Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Med Chem ; 65(21): 14589-14598, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36318612

ABSTRACT

VSA-2 is a recently developed semisynthetic saponin immunostimulant. It is prepared by incorporating a terminal-functionalized side chain to the branched trisaccharide domain at the C3 position of Momordica saponin II (MS II) isolated from the seeds of perennial Momordica cochinchinensis Spreng. Direct comparison of VSA-2 and the clinically proven saponin adjuvant QS-21 shows that VSA-2 is comparable to QS-21 in enhancing humoral and cellular immune responses. Structure-activity relationship studies show that structural changes in the side chain have a significant impact on saponins' adjuvant activity. However, with the VSA-2 molecular framework intact, the new VSA-2 analogues with various substitution(s) at the terminal benzyl group of the side chain retain the ability of potentiating antigen-specific humoral and cellular responses.


Subject(s)
Momordica , Saponins , Momordica/chemistry , Adjuvants, Vaccine , Saponins/chemistry , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemistry , Structure-Activity Relationship
2.
Crit Rev Immunol ; 42(2): 35-47, 2022.
Article in English | MEDLINE | ID: mdl-37017287

ABSTRACT

Follicular regulatory T (TFR) cells are a population of CD4+ T-cells that concomitantly express markers for regulatory T-cells and follicular helper T (TFH) cells, and have been predominantly implicated in the regulation of humoral immunity via their suppressive functions. Rapid and robust progress has been made in the field of TFR cell research since the discovery of this subset over a decade ago. However, there is still a significant gap in our understanding of the mechanisms underlying the phenotypic and functional heterogeneity of TFR cells under various physiologic and pathologic settings. In this review article, we aim to highlight the most up-to-date concepts and investigations in both experimental animal models and human studies to provide a perspective on our understanding of TFR biology with particular emphasis on these cells in the context of disease settings.


Subject(s)
T-Lymphocytes, Helper-Inducer , T-Lymphocytes, Regulatory , Animals , Humans , CD4-Positive T-Lymphocytes , Immunity, Humoral , Biology
3.
Mol Cancer ; 20(1): 150, 2021 11 20.
Article in English | MEDLINE | ID: mdl-34798898

ABSTRACT

BACKGROUND: Accumulation of Foxp3+ regulatory T (Treg) cells in the tumor often represents an important mechanism for cancer immune evasion and a critical barrier to anti-tumor immunity and immunotherapy. Many tumor-infiltrating Treg cells display an activated phenotype and express the transcription factor Blimp1. However, the specific impact of these Blimp1+ Treg cells and their follicular regulatory T (TFR) cell subset on tumor and the underlying mechanisms of action are not yet well-explored. METHODS: Various transplantable tumor models were established in immunocompetent wild-type mice and mice with a Foxp3-specific ablation of Blimp1. Tumor specimens from patients with metastatic melanoma and TCGA datasets were analyzed to support the potential role of Treg and TFR cells in tumor immunity. In vitro culture assays and in vivo adoptive transfer assays were used to understand how Treg, TFR cells and antibody responses influence tumor control. RNA sequencing and NanoString analysis were performed to reveal the transcriptome of tumor-infiltrating Treg cells and tumor cells, respectively. Finally, the therapeutic effects of anti-PD-1 treatment combined with the disruption of Blimp1+ Treg activity were evaluated. RESULTS: Blimp1+ Treg and TFR cells were enriched in the tumors, and higher tumoral TFR signatures indicated increased risk of melanoma metastasis. Deletion of Blimp1 in Treg cells resulted in impaired suppressive activity and a reprogramming into effector T-cells, which were largely restricted to the tumor-infiltrating Treg population. This destabilization combined with increased anti-tumor effector cellular responses, follicular helper T-cell expansion, enhanced tumoral IgE deposition and activation of macrophages secondary to dysregulated TFR cells, remodeled the tumor microenvironment and delayed tumor growth. The increased tumor immunogenicity with MHC upregulation improved response to anti-PD-1 blockade. Mechanistically, Blimp1 enforced intratumoral Treg cells with a unique transcriptional program dependent on Eomesodermin (Eomes) expression; deletion of Eomes in Blimp1-deficient Treg cells restored tumor growth and attenuated anti-tumor immunity. CONCLUSIONS: These findings revealed Blimp1 as a new critical regulator of tumor-infiltrating Treg cells and a potential target for modulating Treg activity to treat cancer. Our study has also revealed two FCERIA-containing immune signatures as promising diagnostic or prognostic markers for melanoma patients.


Subject(s)
Neoplasms/etiology , Neoplasms/metabolism , Positive Regulatory Domain I-Binding Factor 1/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Disease Models, Animal , Fluorescent Antibody Technique , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunity, Humoral/genetics , Immunomodulation/drug effects , Immunophenotyping , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Macrophages/immunology , Macrophages/metabolism , Melanoma, Experimental , Mice , Mice, Knockout , Neoplasms/drug therapy , Neoplasms/pathology , Positive Regulatory Domain I-Binding Factor 1/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , Transcriptome , Treatment Outcome , Tumor Microenvironment/drug effects
4.
Nat Cancer ; 2(10): 1018-1038, 2021 10.
Article in English | MEDLINE | ID: mdl-35121884

ABSTRACT

Expanding the utility of immune-based cancer treatments is a clinical challenge due to tumor-intrinsic factors that suppress the immune response. Here we report the identification of tumoral ring finger protein 2 (RNF2), the core subunit of polycomb repressor complex 1, as a negative regulator of antitumor immunity in various human cancers, including breast cancer. In syngeneic murine models of triple-negative breast cancer, we found that deleting genes encoding the polycomb repressor complex 1 subunits Rnf2, BMI1 proto-oncogene, polycomb ring finger (Bmi1), or the downstream effector of Rnf2, remodeling and spacing factor 1 (Rsf1), was sufficient by itself to induce durable tumor rejection and establish immune memory by enhancing infiltration and activation of natural killer and CD4+ T cells, but not CD8+ T cells, into the tumor and enabled their cooperativity. These findings uncover an epigenetic reprogramming of the tumor-immune microenvironment, which fosters durable antitumor immunity and memory.


Subject(s)
Neoplasms , Polycomb Repressive Complex 1/metabolism , Tumor Microenvironment , Ubiquitin-Protein Ligases/metabolism , Animals , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Humans , Mice , Neoplasms/genetics , Nuclear Proteins , Polycomb Repressive Complex 1/genetics , Polycomb-Group Proteins , Trans-Activators , Tumor Microenvironment/genetics
5.
Nat Commun ; 11(1): 4660, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32938908

ABSTRACT

Intratumor spatial heterogeneity facilitates therapeutic resistance in glioblastoma (GBM). Nonetheless, understanding of GBM heterogeneity is largely limited to the surgically resectable tumor core lesion while the seeds for recurrence reside in the unresectable tumor edge. In this study, stratification of GBM to core and edge demonstrates clinically relevant surgical sequelae. We establish regionally derived models of GBM edge and core that retain their spatial identity in a cell autonomous manner. Upon xenotransplantation, edge-derived cells show a higher capacity for infiltrative growth, while core cells demonstrate core lesions with greater therapy resistance. Investigation of intercellular signaling between these two tumor populations uncovers the paracrine crosstalk from tumor core that promotes malignancy and therapy resistance of edge cells. These phenotypic alterations are initiated by HDAC1 in GBM core cells which subsequently affect edge cells by secreting the soluble form of CD109 protein. Our data reveal the role of intracellular communication between regionally different populations of GBM cells in tumor recurrence.


Subject(s)
Antigens, CD/metabolism , Brain Neoplasms/pathology , Glioblastoma/pathology , Histone Deacetylase 1/metabolism , Neoplasm Proteins/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Female , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/mortality , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Humans , Mice, SCID , Phenylbutyrates/pharmacology , Signal Transduction , Tumor Microenvironment , Xenograft Model Antitumor Assays
6.
Exp Cell Res ; 347(2): 293-300, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27521225

ABSTRACT

Tumor infiltrating monocytes play a crucial role in tumor immune surveillance. As lactate is an important component of the tumor milieu, we investigated its role in the transcriptional regulation of MHC I which is crucial for mounting effective immune responses against tumors. Lactate elevated MHC class I expression in monocytes. Increase in HLAB expression was concomitant with increase in HIF-1α and decrease in PRMT1 levels. Interestingly, a reciprocal relationship was observed between PRMT1 and HIF-1α. While HIF-1α inhibition decreased lactate induced MHC I, both pharmacological inhibition and siRNA mediated knockdown of PRMT1 upregulated HLAB levels. PRMT1 over-expression rescued lactate mediated increase in MHC I expression. Lactate mediated changes in nucleosomal occupancy on HLAB promoter facilitated a chromatin landscape that favoured decreased recruitment of CREB and PRMT1 on CRE site of HLAB locus. The effect of lactate on the chromatin landscape of HLAB was completely mimicked by PRMT1 inhibitor AMI-1 in terms of nucleosomal occupancy and CREB recruitment. Besides demonstrating the importance of lactate in the transcriptional regulation of HLAB, this study highlights for the first time the (i) existence of HIF-1α-PRMT1 regulatory loop and (ii) role of PRMT1 in modulating chromatin landscape crucial for facilitating HLAB gene expression.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lactic Acid/pharmacology , Monocytes/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation/drug effects , Histocompatibility Antigens Class I/genetics , Humans , Models, Biological , Monocytes/drug effects , Nucleosomes/drug effects , Nucleosomes/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors
7.
Exp Cell Res ; 340(1): 116-24, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26597758

ABSTRACT

Hypoxia-inducible Factor-1α (HIF-1α)-regulated expression of Hexokinase-II (HKII) remains a cornerstone in the maintenance of high metabolic demands subserving various pro-tumor functions including immune evasion in gliomas. Since inflammation-induced HIF-1α regulates Major Histocompatibility Complex class I (MHC-I) gene expression, and as cytoskeletal dynamics affect MHC-I membrane clusters, we investigated the involvement of HIF-1α-HKII axis in Tumor Necrosis Factor-α (TNFα)-mediated MHC-I membrane cluster stability in glioma cells and the involvement of actin cytoskeleton in the process. TNFα increased the clustering and colocalization of MHC-I with cortical actin in a HIF-1α dependent manner. siRNA mediated knockdown of HIF-1α as well as enzymatic inhibition of HK II by Lonidamine, delocalized mitochondrially bound HKII. This altered subcellular HKII localization affected TNFα-induced cofilin activation and actin turnover, as pharmacological inhibition of HKII by Lonidamine decreased Actin-related protein 2 (ARP2)/cofilin interaction. Photobleaching studies revealed destabilization of TNFα- induced stable MHC-I membrane clusters in the presence of Lonidamine and ARP2 inhibitor CK666. This work highlights how TNFα triggers a previously unknown function of metabolic protein HKII to influence an immune related outcome. Our study establishes the importance of inflammation induced HIF-1α in integrating two crucial components- the metabolic and immune, through reorganization of cytoskeleton.


Subject(s)
Actin Cytoskeleton/metabolism , Hexokinase/metabolism , Histocompatibility Antigens Class I/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cell Line, Tumor , Humans , Protein Stability
8.
Mol Cell Biochem ; 399(1-2): 105-12, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25300619

ABSTRACT

Transforming growth factor (TGF-ß) is associated with the progression of glioblastoma multiforme (GBM)-the most malignant of brain tumors. Since there is a structural homology between TGF-ß and human chorionic gonadotropin (hCG) and as both TGF-ß and hCG-ß are known regulators of oxidative stress and survival responses in a variety of tumors, the role of TGF-ß in the regulation of hCG-ß and its consequences on redox modulation of glioblastoma cells was investigated. A heightened hCG-ß level was observed in GBM tumors. TGF-ß treatment increased hCG-ß expression in glioma cell lines, and this heightened hCG-ß was found to regulate redox homeostasis in TGF-ß-treated glioma cells, as siRNA-mediated knockdown of hCG-ß (i) elevated reactive oxygen species (ROS) generation, (ii) decreased thioredoxin Trx1 expression and thioredoxin reductase (TrxR) activity, and (iii) abrogated expression of TP53-induced glycolysis and apoptosis regulator (TIGAR). Silencing of hCG-ß abrogated Smad2/3 levels, suggesting the existence of TGF-ß-hCG-ß cross-talk in glioma cells. siRNA-mediated inhibition of elevated TIGAR levels in TGF-ß-treated glioma cells was accompanied by an increase in ROS levels. As a farnesyltransferase inhibitor, Manumycin is known to induce glioma cell apoptosis in a ROS-dependent manner, and we investigated whether Manumycin could induce apoptosis in TGF-ß-treated cells with elevated hCG-ß exhibiting ROS-scavenging property. Manumycin-induced apoptosis in TGF-ß-treated cells was accompanied by elevated ROS levels and decreased expression of hCG-ß, Trx1, Smad2/3, and TIGAR. These findings indicate the existence of a previously unknown TGF-ß-hCG-ß link that regulates redox homeostasis in glioma cells.


Subject(s)
Chorionic Gonadotropin, beta Subunit, Human/physiology , Glioblastoma/metabolism , Homeostasis , Transforming Growth Factor beta/physiology , Cell Line, Tumor , Cell Survival , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Oxidation-Reduction , Polyenes/pharmacology , Polyunsaturated Alkamides/pharmacology , Reactive Oxygen Species/metabolism
9.
Mol Cell Biol ; 33(14): 2718-31, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23671189

ABSTRACT

Hypoxia-inducible factor 1α (HIF-1α) plays a crucial role in the progression of glioblastoma multiforme tumors, which are characterized by their effective immune escape mechanisms. As major histocompatibility complex class I (MHC-I) is involved in glioma immune evasion and since HIF-1α is a pivotal link between inflammation and glioma progression, the role of tumor necrosis factor alpha (TNF-α)-induced inflammation in MHC-I gene regulation was investigated. A TNF-α-induced increase in MHC-I expression and transcriptional activation was concurrent with increased HIF-1α, ΝF-κΒ, and ß-catenin activities. While knockdown of HIF-1α and ß-catenin abrogated TNF-α-induced MHC-I activation, NF-κB had no effect. ß-Catenin inhibition abrogated HIF-1α activation and vice versa, and this HIF-1α-ß-catenin axis positively regulated CREB phosphorylation. Increased CREB activation was accompanied by its increased association with ß-catenin and CBP. Chromatin immunoprecipitation revealed increased CREB enrichment at CRE/site α on the MHC-I promoter in a ß-catenin-dependent manner. ß-Catenin replaced human Brahma (hBrm) with Brg1 as the binding partner for CREB at the CRE site. The hBrm-to-Brg1 switch is crucial for MHC-I regulation, as ATPase-deficient Brg1 abolished TNF-α-induced MHC-I expression. ß-Catenin also increased the association of MHC-I enhanceosome components RFX5 and NF-YB at the SXY module. CREB acts as a platform for assembling coactivators and chromatin remodelers required for MHC-I activation in a HIF-1α/ß-catenin-dependent manner.


Subject(s)
Chromatin Assembly and Disassembly , Genes, MHC Class I , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Transcriptional Activation , Tumor Necrosis Factor-alpha/physiology , beta Catenin/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Binding, Competitive , CCAAT-Binding Factor/metabolism , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Gene Expression , Humans , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Processing, Post-Translational , Regulatory Factor X Transcription Factors , Response Elements , Transcription Factor 4 , Transcription Factors/metabolism
10.
Cell Signal ; 25(3): 682-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23220408

ABSTRACT

The non-classical HLA class I antigen HLA-G contributes to immune escape mechanisms in glioblastoma multiforme (GBM). We have previously shown that IL-1ß-HIF-1α axis connects inflammatory and oncogenic pathways in GBM. In this study, we investigated the role of IL-1ß induced inflammation in regulating HLA-G expression. IL-1ß increased HLA-G and Toll like receptor 4 (TLR4) expression in a HIF-1α dependent manner. Inhibition of TLR4 signaling abrogated IL-1ß induced HLA-G. IL-1ß increased HMGB1 expression and its interaction with TLR4. Inhibition of HMGB1 inhibited TLR4 and vice versa suggesting the existence of HMGB1-TLR4 axis in glioma cells. Interestingly, HMGB1 inhibition prevented IL-1ß induced HLA-G expression. Elevated levels of HMGB1 and ß-defensin 3 were observed in GBM tumors. Importantly, ß-defensin-3 prevented IL-1ß induced HLA-G, TLR4, HMGB1 expression and release of pro-inflammatory mediators. Our studies indicate that ß-defensin-3 abrogates IL-1ß induced HLA-G expression by negatively affecting key molecules associated with its regulation.


Subject(s)
HLA-G Antigens/metabolism , HMGB1 Protein/metabolism , Interleukin-1beta/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , beta-Defensins/pharmacology , Cell Line, Tumor , Glioma/metabolism , Glioma/pathology , HMGB1 Protein/antagonists & inhibitors , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Inflammation/metabolism , Interleukin-1beta/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Toll-Like Receptor 4/antagonists & inhibitors , beta-Defensins/metabolism
11.
J Mol Med (Berl) ; 90(1): 67-80, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21887505

ABSTRACT

The precise role of different toll-like receptor (TLR) superfamily members is just beginning to get elucidated in glioblastoma multiforme (GBM). In this study, we observed heightened TLR4 levels in GBM tumor samples as compared to adjacent normal tissue. Since the pro-inflammatory cytokine tumor necrosis factor (TNF)α induces NF-κB activation in GBM, and as several common signaling mediators are involved in TNFα and TLR4-mediated NF-κB activation, we investigated the role of TLR4 in the regulation of NF-κB activation and inflammatory responses in TNFα-treated glioma cells. TNFα elevated TLR4 expression and inhibition of TLR4 signaling by either signaling inhibitor, neutralizing antibody, or small interfering RNA (siRNA)-attenuated TNFα-induced NF-κB activation. TLR4-mediated NF-κB activation was independent of canonical myeloid differentiation factor 88 signaling but involved toll/IL-1R homology domain-containing adaptor protein-inducing interferon-ß. Inhibition of TLR4 signaling abrogated TNFα-induced increase in (1) transcription factors interferon (IFN) regulatory factor 3 and STAT-1 and (2) IFNß and inflammatory cytokines/chemokines expression. Furthermore, TNFα-induced TLR4-dependent increase in AKT activation and HIF-1α transcriptional activation suggested the existence of TLR4-AKT-HIF-1α axis. Importantly, TNFα-induced TLR4 was abrogated in cells transfected with dominant negative IκB and HIF-1α siRNA. Our studies indicate that TNFα triggered TLR4-HIF-1α and NF-κB-TLR4 feed-forward loops act in tandem to sustain inflammatory response in glioma.


Subject(s)
Glioma/immunology , Glioma/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , NF-kappa B/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Cell Line, Tumor , Enzyme Activation/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , Glioma/pathology , Humans , Inflammation/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Myeloid Differentiation Factor 88/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT1 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/pharmacology
12.
Neurochem Int ; 59(5): 567-71, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21763744

ABSTRACT

Cancer stem-like cells (CSCs) possessing features of neural precursor cells (NPC) influence initiation, recurrence and chemoresistance of glioblastoma multiforme (GBM). As inflammation is crucial for glioblastoma progression we investigated the effect of chronic IL-1ß treatment on CSCs derived from glioblastoma cell line U87MG. Exposure to IL-1ß for 10 days increased (i) accumulation of 8-OHdG - a key biomarker of oxidative DNA damage; (ii) DNA damage response (DDR) indicators γH2AX, ATM and DNA-PK; (iii) nuclear and cytoplasmic p53 and COX-2 levels and (iv) interaction between COX-2 and p53. Despite upregulating p53 expression IL-1ß had no effect on cell cycle progression, apoptosis or self renewal capacity of CSCs. COX-2 inhibitor Celecoxib reduced self renewal capacity and increased apoptosis of both control and IL-1ß treated CSCs. Therefore the ability of COX-2 to regulate proliferation of CSCs irrespective of exposure to IL-1ß, warrants further investigation of COX-2 as a potential anti-glioma target.


Subject(s)
Cyclooxygenase 2/physiology , Glioma/pathology , Neoplastic Stem Cells/physiology , 8-Hydroxy-2'-Deoxyguanosine , Apoptosis/drug effects , Blotting, Western , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , DNA Damage , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/genetics , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Disease Progression , Flow Cytometry , Glioma/enzymology , Histones/metabolism , Humans , Interleukin-1beta/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Tumor Suppressor Protein p53/genetics
13.
BMC Cancer ; 10: 328, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20576128

ABSTRACT

BACKGROUND: The highly resistant nature of glioblastoma multiforme (GBM) to chemotherapy prompted us to evaluate the efficacy of bicyclic triterpenoid Iripallidal against GBM in vitro. METHODS: The effect of Iripallidal on proliferation and apoptosis in glioma cell lines was evaluated by MTS, colony formation and caspase-3 activity. The effect of iripallidal to regulate (i) Akt/mTOR and STAT3 signaling (ii) molecules associated with cell cycle and DNA damage was evaluated by Western blot analysis. The effect of Iripallidal on telomerase activity was also determined. RESULTS: Iripallidal (i) induced apoptosis, (ii) inhibited Akt/mTOR and STAT3 signaling, (iii) altered molecules associated with cell cycle and DNA damage, (iv) inhibited telomerase activity and colony forming efficiency of glioma cells. In addition, Iripallidal displayed anti-proliferative activity against non-glioma cancer cell lines of diverse origin. CONCLUSION: The ability of Iripallidal to serve as a dual-inhibitor of Akt/mTOR and STAT3 signaling warrants further investigation into its role as a therapeutic strategy against GBM.


Subject(s)
Acrolein/analogs & derivatives , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Cyclohexanols/pharmacology , Glioblastoma/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Acrolein/pharmacology , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Telomerase/antagonists & inhibitors , Telomerase/metabolism
14.
Neurochem Int ; 56(1): 194-201, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19836430

ABSTRACT

Inflammation which is an indispensable participant in tumor progression is intricately linked with redox modulation. The pro-inflammatory cytokine Tumor Necrosis Factor (TNFalpha) elevates reactive oxygen species (ROS) in glioblastoma multiforme (GBM). As both TNFalpha and oxidative stress independently play role in regulating cytoskeletal organization and cell survival pathways we investigated whether TNFalpha mediated oxidative stress regulates responses that offer survival advantages to glioblastoma cells. Treatment with TNFalpha elevated Akt phosphorylation in glioma cells. Increased in Akt phosphorylation was concurrent with the decrease in ROS scavenger SOD-1 levels. TNFalpha mediated increase in Akt phosphorylation was dependent on oxidative stress as Akt phosphorylation was abrogated in the presence of ROS inhibitor and elevated in cells transfected with SOD-1 siRNA. TNFalpha altered actin cytoskeletal organization and increased Cdc42 levels. This increase in Cdc42 was concomitant with its increased interaction with scaffold protein IQGAP-1. Also, we report for the first time a ROS dependent interaction between pAkt and IQGAP-1 in TNFalpha treated cells. Importantly, Akt inhibition not only reversed TNFalpha mediated changes in actin cytoskeletal organization but also abrogated anchorage independent growth. Together, these results suggest that TNFalpha induced oxidative stress affects Akt activation to regulate actin organization and growth of glioma cells.


Subject(s)
Actin Cytoskeleton/metabolism , Brain Neoplasms/metabolism , Glioma/metabolism , Oxidative Stress/physiology , Proto-Oncogene Proteins c-akt/metabolism , Tumor Necrosis Factor-alpha/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/physiopathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Cytoskeleton/pathology , Encephalitis/etiology , Encephalitis/metabolism , Encephalitis/physiopathology , Enzyme Inhibitors/pharmacology , Glioma/drug therapy , Glioma/physiopathology , Humans , Oxidative Stress/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/drug effects , RNA Interference , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Superoxide Dismutase/drug effects , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Tumor Necrosis Factor-alpha/pharmacology , cdc42 GTP-Binding Protein/drug effects , cdc42 GTP-Binding Protein/metabolism , ras GTPase-Activating Proteins/drug effects , ras GTPase-Activating Proteins/metabolism
15.
J Cell Mol Med ; 14(8): 2151-61, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19583803

ABSTRACT

The present study identified a novel mechanism of induction of apoptosis in glioblastoma cells by scriptaid - a histone deacetylase (HDAC) inhibitor. Scriptaid reduced glioma cell viability by increasing Jun N-terminal kinase (JNK) activation. Although scriptaid induced activation of both p38MAPK and JNK, it was the inhibition of JNK that attenuated scriptaid-induced apoptosis significantly. Scriptaid also increased the expression of (i) p21 and p27 involved in cell-cycle regulation and (ii) γH2AX associated with DNA damage response in a JNK-dependent manner. Treatment with scriptaid increased Ras activity in glioma cells, and transfection of cells with constitutively active RasV12 further sensitized glioma cells to scriptaid-induced apoptosis. Scriptaid also inhibited telomerase activity independent of JNK. Taken together, our findings indicate that scriptaid (i) induces apoptosis and reduces glioma cell proliferation by elevating JNK activation and (ii) also decreases telomerase activity in a JNK-independent manner.


Subject(s)
Apoptosis/drug effects , Hydroxylamines/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Quinolines/pharmacology , Telomerase/metabolism , Acetylation/drug effects , Anthracenes/pharmacology , Blotting, Western , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme-Linked Immunosorbent Assay , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histones/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Polymerase Chain Reaction/methods , Signal Transduction/drug effects , Telomerase/antagonists & inhibitors , Telomerase/genetics , ras Proteins/metabolism
16.
Free Radic Biol Med ; 47(4): 364-74, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19409983

ABSTRACT

The poor prognosis of glioblastoma multiforme and lack of effective therapy have necessitated the identification of new treatment strategies. We have previously reported that elevation of oxidative stress induces apoptosis of glioma cells. Because the farnesyltransferase inhibitor manumycin is known to induce reactive oxygen species (ROS) generation, we evaluated the effects of manumycin on glioma cells. Manumycin induced glioma cell apoptosis by elevating ROS generation. Treatment with the ROS inhibitor N-acetylcysteine blocked manumycin-induced apoptosis, caspase-3 activity, and PARP expression, indicating the involvement of increased ROS in the proapoptotic activity of manumycin. This heightened ROS level was accompanied by a concurrent decrease in antioxidants such as superoxide dismutase (SOD-1) and thioredoxin (TRX-1). SOD-1 overexpression protects glioma cells from manumycin-induced apoptosis. In addition, small interfering RNA-mediated knockdown of SOD-1 and TRX-1 expression also increased ROS generation and sensitivity of glioma cells to manumycin-induced cell death. Interestingly, suppressing ROS generation prevented manumycin-induced Ras inhibition. This study reports for the first time that Ras inhibition by manumycin is due to heightened ROS levels. We also report for the first time that manumycin inhibits the phosphorylation of signal transducer and activator of transcription 3 and telomerase activity in a ROS-dependent manner, which plays a crucial role in glioma resistance to apoptosis. In addition manumycin (i) induced the DNA-damage repair response, (ii) affected cell-cycle-regulatory molecules, and (iii) impaired the colony-forming ability of glioma cells in a ROS-dependent manner.


Subject(s)
Glioblastoma/pathology , Glioma/pathology , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Telomerase/metabolism , Acetylcysteine/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Farnesyltranstransferase/antagonists & inhibitors , Gene Expression Regulation/drug effects , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/physiopathology , Glioblastoma/therapy , Glioma/enzymology , Glioma/genetics , Glioma/physiopathology , Glioma/therapy , Humans , Oxidative Stress , Phosphorylation , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Polyenes/pharmacology , Polyenes/therapeutic use , Polyunsaturated Alkamides/pharmacology , Polyunsaturated Alkamides/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Small Interfering/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Thioredoxins/genetics , Thioredoxins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...