Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Biochem J ; 480(20): 1675-1691, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37815456

ABSTRACT

Although Microrchidia 2 (MORC2) is widely overexpressed in human malignancies and linked to cancer cell proliferation, metabolism, and metastasis, the mechanism of action of MORC2 in cancer cell migration and invasion is yet undeciphered. Here, we identified for the first time that MORC2, a chromatin remodeler, regulates E-cadherin expression and, subsequently regulates breast cancer cell migration and invasion. We observed a negative correlation between the expression levels of MORC2 and E-cadherin in breast cancer. Furthermore, the overexpression of MORC2 resulted in decreased expression levels of E-cadherin. In addition, co-immunoprecipitation and chromatin immunoprecipitation assays revealed that MORC2 interacts with HDAC1 and gets recruited onto the E-cadherin promoter to inhibit its transcription, thereby suppress its expression. Consequently, knockdown of HDAC1 in MORC2-overexpressing cells led to reduced cancer cell migration and invasion. Interestingly, we noticed that MORC2-regulated glucose metabolism via c-Myc, and LDHA, also modulates the expression of E-cadherin. Collectively, these results demonstrate for the first time a mechanistic role for MORC2 as an upstream regulator of E-cadherin expression and its associated functions in breast cancer.


Subject(s)
Breast Neoplasms , Histone Deacetylase 1 , Humans , Female , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Cell Line, Tumor , Cadherins/genetics , Cadherins/metabolism , Breast Neoplasms/genetics , Gene Expression , Gene Expression Regulation, Neoplastic , Transcription Factors/metabolism
2.
Front Oncol ; 10: 626836, 2020.
Article in English | MEDLINE | ID: mdl-33680951

ABSTRACT

Forkhead box transcription factor, FOXM1 is implicated in several cellular processes such as proliferation, cell cycle progression, cell differentiation, DNA damage repair, tissue homeostasis, angiogenesis, apoptosis, and redox signaling. In addition to being a boon for the normal functioning of a cell, FOXM1 turns out to be a bane by manifesting in several disease scenarios including cancer. It has been given an oncogenic status based on several evidences indicating its role in tumor development and progression. FOXM1 is highly expressed in several cancers and has also been implicated in poor prognosis. A comprehensive understanding of various aspects of this molecule has revealed its role in angiogenesis, invasion, migration, self- renewal and drug resistance. In this review, we attempt to understand various mechanisms underlying FOXM1 gene and protein regulation in cancer including the different signaling pathways, post-transcriptional and post-translational modifications. Identifying crucial molecules associated with these processes can aid in the development of potential pharmacological approaches to curb FOXM1 mediated tumorigenesis.

3.
J Cell Physiol ; 233(10): 6938-6950, 2018 10.
Article in English | MEDLINE | ID: mdl-29665004

ABSTRACT

Mutations in p53 gene are one of the hallmarks of tumor development. Specific targeting of mutant p53 protein has a promising role in cancer therapeutics. Our preliminary observation showed destabilization of mutant p53 protein in SW480, MiaPaCa and MDAMB231 cell lines upon thiostrepton treatment. In order to elucidate the mechanism of thiostrepton triggered mutant p53 degradation, we explored the impact of proteasome inhibition on activation of autophagy. Combined treatment of thiostrepton and cycloheximide/chloroquine prevented the degradation of mutant p53 protein, reinforcing autophagy as the means of mutant p53 destabilization. Our initial studies suggested that mutant p53 degradation post THSP treatment was carried out by BAG3 mediated autophagy, based on the evidence of BAG1 to BAG3 switching. Subsequent interactome analysis performed post thiostrepton treatment revealed an association of p53 with autophagosome complex associated proteins such as BAG3, p62 and HSC70. Reaccumulation of p53 was seen in BAG3 silenced cells treated with thiostrepton, thereby confirming the role of BAG3 in destabilization of this molecule. Further, localization of p53 into the lysosome upon THSP treatment substantiated our findings that mutant p53 was degraded by an autopahgic process.


Subject(s)
Adaptor Proteins, Signal Transducing/drug effects , Autophagy/drug effects , Thiostrepton/pharmacology , Tumor Suppressor Protein p53/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Autophagosomes/drug effects , Autophagosomes/metabolism , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism
4.
BMC Cancer ; 18(1): 284, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29534682

ABSTRACT

BACKGROUND: Smurf2 E3 ubiquitin ligase physically associates with and regulate the stability of distinct cellular protein substrates. The multi-functional scaffold protein Connector enhancer of kinase suppressor of ras 2 (CNKSR2) plays a key role in regulating cell proliferation, and differentiation through multiple receptor tyrosine kinase pathways. The aim of this study was to investigate whether the interaction between Smurf2 and CNKSR2 has any significant role in the post transcriptional regulation of CNKSR2 expression in breast cancer. METHODS: Here we demonstrate a novel interaction of CNKSR2 with Smurf2 by co-immunoprecipitation, indirect immunofluorescence studies, and surface plasmon resonance (SPR) analysis, which can ubiquitinate, but stabilize CNKSR2 by protecting it from proteasome mediated degradation. RESULTS: CNKSR2 protein levels were significantly increased upon forced overexpression of Smurf2, indicating the role of Smurf2 in regulating the stability of CNKSR2. Conversely, Smurf2 knockdown resulted in a marked decrease in the protein level expression of CNKSR2 by facilitating enhanced polyubiquitination and proteasomal degradation and reduced the proliferation and clonogenic survival of MDA-MB-231 breast cancer cell lines. Tissue microarray data from 84 patients with various stages of mammary carcinoma, including (in order of increasing malignant potential) normal, usual hyperplasia, fibrocystic changes, fibroadenoma, carcinoma-in-situ, and invasive ductal carcinoma showed a statistically significant association between Smurf2 and CNKSR2 expression, which is also well correlated with the ER, PR, and HER2 status of the tissue samples. A comparatively high expression of Smurf2 and CNKSR2 was observed when the expression of ER and PR was low, and HER2 was high. Consistently, both Smurf2 and CNKSR2 showed an integrated expression in MCF10 breast progression model cell lines. CONCLUSIONS: Altogether, our findings reveal that Smurf2 is a novel positive regulator of CNKSR2 and suggest that Smurf2-CNKSR2 interaction may serve as a common strategy to control proliferation of human breast cancer cells by modulating CNKSR2 protein stability.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Proteasome Endopeptidase Complex/metabolism , Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Female , Humans , Intracellular Signaling Peptides and Proteins , Protein Interaction Maps , Protein Processing, Post-Translational , Protein Stability , Tumor Cells, Cultured , Ubiquitination
5.
BMC Cancer ; 16: 336, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27229859

ABSTRACT

BACKGROUND: Studies over the past decade and half have identified cancer stem cells (CSCs) to be responsible for tumorigenesis, invasion, sustenance of metastatic disease, radio- and chemo-resistance and tumor relapse. Recent reports have described the plasticity of breast CSCs (BCSCs) to shift between the epithelial and mesenchymal phenotypes via Epithelial-Mesenchymal Transition (EMT) and Mesenchymal-Epithelial Transition (MET) states as the reason for their invasive capabilities. Additionally, BRCA1 has been found to be a mammary stem cell fate determinant. However, it is not clear what would be the best marker that can be used for identifying CSCs in BRCA1 mutated cancers. Also, anticancer agents that can reduce CSC population in a BRCA1 defective condition have not been addressed so far. METHODS: Putative BCSCs were identified based on Hoechst exclusion, CD44(+)/24(-/low) expression and Aldehyde Dehydrogenase 1 (ALDH1) positivity using flow cytometry. The 'stemness' of the isolated ALDH1+ cells were analysed by immunofluorescence, western blotting for stem cell and EMT markers as well as in vitro mammosphere assays. Induction of Reactive Oxygen Species (ROS) by Plumbagin (PB) in BCSCs was assayed by Dichloro-dihydro-fluorescein diacetate (DCF-DA) staining. Ovarian cancer xenografts treated with PB were subjected to immunohistochemical analysis to study the ability of PB to target CSCs. RESULTS: We have confirmed that ALDH1 positivity is the best marker for the identification of BCSCs in BRCA1-defective breast cancer cell lines when compared to the CD marker profile and Side Population (SP) analysis. BRCA1 status was observed to be a determinant of the abundance of epithelial-like (ALDH1+) or mesenchymal-like (CD44(+)/24(-/low)) BCSCs, and the reconstitution of a full length, wild type BRCA1 in HCC1937 breast cancer cells possessing a mutated BRCA1, transforms them from 'stem-like' to more 'mesenchymal'. For the first time we have shown that Plumbagin (PB), a naturally occurring naphthoquinone which is predominantly a ROS inducer, could reduce BCSCs specifically in BRCA1-defective, basal-like cancer cells. CONCLUSIONS: The best marker for identifying BCSCs in BRCA1 defective condition could be ALDH1 and that BRCA1 mutated BCSCs would be mostly 'stem like' than 'mesenchymal'. Also ROS inducers like PB could reduce BCSCs in BRCA1 defective cancers.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , BRCA1 Protein/genetics , Breast Neoplasms/pathology , Naphthoquinones/pharmacology , Neoplastic Stem Cells/drug effects , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , BRCA1 Protein/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Carboplatin/pharmacology , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Humans , Inhibitory Concentration 50 , Mice, SCID , Naphthoquinones/therapeutic use , Neoplastic Stem Cells/metabolism , Xenograft Model Antitumor Assays
6.
Dalton Trans ; 44(35): 15591-601, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26244289

ABSTRACT

Novel biomimetic mononuclear complexes, [Fe()Cl2](+) () and [Cu()(H2O)](2+) () based on naphthalimide appended tripodal tetradentate ligand ( = 2,2',2''-(3,3',3''-(2,2',2''-nitrilotris(methylene)tris(1H-benzo[d]imidazole-2,1-diyl))tris(propane-3,1-diyl))tris(1H-benzo-[de]isoquinoline-1,3(2H)-dione)) have been synthesized and characterized by various analytical and spectral techniques. In addition, the structures of the ligand () and complex were established unambiguously through X-ray crystal structure analysis. Uniquely, the coordination with a metal ion modified the ligand scaffold to interact efficiently with ct-DNA (groove binding) as well as protein (hydrophobic and/or electrostatic interactions). We have determined the affinity of these complexes for DNA/protein and the values are found to be in the range, KDNA = 0.34-1.01 × 10(4) M(-1) and KBSA = 4.1-5.0 × 10(5) M(-1). Furthermore, the fluorescence quenching of BSA with complexes and occurs through a static mechanism and affects the conformation of BSA around the tryptophan residues. The in vitro biological studies of these systems employing HeLa cell lines indicated that both these complexes exhibited enhanced cytotoxicity (IC50 = 32 ± 0.19 and 10 ± 0.21 µM for complexes and , respectively), when compared to the ligand () (IC50 = 150 µM). Interestingly, both the complexes ( and ) were found to be non-toxic to normal H9C2 cell lines. The mechanism of in vitro biological activity of these complexes has been evaluated through a variety of techniques: acridine orange/ethidium bromide, DAPI staining studies, annexin V-FITC/PI and poly(ADP-ribose)-polymerase (PARP) cleavage, which confirmed the apoptotic mediated cell death. Our results demonstrate the importance of complexation of the naphthalimide ligand () as well as the potential of these biomimetic metal complexes as cytotoxic and anticancer agents.


Subject(s)
Antineoplastic Agents/chemistry , Coordination Complexes/chemistry , Copper/chemistry , Ferrous Compounds/chemistry , Naphthalimides/chemistry , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cattle , Coordination Complexes/pharmacology , Copper/pharmacology , Crystallography, X-Ray , DNA/metabolism , Ferrous Compounds/pharmacology , HeLa Cells , Humans , Ligands , Models, Molecular , Naphthalimides/pharmacology , Neoplasms/drug therapy
7.
ACS Chem Biol ; 8(1): 127-32, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23092119

ABSTRACT

We synthesized a novel water-soluble porphyrin THPP and its metalated derivative Zn-THPP having excellent triplet excited state quantum yields and singlet oxygen generation efficiency. When compared to U.S. Food and Drug Administration approved and clinically used sensitizer Photofrin, THPP showed ca. 2-3-fold higher in vitro photodynamic activity in different cell lines under identical conditions. The mechanism of the biological activity of these porphyrin systems has been evaluated through a variety of techniques: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, comet assay, poly(ADP-ribose)polymerase (PARP) cleavage, CM-H(2)DCFDA assay, DNA fragmentation, flow cytometric analysis, fluorescence, and confocal microscopy, which confirm the apoptotic cell death through predominantly reactive oxygen species (ROS). Moreover, THPP showed rapid cellular uptake and are localized in the nucleus of the cells as compared to Hoechst dye and Photofrin, thereby demonstrating its use as an efficient sensitizer in photodynamic therapy and live cell NIR nucleus imaging applications.


Subject(s)
Coordination Complexes/chemical synthesis , Photosensitizing Agents/chemical synthesis , Porphyrins/chemical synthesis , Zinc/chemistry , Animals , Apoptosis/drug effects , Cell Line, Tumor , Coordination Complexes/pharmacology , Humans , Microscopy, Confocal , Molecular Structure , Photosensitizing Agents/pharmacology , Porphyrins/chemistry , Porphyrins/pharmacology
8.
Clin Exp Metastasis ; 29(5): 471-92, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22419013

ABSTRACT

Nitric oxide (NO), an uncharged free radical is implicated in various physiological and pathological processes. The present study is an investigation on the effect of NO on proliferation, apoptosis and migration of colon cancer cells. Colon adenocarcinoma cells, WiDr, were used for the in vitro experiments. Tissues from colon adenocarcinoma, adjacent normal and inflammatory tissue and lymph node with metastasis were evaluated for iNOS, MMP-2/9 and Fra-1/Fra-2. NO increases the proliferation of cancer cells and simultaneously prevents apoptosis. Expression of MMP-2/9, RhoB and Rac-1 was enhanced by NO in a time dependent manner. Further, NO increased phosphorylation of ERK1/2 and induced nuclear translocation of Fra-1 and Fra-2. Electrophoretic mobility shift analysis and use of deletion mutant promoter constructs identified role of AP-1 in NO-mediated regulation of MMP-2/9. iNOS, MMP-2/9, Fra-1 and Fra-2 in normal and colon adenocarcinoma tissues were analyzed and it was found that increased expression of these proteins in cancer when compared to normal provides support to our in vitro findings. The study showed that the NO-cGMP-PKG promotes MMP-2/9 expression by activating ERK-1/2 and AP-1. This study reveals the insidious role of NO in imparting tumor aggressiveness.


Subject(s)
Cell Movement , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Nitric Oxide/metabolism , Signal Transduction , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Blotting, Western , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/genetics , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Humans , MAP Kinase Signaling System , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Up-Regulation
9.
J Exp Clin Cancer Res ; 30: 56, 2011 May 16.
Article in English | MEDLINE | ID: mdl-21575192

ABSTRACT

BACKGROUND: Signal transducer and activator of transcription 3 (STAT3) is a key signaling molecule and a central cytoplasmic transcription factor, implicated in the regulation of growth. Its aberrant activation has been demonstrated to correlate with many types of human malignancy. However, whether constitutive STAT3 signaling plays a key role in the survival and growth of soft-tissue tumors is still unclear and hence needs to be elucidated further. In our study we examined the expression levels of STAT3 and pSTAT3 in different grades of soft tissue tumors and correlated with its clinicopathological characteristics. METHODS: Expression levels of STAT3 and pSTAT3 in soft tissue tumors were studied using Immunohistochemistry, Western blotting and Reverse transcriptase- PCR and correlated with its clinicopathological characteristics using Chi squared or Fisher's exact test and by logistic regression analysis. Statistical analysis was done using Intercooled Stata software (Intercooled Stata 8.2 version). RESULTS: Of the 82 soft tissue tumor samples, fifty four (65.8%) showed immunoreactivity for STAT3 and twenty eight (34.1%) for pSTAT3. Expression of STAT3 and pSTAT3 was significantly associated with tumor grade (P < 0.001; P < 0.001), tumor location (P = 0.025; P = 0.027), plane of tumor (P = 0.011; P = 0.006), and tumor necrosis (P = 0.001; P = 0.002). Western blotting and RT-PCR analysis showed increased expression of STAT3 and p-STAT3 as grade of malignancy increased. CONCLUSION: These findings suggest that constitutive activation of STAT3 is an important factor related to carcinogenesis of human soft tissue tumors and is significantly associated with its clinicopathological parameters which may possibly have potential diagnostic implications.


Subject(s)
Gene Expression Regulation, Neoplastic , STAT3 Transcription Factor/metabolism , Soft Tissue Neoplasms/physiopathology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Neoplasm Staging , Phosphorylation , Prognosis , RNA, Messenger/metabolism , STAT3 Transcription Factor/genetics , Soft Tissue Neoplasms/diagnosis , Young Adult
10.
Mol Cancer Res ; 7(1): 118-28, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19147543

ABSTRACT

Activation of signal transducers and activators of transcription-3 (STAT-3) has been linked with survival, proliferation, chemoresistance, and angiogenesis of tumor cells, including human multiple myeloma (MM). Thus, agents that can suppress STAT3 activation have potential as cancer therapeutics. In our search for such agents, we identified acetyl-11-keto-beta-boswellic acid (AKBA), originally isolated from Boswellia serrata. Our results show that AKBA inhibited constitutive STAT3 activation in human MM cells. AKBA suppressed IL-6-induced STAT3 activation, and the inhibition was reversible. The phosphorylation of both Jak 2 and Src, constituents of the STAT3 pathway, was inhibited by AKBA. Interestingly, treatment of cells with pervanadate suppressed the effect of AKBA to inhibit the phosphorylation of STAT3, thus suggesting the involvement of a protein tyrosine phosphatase. We found that AKBA induced Src homology region 2 domain-containing phosphatase 1 (SHP-1), which may account for its role in dephosphorylation of STAT3. Moreover, deletion of the SHP-1 gene by small interfering RNA abolished the ability of AKBA to inhibit STAT3 activation. The inhibition of STAT3 activation by AKBA led to the suppression of gene products involved in proliferation (cyclin D1), survival (Bcl-2, Bcl-xL, and Mcl-1), and angiogenesis (VEGF). This effect correlated with the inhibition of proliferation and apoptosis in MM cells. Consistent with these results, overexpression of constitutive active STAT3 significantly reduced the AKBA-induced apoptosis. Overall, our results suggest that AKBA is a novel inhibitor of STAT3 activation and has potential in the treatment of cancer.


Subject(s)
Cell Survival/drug effects , Multiple Myeloma/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Transcription, Genetic/physiology , Triterpenes/pharmacology , Actins/genetics , Cell Line, Tumor , Flow Cytometry , Humans , Multiple Myeloma/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , RNA, Neoplasm/drug effects , RNA, Neoplasm/genetics , RNA, Small Interfering/genetics , STAT3 Transcription Factor/antagonists & inhibitors , Transcription, Genetic/drug effects
11.
Indian J Gastroenterol ; 27(4): 162-4, 2008.
Article in English | MEDLINE | ID: mdl-18974468

ABSTRACT

Isolated cases and small series of acute pancreatitis complicating viral infections have been reported. However, data on the natural history of such patients are sparse. We report a series of five patients of acute pancreatitis complicating diverse viral infections. During follow-up ranging from 6 to 36 months, two of these five patients developed evidence of chronic pancreatitis.


Subject(s)
Pancreatitis/etiology , Virus Diseases/complications , Abdominal Pain/blood , Abdominal Pain/diagnosis , Abdominal Pain/etiology , Acute Disease , Adolescent , Adult , Amylases/blood , Chronic Disease , Disease Progression , Female , Follow-Up Studies , Humans , Male , Middle Aged , Pancreatitis/blood , Ultrasonography , Virus Diseases/therapy
12.
Trop Gastroenterol ; 29(3): 175-6, 2008.
Article in English | MEDLINE | ID: mdl-19115613

ABSTRACT

Tropical pancreatitis and fibrocalculous pancreatitic diabetes are usually considered two ends of the same disease although some studies suggest the likelihood of distinct entities. We report the occurrence of these two diseases in two siblings. The literature comparing the similarities and differences between them is briefly reviewed.


Subject(s)
Calcinosis/diagnosis , Diabetes Complications/diagnosis , Pancreatitis/etiology , Pancreatitis/pathology , Adult , Calcinosis/complications , Diabetes Complications/complications , Humans , Male
13.
Mol Cancer Res ; 5(9): 943-55, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17855663

ABSTRACT

The activation of signal transducers and activators of transcription 3 (STAT3) has been linked with the proliferation of a variety of human cancer cells, including multiple myeloma. Agents that can suppress STAT3 activation have potential for prevention and treatment of cancer. In the present report, we tested an agent, ursolic acid, found in basil, apples, prunes, and cranberries, for its ability to suppress STAT3 activation. We found that ursolic acid, a pentacyclic triterpenoid, inhibited both constitutive and interleukin-6-inducible STAT3 activation in a dose- and time-dependent manner in multiple myeloma cells. The suppression was mediated through the inhibition of activation of upstream kinases c-Src, Janus-activated kinase 1, Janus-activated kinase 2, and extracellular signal-regulated kinase 1/2. Vanadate treatment reversed the ursolic acid-induced down-regulation of STAT3, suggesting the involvement of a tyrosine phosphatase. Indeed, we found that ursolic acid induced the expression of tyrosine phosphatase SHP-1 protein and mRNA. Moreover, knockdown of SHP-1 by small interfering RNA suppressed the induction of SHP-1 and reversed the inhibition of STAT3 activation, thereby indicating the critical role of SHP-1 in the action of this triterpene. Ursolic acid down-regulated the expression of STAT3-regulated gene products such as cyclin D1, Bcl-2, Bcl-xL, survivin, Mcl-1, and vascular endothelial growth factor. Finally, ursolic acid inhibited proliferation and induced apoptosis and the accumulation of cells in G1-G0 phase of cell cycle. This triterpenoid also significantly potentiated the apoptotic effects of thalidomide and bortezomib in multiple myeloma cells. Overall, these results suggest that ursolic acid is a novel blocker of STAT3 activation that may have a potential in prevention and treatment of multiple myeloma and other cancers.


Subject(s)
Cell Division/drug effects , STAT3 Transcription Factor/genetics , Triterpenes/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Immunohistochemistry , Multiple Myeloma/pathology , Oligonucleotide Probes , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/drug effects , Transfection , Ursolic Acid
14.
Clin Cancer Res ; 13(10): 3024-32, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17505005

ABSTRACT

PURPOSE: Capsaicin, a constituent of green and red peppers, has been linked with suppression of tumorigenesis through a mechanism that is not well understood. Because the transcription factor signal transducer and activator of transcription 3 (STAT3) has been closely linked with tumorigenesis, we investigated the effect of this vanilloid on the STAT3 pathway in human multiple myeloma cells. EXPERIMENTAL DESIGN: The effect of capsaicin on both constitutive and interleukin-6-induced STAT3 activation, associated protein kinases, and STAT3-regulated gene products involved in proliferation, survival and angiogenesis, cellular proliferation, and apoptosis in multiple myeloma cells was investigated. RESULTS: We found that capsaicin inhibited constitutive activation of STAT3 in multiple myeloma cells in a dose- and time-dependent manner, with minimum effect on STAT5. Capsaicin also inhibited the interleukin-6-induced STAT3 activation. The activation of Janus-activated kinase 1 and c-Src, implicated in STAT3 activation, was also inhibited by the vanilloid, with no effect on extracellular signal-regulated kinase 1/2 activation. Pervanadate reversed the capsaicin-induced down-regulation of STAT3, suggesting the involvement of a protein tyrosine phosphatase. Capsaicin down-regulated the expression of the STAT3-regulated gene products, such as cyclin D1, Bcl-2, Bcl-xL, survivin, and vascular endothelial growth factor. Finally, capsaicin induced the accumulation of cells in G(1) phase, inhibited proliferation, and induced apoptosis, as indicated by caspase activation. Capsaicin also significantly potentiated the apoptotic effects of Velcade and thalidomide in multiple myeloma cells. When administered i.p., capsaicin inhibited the growth of human multiple myeloma xenograft tumors in male athymic nu/nu mice. CONCLUSION: Overall, these results suggest that capsaicin is a novel blocker of the STAT3 activation pathway, with a potential role in the prevention and treatment of multiple myeloma and other cancers.


Subject(s)
Capsaicin/pharmacology , Multiple Myeloma/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis , Boronic Acids/pharmacology , Bortezomib , Caspase 3/metabolism , Caspase Inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/antagonists & inhibitors , DNA/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , G1 Phase/drug effects , Humans , Inhibitor of Apoptosis Proteins , Interleukin-6/pharmacology , Janus Kinase 1/antagonists & inhibitors , Microtubule-Associated Proteins/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Pyrazines/pharmacology , STAT3 Transcription Factor/metabolism , Survivin , Thalidomide/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , bcl-X Protein/antagonists & inhibitors
15.
Blood ; 109(12): 5112-21, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17332240

ABSTRACT

Gossypin, a flavone originally isolated from Hibiscus vitifolius, has been shown to suppress angiogenesis, inflammation, and carcinogenesis. The mechanisms of these activities, however, are unknown. Because nuclear factor-kappaB (NF-kappaB) is associated with inflammation, carcinogenesis, hyperproliferation, invasion, and angiogenesis, we hypothesized that gossypin mediates its effects through modulation of NF-kappaB activation. In the present study, we demonstrate that gossypin (and not gossypetin, an aglycone analog) inhibited NF-kappaB activation induced by inflammatory stimuli and carcinogens. Constitutive NF-kappaB activation in tumor cells was also inhibited by this flavone. Inhibition of I kappa B alpha kinase by gossypin led to the suppression of I kappa B alpha phosphorylation and degradation, p65 nuclear translocation, and NF-kappaB-regulated gene expression. This, in turn, led to the down-regulation of gene products involved in cell survival (IAP2, XIAP, Bcl-2, Bcl-xL, survivin, and antiFas-associated death domain-like interleukin-1 beta-converting enzyme-inhibitory protein), proliferation (c-myc, cyclin D1, and cyclooxygenase-2), angiogenesis (vascular endothelial growth factor), and invasion (matrix metalloprotease-9). Suppression of these gene products by gossypin enhanced apoptosis induced by tumor necrosis factor and chemotherapeutic agents, suppressed tumor necrosis factor-induced cellular invasion, abrogated receptor activator of NF-kappaB ligand-induced osteoclastogenesis, and vascular endothelial growth factor-induced migration of human umbilical vein endothelial cells. Overall, our results demonstrate that gossypin inhibits the NF-kappaB activation pathway, which may explain its role in the suppression of inflammation, carcinogenesis, and angiogenesis.


Subject(s)
Apoptosis/drug effects , Cell Movement/drug effects , Flavonoids/pharmacology , I-kappa B Kinase/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Osteoclasts/drug effects , Transforming Growth Factor beta1/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Angiogenesis Inhibitors/pharmacology , Anti-Inflammatory Agents/pharmacology , Anticarcinogenic Agents/pharmacology , Cell Line, Tumor , Cells, Cultured , Down-Regulation/genetics , Endothelium, Vascular/cytology , Enzyme Activation/drug effects , Flavones , Humans , NF-kappa B/metabolism
16.
Blood ; 109(6): 2293-302, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17164350

ABSTRACT

Whether resveratrol, a component of red grapes, berries, and peanuts, could suppress the proliferation of multiple myeloma (MM) cells by interfering with NF-kappaB and STAT3 pathways, was investigated. Resveratrol inhibited the proliferation of human multiple myeloma cell lines regardless of whether they were sensitive or resistant to the conventional chemotherapy agents. This stilbene also potentiated the apoptotic effects of bortezomib and thalidomide. Resveratrol induced apoptosis as indicated by accumulation of sub-G(1) population, increase in Bax release, and activation of caspase-3. This correlated with down-regulation of various proliferative and antiapoptotic gene products, including cyclin D1, cIAP-2, XIAP, survivin, Bcl-2, Bcl-xL, Bfl-1/A1, and TRAF2. In addition, resveratrol down-regulated the constitutive activation of AKT. These effects of resveratrol are mediated through suppression of constitutively active NF-kappaB through inhibition of IkappaBalpha kinase and the phosphorylation of IkappaBalpha and of p65. Resveratrol inhibited both the constitutive and the interleukin 6-induced activation of STAT3. When we examined CD138(+) plasma cells from patients with MM, resveratrol inhibited constitutive activation of both NF-kappaB and STAT3, leading to down-regulation of cell proliferation and potentiation of apoptosis induced by bortezomib and thalidomide. These mechanistic findings suggest that resveratrol may have a potential in the treatment of multiple myeloma.


Subject(s)
Apoptosis/drug effects , Down-Regulation/drug effects , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Stilbenes/pharmacology , Boronic Acids/pharmacology , Bortezomib , Caspase 3/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Enzyme Activation , Gene Expression Regulation, Neoplastic , Humans , Interleukin-6/metabolism , Multiple Myeloma/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyrazines/pharmacology , Resveratrol , Syndecan-1/metabolism , Thalidomide/pharmacology , Tumor Cells, Cultured , bcl-2-Associated X Protein/metabolism
17.
J Immunol ; 177(8): 5612-22, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17015749

ABSTRACT

Deguelin, a constituent of the bark of the African plant Mundulea sericea (Leguminosae), exhibits antiproliferative and anticarcinogenic activities through a mechanism that is not well understood. Because various steps in carcinogenesis are regulated by NF-kappaB, we postulated that the activity of deguelin is mediated through this transcription factor. We found that deguelin suppressed NF-kappaB activation induced by carcinogens, tumor promoters, growth factors, and inflammatory stimuli. This suppression was not cell-type specific, because NF-kappaB activation was suppressed in both lymphoid and epithelial cells. Moreover, constitutive NF-kappaB activation was also blocked by deguelin. The suppression of TNF-induced NF-kappaB activation by deguelin occurred through the inhibition of the activation of IkappaBalpha kinase, leading to sequential suppression of IkappaBalpha phosphorylation, IkappaBalpha degradation, p65 phosphorylation, p65 nuclear translocation, and NF-kappaB-dependent reporter gene expression. Deguelin also suppressed the NF-kappaB reporter activity induced by TNFR1, TNFR-associated death domain, TNFR-associated factor 2, and IkappaBalpha kinase, but not that induced by p65. The inhibition of NF-kappaB activation thereby led to the down-regulation of gene products involved in cell survival, proliferation, and invasion. Suppression of these gene products by deguelin enhanced the apoptosis induced by TNF and chemotherapeutic agents and suppressed TNF-induced cellular invasion. Our results demonstrate that deguelin inhibits the NF-kappaB activation pathway, which may explain its role in the suppression of carcinogenesis and cellular proliferation.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , I-kappa B Kinase/antagonists & inhibitors , NF-kappa B/physiology , Neoplasm Invasiveness/prevention & control , Rotenone/analogs & derivatives , Antineoplastic Agents/pharmacology , Cells, Cultured , Drug Interactions , Epithelial Cells/drug effects , Humans , I-kappa B Kinase/metabolism , Lymphocytes/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Rotenone/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...