunology , Humans , Immunotherapy, Adoptive/methods , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Receptors, Antigen, T-Cell/immunology
7.
Int J Cancer ; 138(10): 2477-86, 2016 May 15.
Article in English | MEDLINE | ID: mdl-26704363

ABSTRACT

Histone/protein deacetylases (HDACs) are frequently upregulated in human malignancies and have therefore become therapeutic targets in cancer therapy. However, inhibiting certain HDAC isoforms can have protolerogenic effects on the immune system, which could make it easier for tumor cells to evade the host immune system. Therefore, a better understanding of how each HDAC isoform affects immune biology is needed to develop targeted cancer therapy. Here, we studied the immune phenotype of HDAC5(-/-) mice on a C57BL/6 background. While HDAC5(-/-) mice replicate at expected Mendelian ratios and do not develop overt autoimmune disease, their T-regulatory (Treg) cells show reduced suppressive function in vitro and in vivo. Likewise, CD4(+) T-cells lacking HDAC5 convert poorly to Tregs under appropriately polarizing conditions. To test if this attenuated Treg formation and suppressive function translated into improved anticancer immunity, we inoculated HDAC5(-/-) mice and littermate controls with a lung adenocarcinoma cell line. Cumulatively, lack of HDAC5 did not lead to better anticancer immunity. We found that CD8(+) T cells missing HDAC5 had a reduced ability to produce the cytokine, IFN-γ, in vitro and in vivo, which may offset the benefit of weakened Treg function and formation. Taken together, targeting HDAC5 weakens suppressive function and de-novo induction of Tregs, but also reduces the ability of CD8(+) T cells to produce IFN-γ.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Line, Tumor , Cytokines/biosynthesis , Forkhead Transcription Factors/metabolism , Gene Deletion , Gene Targeting , Interferon-gamma/biosynthesis , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
8.
Cancer Immunol Res ; 3(7): 815-26, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25941351

ABSTRACT

Chimeric antigen receptors (CAR) bearing an antigen-binding domain linked in cis to the cytoplasmic domains of CD3ζ and costimulatory receptors have provided a potent method for engineering T-cell cytotoxicity toward B-cell leukemia and lymphoma. However, resistance to immunotherapy due to loss of T-cell effector function remains a significant barrier, especially in solid malignancies. We describe an alternative chimeric immunoreceptor design in which we have fused a single-chain variable fragment for antigen recognition to the transmembrane and cytoplasmic domains of KIR2DS2, a stimulatory killer immunoglobulin-like receptor (KIR). We show that this simple, KIR-based CAR (KIR-CAR) triggers robust antigen-specific proliferation and effector function in vitro when introduced into human T cells with DAP12, an immunotyrosine-based activation motifs-containing adaptor. T cells modified to express a KIR-CAR and DAP12 exhibit superior antitumor activity compared with standard first- and second-generation CD3ζ-based CARs in a xenograft model of mesothelioma highly resistant to immunotherapy. The enhanced antitumor activity is associated with improved retention of chimeric immunoreceptor expression and improved effector function of isolated tumor-infiltrating lymphocytes. These results support the exploration of KIR-CARs for adoptive T-cell immunotherapy, particularly in immunotherapy-resistant solid tumors.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , B-Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Membrane Proteins/genetics , Mesothelioma/immunology , Receptors, Antigen, T-Cell/immunology , Single-Chain Antibodies/immunology , Animals , Cell Line, Tumor , Humans , Immunotherapy, Adoptive , Mice , Mice, SCID , Xenograft Model Antitumor Assays
9.
PLoS One ; 10(5): e0127310, 2015.
Article in English | MEDLINE | ID: mdl-26011724

ABSTRACT

Dysregulation of signaling pathways and energy metabolism in cancer cells enhances production of mitochondrial hydrogen peroxide that supports tumorigenesis through multiple mechanisms. To counteract the adverse effects of mitochondrial peroxide many solid tumor types up-regulate the mitochondrial thioredoxin reductase 2--thioredoxin 2 (TRX2)--peroxiredoxin 3 (PRX3) antioxidant network. Using malignant mesothelioma cells as a model, we show that thiostrepton (TS) irreversibly disables PRX3 via covalent crosslinking of peroxidatic and resolving cysteine residues in homodimers, and that targeting the oxidoreductase TRX2 with the triphenylmethane gentian violet (GV) potentiates adduction by increasing levels of disulfide-bonded PRX3 dimers. Due to the fact that activity of the PRX3 catalytic cycle dictates the rate of adduction by TS, immortalized and primary human mesothelial cells are significantly less sensitive to both compounds. Moreover, stable knockdown of PRX3 reduces mesothelioma cell proliferation and sensitivity to TS. Expression of catalase in shPRX3 mesothelioma cells restores defects in cell proliferation but not sensitivity to TS. In a SCID mouse xenograft model of human mesothelioma, administration of TS and GV together reduced tumor burden more effectively than either agent alone. Because increased production of mitochondrial hydrogen peroxide is a common phenotype of malignant cells, and TS and GV are well tolerated in mammals, we propose that targeting PRX3 is a feasible redox-dependent strategy for managing mesothelioma and other intractable human malignancies.


Subject(s)
Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mesothelioma/drug therapy , Mesothelioma/metabolism , Mitochondria/metabolism , Peroxides/metabolism , Peroxiredoxin III/metabolism , Thiostrepton/pharmacology , Animals , Catalase/metabolism , Cell Proliferation/drug effects , Epithelium/drug effects , Epithelium/metabolism , Humans , Male , Mesothelioma, Malignant , Mice , Mice, SCID , Mitochondria/drug effects , Oxidation-Reduction/drug effects , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Thioredoxins/metabolism
10.
Cancer Res ; 75(14): 2800-2810, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25979873

ABSTRACT

Malignant cells drive the generation of a desmoplastic and immunosuppressive tumor microenvironment. Cancer-associated stromal cells (CASC) are a heterogeneous population that provides both negative and positive signals for tumor cell growth and metastasis. Fibroblast activation protein (FAP) is a marker of a major subset of CASCs in virtually all carcinomas. Clinically, FAP expression serves as an independent negative prognostic factor for multiple types of human malignancies. Prior studies established that depletion of FAP(+) cells inhibits tumor growth by augmenting antitumor immunity. However, the potential for immune-independent effects on tumor growth have not been defined. Herein, we demonstrate that FAP(+) CASCs are required for maintenance of the provisional tumor stroma because depletion of these cells, by adoptive transfer of FAP-targeted chimeric antigen receptor (CAR) T cells, reduced extracellular matrix proteins and glycosaminoglycans. Adoptive transfer of FAP-CAR T cells also decreased tumor vascular density and restrained growth of desmoplastic human lung cancer xenografts and syngeneic murine pancreatic cancers in an immune-independent fashion. Adoptive transfer of FAP-CAR T cells also restrained autochthonous pancreatic cancer growth. These data distinguish the function of FAP(+) CASCs from other CASC subsets and provide support for further development of FAP(+) stromal cell-targeted therapies for the treatment of solid tumors.


Subject(s)
Extracellular Matrix/pathology , Gelatinases/metabolism , Membrane Proteins/metabolism , Neoplasms/pathology , Serine Endopeptidases/metabolism , Stromal Cells/physiology , Tumor Microenvironment/physiology , Animals , Endopeptidases , Epithelial-Mesenchymal Transition/genetics , Gelatinases/genetics , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasms/immunology , Serine Endopeptidases/genetics , Stromal Cells/metabolism , Tumor Cells, Cultured
11.
Oncoimmunology ; 2(8): e26218, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-24179709

ABSTRACT

We evaluated a neutralizing anti-TGFß antibody (GC1008) in cancer patients with malignant pleura mesothelioma (MPM). The goal of this study was to assess immunoregulatory effects in relation to clinical safety and clinical response. Patients with progressive MPM and 1-2 prior systemic therapies received GC1008 at 3mg/kg IV over 90 min every 21 d as part of an open-label, two-center Phase II trial. Following TGFß blockade therapy, clinical safety and patient survival were monitored along with the effects of anti-TGFß antibodies on serum biomarkers and peripheral blood mononuclear cells (PBMC). Although designed as a larger trial, only 13 patients were enrolled when the manufacturer discontinued further development of the antibody for oncology indications. All participants tolerated therapy. Although partial or complete radiographic responses were not observed, three patients showed stable disease at 3 mo. GC1008 had no effect in the expression of NK, CD4+, or CD8+ T cell activating and inhibitory markers, other than a decrease in the expression of 2B4 and DNAM-1 on NK cells. However, serum from 5 patients showed new or enhanced levels of antibodies against MPM tumor lysates as measured by immunoblotting. Patients who produced anti-tumor antibodies had increased median overall survival (OS) (15 vs 7.5 mo, p < 0.03) compared with those who did not. To our knowledge, these data represent the first immune analysis of TGFß- blockade in human cancer patients.

12.
J Cell Physiol ; 228(4): 835-45, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23018647

ABSTRACT

Malignant mesothelioma (MM) is an intractable tumor of the peritoneal and pleural cavities primarily linked to exposure to asbestos. Recently, we described an interplay between mitochondrial-derived oxidants and expression of FOXM1, a redox-responsive transcription factor that has emerged as a promising therapeutic target in solid malignancies. Here we have investigated the effects of nitroxides targeted to mitochondria via triphenylphosphonium (TPP) moieties on mitochondrial oxidant production, expression of FOXM1 and peroxiredoxin 3 (PRX3), and cell viability in MM cells in culture. Both Mito-carboxy-proxyl (MCP) and Mito-TEMPOL (MT) caused dose-dependent increases in mitochondrial oxidant production that was accompanied by inhibition of expression of FOXM1 and PRX3 and loss of cell viability. At equivalent concentrations TPP, CP, and TEMPOL had no effect on these endpoints. Live cell ratiometric imaging with a redox-responsive green fluorescent protein targeted to mitochondria (mito-roGFP) showed that MCP and MT, but not CP, TEMPOL, or TPP, rapidly induced mitochondrial fragmentation and swelling, morphological transitions that were associated with diminished ATP levels and increased production of mitochondrial oxidants. Mdivi-1, an inhibitor of mitochondrial fission, did not rescue mitochondria from fragmentation by MCP. Immunofluorescence microscopy experiments indicate a fraction of FOXM1 coexists in the cytoplasm with mitochondrial PRX3. Our results indicate that MCP and MT inhibit FOXM1 expression and MM tumor cell viability via perturbations in redox homeostasis caused by marked disruption of mitochondrial architecture, and suggest that both compounds, either alone or in combination with thiostrepton or other agents, may provide credible therapeutic options for the management of MM.


Subject(s)
Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/biosynthesis , Mesothelioma/metabolism , Mitochondria/metabolism , Oxidants/metabolism , Peroxiredoxin III/antagonists & inhibitors , Peroxiredoxin III/biosynthesis , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cytoplasm/drug effects , Cytoplasm/genetics , Cytoplasm/metabolism , Cytoplasm/physiology , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeostasis/drug effects , Homeostasis/genetics , Homeostasis/physiology , Humans , Mesothelioma/pathology , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Mitochondrial Dynamics/genetics , Mitochondrial Dynamics/physiology , Organophosphorus Compounds/pharmacology , Oxidation-Reduction/drug effects , Peroxiredoxin III/genetics , Peroxiredoxin III/metabolism , Quinazolinones/pharmacology
13.
PLoS One ; 7(6): e39404, 2012.
Article in English | MEDLINE | ID: mdl-22761781

ABSTRACT

Thiostrepton (TS) is a thiazole antibiotic that inhibits expression of FOXM1, an oncogenic transcription factor required for cell cycle progression and resistance to oncogene-induced oxidative stress. The mechanism of action of TS is unclear and strategies that enhance TS activity will improve its therapeutic potential. Analysis of human tumor specimens showed FOXM1 is broadly expressed in malignant mesothelioma (MM), an intractable tumor associated with asbestos exposure. The mechanism of action of TS was investigated in a cell culture model of human MM. As for other tumor cell types, TS inhibited expression of FOXM1 in MM cells in a dose-dependent manner. Suppression of FOXM1 expression and coincidental activation of ERK1/2 by TS were abrogated by pre-incubation of cells with the antioxidant N-acetyl-L-cysteine (NAC), indicating its mechanism of action in MM cells is redox-dependent. Examination of the mitochondrial thioredoxin reductase 2 (TR2)-thioredoxin 2 (TRX2)-peroxiredoxin 3 (PRX3) antioxidant network revealed that TS modifies the electrophoretic mobility of PRX3. Incubation of recombinant human PRX3 with TS in vitro also resulted in PRX3 with altered electrophoretic mobility. The cellular and recombinant species of modified PRX3 were resistant to dithiothreitol and SDS and suppressed by NAC, indicating that TS covalently adducts cysteine residues in PRX3. Reduction of endogenous mitochondrial TRX2 levels by the cationic triphenylmethane gentian violet (GV) promoted modification of PRX3 by TS and significantly enhanced its cytotoxic activity. Our results indicate TS covalently adducts PRX3, thereby disabling a major mitochondrial antioxidant network that counters chronic mitochondrial oxidative stress. Redox-active compounds like GV that modify the TR2/TRX2 network may significantly enhance the efficacy of TS, thereby providing a combinatorial approach for exploiting redox-dependent perturbations in mitochondrial function as a therapeutic approach in mesothelioma.


Subject(s)
Anti-Bacterial Agents/pharmacology , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Peroxiredoxin III/metabolism , Thiostrepton/pharmacology , Dose-Response Relationship, Drug , Forkhead Box Protein M1 , Forkhead Transcription Factors/metabolism , Humans , Mesothelioma, Malignant , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction , Signal Transduction/drug effects , Superoxides/metabolism , Tumor Cells, Cultured
14.
DNA Repair (Amst) ; 9(2): 134-43, 2010 Feb 04.
Article in English | MEDLINE | ID: mdl-20005182

ABSTRACT

Although DNA in eukaryotes is packaged in nucleosomes, it remains vulnerable to oxidative damage that can result from normal cellular metabolism, ionizing radiation, and various chemical agents. Oxidatively damaged DNA is repaired in a stepwise fashion via the base excision repair (BER) pathway, which begins with the excision of damaged bases by DNA glycosylases. We reported recently that the human DNA glycosylase hNTH1 (human Endonuclease III), a member of the HhH GpG superfamily of glycosylases, can excise thymine glycol lesions from nucleosomes without requiring or inducing nucleosome disruption; optimally oriented lesions are excised with an efficiency approaching that seen for naked DNA [1]. To determine if this property is shared by human DNA glycoylases in the Fpg/Nei family, we investigated the activity of NEIL1 on defined nucleosome substrates. We report here that the cellular concentrations and apparent k(cat)/K(M) ratios for hNTH1 and NEIL1 are similar. Additionally, after adjustment for non-specific DNA binding, hNTH1 and NEIL1 proved to have similar intrinsic activities toward nucleosome substrates. However, NEIL1 and hNTH1 differ in that NEIL1 binds undamaged DNA far more avidly than hNTH1. As a result, hNTH1 is able to excise both accessible and sterically occluded lesions from nucleosomes at physiological concentrations, while the high non-specific DNA affinity of NEIL1 would likely hinder its ability to process sterically occluded lesions in cells. These results suggest that, in vivo, NEIL1 functions either at nucleosome-free regions (such as those near replication forks) or with cofactors that limit its non-specific binding to DNA.


Subject(s)
Chromatin/metabolism , DNA Damage , DNA Glycosylases/metabolism , DNA/metabolism , Cell Line, Tumor , Deoxyribonuclease (Pyrimidine Dimer)/metabolism , Humans , Kinetics , Nucleic Acid Conformation , Nucleosomes/chemistry , Nucleosomes/metabolism , Oxidation-Reduction , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...