Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Am Nutr Assoc ; 43(3): 261-271, 2024.
Article in English | MEDLINE | ID: mdl-37905950

ABSTRACT

OBJECTIVE: Virgin coconut oil (VCNO), an unrefined kernel oil from Cocos nucifera L., has considerable medicinal and nutritive value. Experimental evidence suggests its antioxidant, anti-inflammatory, chemoprotective, analgesic, and hypolipidemic effects. Presently, the effect of VCNO on ameliorating dextran sodium sulfate (DSS)-induced inflammatory bowel disease and cyclophosphamide (CTX)-induced immunosuppression in experimental animals was analyzed. METHOD: DSS (4%) was administered to BALB/c mice through drinking water for 12 days to induce inflammatory bowel disease, and VCNO (500, 750, and 1000 mg/kg bwt) was supplemented orally for 12 days. For anti-inflammatory studies, lipopolysaccharide (LPS, 250 µg/animal) was injected into the intraperitoneal cavity of Swiss albino mice followed by 7 days' pretreatment of VCNO (500, 750, and 1000 mg/kg bwt). To understand the mechanism of action, serum from all animals was collected after 6 hours of LPS challenge and levels of proinflammatory cytokines were analyzed using enzyme-inked immunosorbent assay. In addition to this, immunosuppression was induced by CTX (50 mg/kg bwt, po) in Swiss albino mice. RESULTS: Oral administration of VCNO effectively reversed the pathologies associated with inflammatory bowel disease induced by DSS, including loss of body weight, increased disease activity index, shortening of colon length, diarrhea, and rectal bleeding. Histopathological examination showed that VCNO restored the damage in colon tissue induced by DSS. Similar trends were noticed in levels of myeloperoxidase and mRNA expression of proinflammatory cytokines in colon tissue. In addition to this, supplementation of VCNO markedly reduced the hike in the level of serum proinflammatory cytokines in LPS-challenged mice. Further, administration of VCNO effectively increased spleen and thymus indexes and stimulated the production of interferon-γ in serum. CONCLUSIONS: Overall, this study revealed that VCNO alleviates inflammatory bowel disease and inflammation; concurrently, it can revert immunosuppression.


Subject(s)
Inflammatory Bowel Diseases , Lipopolysaccharides , Animals , Mice , Coconut Oil , Dextran Sulfate/toxicity , Lipopolysaccharides/toxicity , Inflammation/drug therapy , Inflammatory Bowel Diseases/chemically induced , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Immunity
5.
J Oncol ; 2022: 3817724, 2022.
Article in English | MEDLINE | ID: mdl-36624801

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common primary liver cancer in patients with liver cirrhosis of various etiologies. In recent years, there has been an advance in the knowledge of molecular mechanisms and a better staging definition of patients which has allowed the development of new therapies that have entered the therapeutic workup of these patients. Deep information on molecular drivers of HCC contributed to the development of targeted therapies with remarkable benefits. The novel strategies of targeting immune evasion using immune checkpoint inhibitors and CAR-T and TCR-T therapeutics have also shown promising results. For advanced diseases, the therapeutic algorithm has been recently updated, thanks to the efficacy of combining immunotherapy and antiangiogenic therapy in the first-line setting, and new drugs, both as single-agents or combinations, are currently under investigation.

6.
IUBMB Life ; 73(11): 1348-1362, 2021 11.
Article in English | MEDLINE | ID: mdl-34514729

ABSTRACT

STAT3 is a key oncogenic transcription factor, often overactivated in several human cancers including hepatocellular carcinoma (HCC). STAT3 modulates the expression of genes that are connected with cell proliferation, antiapoptosis, metastasis, angiogenesis, and immune evasion in tumor cells. In this study, we investigated the effect of crocetin on the growth of HCC cells and dissected its underlying molecular mechanism in imparting a cytotoxic effect. Crocetin suppressed proliferation, promoted apoptosis, and counteracted the invasive capacity of HCC cells. Besides, crocetin downregulated the constitutive/inducible STAT3 activation (STAT3Y705 ), nuclear accumulation of STAT3 along with suppression of its DNA binding activity in HCC cells with no effect on STAT5 activation. Crocetin suppressed the activity of upstream kinases such as Src, JAK1, and JAK2. Sodium pervanadate treatment terminated the crocetin-propelled STAT3 inhibition suggesting the involvement of tyrosine phosphatases. Crocetin increased the expression of SHP-1 and siRNA-mediated SHP-1 silencing resulted in the negation of crocetin-driven STAT3 inhibition. Further investigation revealed that crocetin treatment inhibited the expression of STAT3 regulated genes (Bcl-2, Bcl-xL, cyclin D1, survivin, VEGF, COX-2, and MMP-9). Taken together, this report presents crocetin as a novel abrogator of the STAT3 pathway in HCC cell lines.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carotenoids/pharmacology , STAT3 Transcription Factor/metabolism , Vitamin A/analogs & derivatives , Caspase 3/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Interleukin-6/metabolism , Interleukin-6/pharmacology , Janus Kinase 2/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Vitamin A/pharmacology
7.
Oncogene ; 39(31): 5373-5389, 2020 07.
Article in English | MEDLINE | ID: mdl-32561852

ABSTRACT

Cancer stem cells possess the capacity for self-renewal and resistance to chemotherapy. It is therefore crucial to understand the molecular regulators of stemness in the quest to develop effective cancer therapies. TAZ is a transcription activator that promotes stem cell functions in post-development mammalian cells; suppression of TAZ activity reduces or eliminates cancer stemness in select cancers. Isoprenylcysteine carboxylmethyltransferase (ICMT) is the unique enzyme of the last step of posttranslational prenylation processing pathway that modifies several oncogenic proteins, including RAS. We found that suppression of ICMT results in reduced self-renewal/stemness in KRAS-driven pancreatic and breast cancer cells. Silencing of ICMT led to significant reduction of TAZ protein levels and loss of self-renewal ability, which could be reversed by overexpressing mutant KRAS, demonstrating the functional impact of ICMT modification on the ability of KRAS to control TAZ stability and function. Contrary to expectation, YAP protein levels appear to be much less susceptible than TAZ to the regulation by ICMT and KRAS, and YAP is less consequential in regulating stemness characteristics in these cells. Further, we found that the ICMT-dependent KRAS regulation of TAZ was mediated through RAF, but not PI3K, signaling. Functionally, we demonstrate that a signaling cascade from ICMT modification of KRAS to TAZ protein stability supports cancer cell self-renewal abilities in both in vitro and in vivo settings. In addition, studies using the proof-of-concept small molecule inhibitors of ICMT confirmed its role in regulating TAZ and self-renewal, demonstrating the potential utility of targeting ICMT to control aggressive KRAS-driven cancers.


Subject(s)
Breast Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/metabolism , Protein Methyltransferases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Trans-Activators/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Self Renewal/physiology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Doxorubicin/pharmacology , Female , HEK293 Cells , Heterografts , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Protein Methyltransferases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Trans-Activators/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Gemcitabine
8.
Cancers (Basel) ; 11(8)2019 Aug 03.
Article in English | MEDLINE | ID: mdl-31382612

ABSTRACT

Cancer cells possess metabolic properties that are different from benign cells. These unique characteristics have become attractive targets that are being actively investigated for cancer therapy. p21cip1/waf1, also known as Cyclin-Dependent Kinase inhibitor 1A, is encoded by the CDKN1A gene. It is a major p53 target gene involved in cell cycle progression that has been extensively evaluated. To date, p21 has been reported to regulate various cell functions, both dependent and independent of p53. Besides regulating the cell cycle, p21 also modulates apoptosis, induces senescence, and maintains cellular quiescence in response to various stimuli. p21 transcription is induced in response to stresses, including those from oxidative and chemotherapeutic treatment. A recent study has shown that in response to metabolic stresses such as nutrient and energy depletion, p21 expression is induced to regulate various cell functions. Despite the biological significance, the mechanism of p21 regulation in cancer adaptation to metabolic stress is underexplored and thus represents an exciting field. This review focuses on the recent development of p21 regulation in response to metabolic stress and its impact in inducing cell cycle arrest and death in cancer cells.

9.
Mol Cancer Ther ; 16(5): 914-923, 2017 05.
Article in English | MEDLINE | ID: mdl-28167504

ABSTRACT

Pancreatic cancer remains one of the most difficult to treat human cancers despite recent advances in targeted therapy. Inhibition of isoprenylcysteine carboxylmethyltransferase (ICMT), an enzyme that posttranslationally modifies a group of proteins including several small GTPases, suppresses proliferation of some human cancer cells. However, the efficacy of ICMT inhibition on human pancreatic cancer has not been evaluated. In this study, we have evaluated a panel of human pancreatic cancer cell lines and identified those that are sensitive to ICMT inhibition. In these cells, ICMT suppression inhibited proliferation and induced apoptosis. This responsiveness to ICMT inhibition was confirmed in in vivo xenograft tumor mouse models using both a small-molecule inhibitor and shRNA-targeting ICMT. Mechanistically, we found that, in sensitive pancreatic cancer cells, ICMT inhibition induced mitochondrial respiratory deficiency and cellular energy depletion, leading to significant upregulation of p21. Furthermore, we characterized the role of p21 as a regulator and coordinator of cell signaling that responds to cell energy depletion. Apoptosis, but not autophagy, that is induced via p21-activated BNIP3 expression accounts for the efficacy of ICMT inhibition in sensitive pancreatic cancer cells in both in vitro and in vivo models. In contrast, cells resistant to ICMT inhibition demonstrated no mitochondria dysfunction or p21 signaling changes under ICMT suppression. These findings not only identify pancreatic cancers as potential therapeutic targets for ICMT suppression but also provide an avenue for identifying those subtypes that would be most responsive to agents targeting this critical enzyme. Mol Cancer Ther; 16(5); 914-23. ©2017 AACR.


Subject(s)
Membrane Proteins/genetics , Pancreatic Neoplasms/drug therapy , Protein Methyltransferases/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , rho GTP-Binding Proteins/genetics , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Protein Methyltransferases/genetics , Signal Transduction/drug effects , Small Molecule Libraries/administration & dosage , Xenograft Model Antitumor Assays
11.
PLoS One ; 8(3): e57015, 2013.
Article in English | MEDLINE | ID: mdl-23472074

ABSTRACT

Accumulating evidence(s) indicate that CXCL12-CXCR4 signaling cascade plays an important role in the process of invasion and metastasis that accounts for more than 80% of deaths in hepatocellular carcinoma (HCC) patients. Thus, identification of novel agents that can downregulate CXCR4 expression and its associated functions have a great potential in the treatment of metastatic HCC. In the present report, we investigated an anthraquinone derivative, emodin for its ability to affect CXCR4 expression as well as function in HCC cells. We observed that emodin downregulated the expression of CXCR4 in a dose-and time-dependent manner in HCC cells. Treatment with pharmacological proteasome and lysosomal inhibitors did not have substantial effect on emodin-induced decrease in CXCR4 expression. When investigated for the molecular mechanism(s), it was observed that the suppression of CXCR4 expression was due to downregulation of mRNA expression, inhibition of NF-κB activation, and abrogation of chromatin immunoprecipitation activity. Inhibition of CXCR4 expression by emodin further correlated with the suppression of CXCL12-induced migration and invasion in HCC cell lines. In addition, emodin treatment significantly suppressed metastasis to the lungs in an orthotopic HCC mice model and CXCR4 expression in tumor tissues. Overall, our results show that emodin exerts its anti-metastatic effect through the downregulation of CXCR4 expression and thus has the potential for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Movement/drug effects , Emodin/pharmacology , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Receptors, CXCR4/metabolism , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Down-Regulation , Female , Genes, Reporter , Humans , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation , RNA, Messenger/metabolism , Signal Transduction , Wound Healing
12.
BMC Chem Biol ; 12: 5, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23033888

ABSTRACT

BACKGROUND: Dibenzoazepine (DB) derivatives are important and valuable compounds in medicinal chemistry. The synthesis and chemotherapeutic properties of naturally occurring DBs and different heterocyclic moiety tethered DBs are reported. Herein, we report the DB-fused hybrid structure that containing isoxazolines (DBIs) and their anti-cancer activity, which could throw light on the structural and functional features of new molecules. RESULTS AND CONCLUSION: The synthesis and characterization of novel ring DB tethered isoxazoline derivatives (DBIs) were carried out. After the detailed structural characterization using 2D-NMR experiments, the compounds were identified as 5-substituted isoxazolines. The effect of newly synthesized DBIs against the invasion of murine osteosarcoma (LM8G7) cells was studied. Among the tested molecules, compound 4g (5-[-3-(4-chlorophenyl)-4,5-dihydroisoxazol-5-yl-methyl]-5 H-dibenzo[b,f]azepine), was found to inhibit the invasion of LM8G7 cells strongly, when compared to other structurally related compounds. Cumulatively, the compound 4g inhibited the invasion MDA-MB-231 cells completely at 10 µM. In addition to anti-invasion property the compound 4g also inhibited the migration of LM8G7 and human ovarian cancer cells (OVSAHO) dose-dependently. Compound 4g inhibited the proliferation of LM8G7, OVSAHO, human breast cancer cells (MCF-7) and human melphalan-resistant multiple myeloma (RPMI8226-LR5) cells that are comparable to cisplatin and suramin.

13.
J Biol Chem ; 287(45): 38028-40, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-22992727

ABSTRACT

Gastric cancer (GC) is a lethal malignancy and the second most common cause of cancer-related deaths. Although treatment options such as chemotherapy, radiotherapy, and surgery have led to a decline in the mortality rate due to GC, chemoresistance remains as one of the major causes for poor prognosis and high recurrence rate. In this study, we investigated the potential effects of isorhamnetin (IH), a 3'-O-methylated metabolite of quercetin on the peroxisome proliferator-activated receptor γ (PPAR-γ) signaling cascade using proteomics technology platform, GC cell lines, and xenograft mice model. We observed that IH exerted a strong antiproliferative effect and increased cytotoxicity in combination with chemotherapeutic drugs. IH also inhibited the migratory/invasive properties of GC cells, which could be reversed in the presence of PPAR-γ inhibitor. We found that IH increased PPAR-γ activity and modulated the expression of PPAR-γ regulated genes in GC cells. Also, the increase in PPAR-γ activity was reversed in the presence of PPAR-γ-specific inhibitor and a mutated PPAR-γ dominant negative plasmid, supporting our hypothesis that IH can act as a ligand of PPAR-γ. Using molecular docking analysis, we demonstrate that IH formed interactions with seven polar residues and six nonpolar residues within the ligand-binding pocket of PPAR-γ that are reported to be critical for its activity and could competitively bind to PPAR-γ. IH significantly increased the expression of PPAR-γ in tumor tissues obtained from xenograft model of GC. Overall, our findings clearly indicate that antitumor effects of IH may be mediated through modulation of the PPAR-γ activation pathway in GC.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , PPAR gamma/metabolism , Quercetin/analogs & derivatives , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Anilides/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Drug Synergism , Female , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , PPAR gamma/antagonists & inhibitors , Protein Binding/drug effects , Proteomics , Quercetin/metabolism , Quercetin/pharmacology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays
14.
Pharm Biol ; 49(10): 995-1007, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21936626

ABSTRACT

CONTEXT: Cancer is a major public health problem in India and many other parts of the world. Its two main characteristics are uncontrolled cell growth and metastasis. Natural products represent a rich source of compounds that have found many applications in various fields of medicines and therapy including cancer therapy. Effective ingredients in several plant-derived medicinal extracts are terpenoid compounds and many terpenes have biological activities and are used for the treatment of human diseases. OBJECTIVES: This review attempted to collect all available published scientific literature of eight naturally occurring terpenoids and their effect on inhibition of tumor progression. METHODS: The present review is about eight potent naturally occurring terpenoids that have been studied for their pharmacological properties in our lab and this review includes 130 references compiled from all major databases. RESULTS: Literature survey revealed that triterpenoids, such as glycyrrhizic acid, ursolic acid, oleanolic acid, and nomilin, the diterpene andrographolide, and the monoterpenoids like limonene and perillic acid had shown immunomodulatory and antitumor activities. All of them could induce apoptosis in various cancer cells by activating various proapoptotic signaling cascades. Many of these terpenoids found to inhibit metastatic progression and tumor-induced angiogenesis. The molecular mechanisms that involved in these activities include inhibition of various oncogenic and anti-apoptotic signaling pathways and suppression or nuclear translocation of various transcription factors including nuclear factor kappa B (NF-κB). CONCLUSION: The chemopreventive and chemoprotective effects of these compounds point toward their possible role in modern anticancer therapies.


Subject(s)
Disease Progression , Neoplasms/drug therapy , Plant Extracts/pharmacology , Terpenes/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Humans , Neoplasms/pathology , Neoplasms/prevention & control , Plant Extracts/therapeutic use , Plant Extracts/toxicity , Terpenes/therapeutic use , Terpenes/toxicity
15.
Mol Cancer ; 10: 107, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21880153

ABSTRACT

BACKGROUND: Increasing evidence indicates that the interaction between the CXC chemokine receptor-4 (CXCR4) and its ligand CXCL12 is critical in the process of metastasis that accounts for more than 90% of cancer-related deaths. Thus, novel agents that can downregulate the CXCR4/CXCL12 axis have therapeutic potential in inhibiting cancer metastasis. METHODS: In this report, we investigated the potential of an agent, plumbagin (5-hydroxy-2-methyl-1, 4-naphthoquinone), for its ability to modulate CXCR4 expression and function in various tumor cells using Western blot analysis, DNA binding assay, transient transfection, real time PCR analysis, chromatin immunoprecipitation, and cellular migration and invasion assays. RESULTS: We found that plumbagin downregulated the expression of CXCR4 in breast cancer cells irrespective of their HER2 status. The decrease in CXCR4 expression induced by plumbagin was not cell type-specific as the inhibition also occurred in gastric, lung, renal, oral, and hepatocellular tumor cell lines. Neither proteasome inhibition nor lysosomal stabilization had any effect on plumbagin-induced decrease in CXCR4 expression. Detailed study of the underlying molecular mechanism(s) revealed that the regulation of the downregulation of CXCR4 was at the transcriptional level, as indicated by downregulation of mRNA expression, inhibition of NF-κB activation, and suppression of chromatin immunoprecipitation activity. In addition, using a virtual, predictive, functional proteomics-based tumor pathway platform, we tested the hypothesis that NF-κB inhibition by plumbagin causes the decrease in CXCR4 and other metastatic genes. Suppression of CXCR4 expression by plumbagin was found to correlate with the inhibition of CXCL12-induced migration and invasion of both breast and gastric cancer cells. CONCLUSIONS: Overall, our results indicate, for the first time, that plumbagin is a novel blocker of CXCR4 expression and thus has the potential to suppress metastasis of cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Naphthoquinones/pharmacology , Receptors, CXCR4/metabolism , Breast Neoplasms , Cell Line, Tumor , Chemokine CXCL12/pharmacology , Chemokine CXCL12/physiology , Computer Simulation , Down-Regulation , Female , Genes, Reporter , Humans , Luciferases/biosynthesis , Luciferases/genetics , Models, Biological , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Invasiveness , Protein Binding , Receptors, CXCR4/genetics , Stomach Neoplasms , Transcription, Genetic/drug effects
16.
Apoptosis ; 16(10): 1028-41, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21786165

ABSTRACT

Celastrol, a plant triterpene has attracted great interest recently, especially for its potential anti-inflammatory and anti-cancer activities. In the present report, we investigated the effect of celastrol on proliferation of various cancer cell lines. The mechanism, by which this triterpene exerts its apoptotic effects, was also examined in detail. We found that celastrol inhibited the proliferation of wide variety of human tumor cell types including multiple myeloma, hepatocellular carcinoma, gastric cancer, prostate cancer, renal cell carcinoma, head and neck carcinoma, non-small cell lung carcinoma, melanoma, glioma, and breast cancer with concentrations as low as 1 µM. Growth inhibitory effects of celastrol correlated with a decrease in the levels of cyclin D1 and cyclin E, but concomitant increase in the levels of p21 and p27. The apoptosis induced by celastrol was indicated by the activation of caspase-8, bid cleavage, caspase-9 activation, caspase-3 activation, PARP cleavage and through the down regulation of anti-apoptototic proteins. The apoptotic effects of celastrol were preceded by activation of JNK and down-regulation of Akt activation. JNK was needed for celastrol-induced apoptosis, and inhibition of JNK by pharmacological inhibitor abolished the apoptotic effects. Overall, our results indicate that celastrol can inhibit cell proliferation and induce apoptosis through the activation of JNK, suppression of Akt, and down-regulation of anti-apoptotic protein expression.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Triterpenes/pharmacology , Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Humans , Neoplasms/drug therapy , Pentacyclic Triterpenes , Phosphoinositide-3 Kinase Inhibitors
17.
Br J Pharmacol ; 163(2): 283-98, 2011 May.
Article in English | MEDLINE | ID: mdl-21198544

ABSTRACT

BACKGROUND AND PURPOSE: Activation of signal transducer and activator of transcription 3 (STAT3) play a critical role in the survival, proliferation, angiogenesis and chemoresistance of tumour cells. Thus, agents that suppress STAT3 phosphorylation have potential as cancer therapies. In the present study, we investigated whether the apoptotic, antiproliferative and chemosensitizing effects of γ-tocotrienol are associated with its ability to suppress STAT3 activation in hepatocellular carcinoma (HCC). EXPERIMENTAL APPROACH: The effect of γ-tocotrienol on STAT3 activation, associated protein kinases and phosphatase, STAT3-regulated gene products, cellular proliferation and apoptosis in HCC cells was investigated. KEY RESULTS: γ-Tocotrienol inhibited both the constitutive and inducible activation of STAT3 with minimum effect on STAT5. γ-Tocotrienol also inhibited the activation of Src, JAK1 and JAK2 implicated in STAT3 activation. Pervanadate reversed the γ-tocotrienol-induced down-regulation of STAT3, suggesting the involvement of a protein tyrosine phosphatase. Indeed, we found that γ-tocotrienol induced the expression of the tyrosine phosphatase SHP-1 and deletion of the SHP-1 gene by small interfering RNA abolished the ability of γ-tocotrienol to inhibit STAT3 activation. γ-Tocotrienol also down-regulated the expression of STAT3-regulated gene products, including cyclin D1, Bcl-2, Bcl-xL, survivin, Mcl-1 and vascular endothelial growth factor. Finally, γ-tocotrienol inhibited proliferation, induced apoptosis and significantly potentiated the apoptotic effects of chemotherapeutic drugs (paclitaxel and doxorubicin) used for the treatment of HCC. CONCLUSIONS AND IMPLICATIONS: Overall, these results suggest that γ-tocotrienol is a novel blocker of the STAT3 activation pathway, with a potential role in future therapies for HCC and other cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Chromans/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Vitamin E/analogs & derivatives , Carcinoma, Hepatocellular , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Drug Synergism , Enzyme Activation , Enzyme Induction , Genes, Reporter , Humans , Janus Kinases/metabolism , Liver Neoplasms , NF-kappa B/metabolism , Paclitaxel/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatases/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Vitamin E/pharmacology , src-Family Kinases/metabolism
18.
Immunopharmacol Immunotoxicol ; 31(3): 377-87, 2009.
Article in English | MEDLINE | ID: mdl-19555203

ABSTRACT

The effect of Punarnavine on the immune system was studied using Balb/c mice. Intraperitoneal administration of Punarnavine (40 mg/kg body weight) was found to enhance the total WBC count on 6(th) day. Bone marrow cellularity and number of alpha-esterase positive cells were also increased by the administration of Punarnavine. Treatment of Punarnavine along with the antigen, sheep red blood cells (SRBC), produced an enhancement in the circulating antibody titer and the number of plaque forming cells (PFC) in the spleen. Maximum number of PFC was obtained on the 6(th) day. Punarnavine also showed enhanced proliferation of splenocytes, thymocytes and bone marrow cells both in the presence and absence of specific mitogens in vitro and in vivo. More over administration of Punarnavine significantly reduced the LPS induced elevated levels of proinflammatory cytokines such as TNF-alpha, IL-1beta, and IL-6 in mice. These results indicate the immunomodulatory activity of Punarnavine.


Subject(s)
Alkaloids/pharmacology , Bone Marrow Cells/immunology , Cell Proliferation/drug effects , Immunologic Factors/pharmacology , Nyctaginaceae/chemistry , Alkaloids/chemistry , Animals , Antibodies/immunology , Antibody Formation/drug effects , Antibody Formation/immunology , Bone Marrow Cells/cytology , Cytokines/immunology , Immunologic Factors/chemistry , Inflammation Mediators/immunology , Leukocyte Count , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , Spleen/cytology , Spleen/immunology , Thymus Gland/cytology , Thymus Gland/immunology
19.
Asian Pac J Cancer Prev ; 10(6): 1031-7, 2009.
Article in English | MEDLINE | ID: mdl-20192578

ABSTRACT

The objective of this study was to assess the effect of Punarnavine, an alkaloid isolated from Boerhaavia diffusa, on apoptosis in B16F-10 melanoma cells. Treatment of B16F-10 melanoma cells with nontoxic concentrations of Punarnvine resulted in the presence of apoptotic bodies and DNA fragmentation in a dose dependent manner. Cell cycle analysis and TUNEL assays also confirmed the observation. The apoptotic genes p53 and caspase-3 were found upregulated in Punarnavine treated cells, whereas the antiapoptotic gene Bcl-2 was downregulated. The inhibited nuclear translocation of NF-kappaBp65, NF-kappaBp50, NF-kappaB-c-Rel, c-Fos, ATF-2 and CREB-1 in Punarnavine treated B16 F-10 cells pointed to suppression of NF-kappaB signaling by Punarnavine. All these results demonstrate that Punarnavine induces apoptosis via activation of p53 induced caspase-3 mediated pro-apoptotic signaling and suppression of NF-kappaB induced Bcl-2 mediated survival signaling.


Subject(s)
Alkaloids/pharmacology , Apoptosis/drug effects , Melanoma, Experimental/drug therapy , NF-kappa B/metabolism , Animals , Caspase 3/genetics , Caspase 3/metabolism , Cell Cycle , Cell Line, Tumor , DNA Fragmentation , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Genes, bcl-2/genetics , Genes, p53/genetics , In Situ Nick-End Labeling , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , NF-kappa B/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...