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










Publication year range
1.
Comput Biol Med ; 145: 105404, 2022 06.
Article in English | MEDLINE | ID: mdl-35339097

ABSTRACT

BACKGROUND: Matrix metalloproteinase 3 (MMP3) plays a crucial role in cancer progression and development by proteolyzing extracellular matrix substrates. Primarily, the expression of MMP3 is regulated at the transcriptional level. The minute interplay of various transcription factor binding motifs at the promoter region is responsible for the altered expression of the genes. Single nucleotide polymorphism (SNP) at the transcription factor binding sites shows specific effects on gene expressions. Genome-wide association study (GWAS) strongly reported the association of common SNPs (rs3025058, rs522616, and rs617819) of MMP3 promoter with disease progression. The insufficient functional analysis of these promoter SNPs indicates the need for extensive mechanistic analysis on the effects of allelic variants upon transcription factor binding at MMP3 promoter. METHODS: The binding of transcription factors on the MMP3 promoter sequence was investigated by a virtual laboratory. The interaction between the specific transcription factor and promoter DNA with allelic variants was analyzed by computational tools. RESULTS: It was found that transcription factor c-Myb and Smad4 binding on MMP3 promoter were altered due to the presence of rs522616 and rs617819 SNPs, respectively. Further, the binding affinity of Smad4 to the MMP3 promoter containing C allele at -375 region is higher than that of its allelic variant G. CONCLUSION: This study presented that the complex of Smad4-DNA fragment containing C allele has higher binding affinity and stability as compared with its allelic variant. Hence, it is predicted that rs617819 polymorphism directly affects the Smad4 binding motif on MMP3 promoter and alters its gene expression.


Subject(s)
Genome-Wide Association Study , Matrix Metalloproteinase 3 , DNA , Matrix Metalloproteinase 3/genetics , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/genetics
4.
Oncotarget ; 11(13): 1097-1108, 2020 Mar 31.
Article in English | MEDLINE | ID: mdl-32284788

ABSTRACT

Glioblastoma (GBM) is the most common primary brain tumor and is invariably fatal. Heat shock proteins (HSPs) provide protein signatures/biomarkers for GBM that afford potential as targets for developing anti-GBM drugs. In GBM, elevated expression of hypoxia inducible factors under the influence of Ets family proteins significantly promotes the expression of HSPs. RNAseq analysis identified HSPB1 as a prominent upregulated HSP in GBM and in radiation resistant/temozolomide resistant (radio/TMZR) GBM. Here, we established friend leukemia integration 1 (Fli-1), a member of Ets family to be playing a transcriptional regulatory role on the HSPB1 gene. Fli-1 binds to nucleotide residues GGAA at binding sites 3, 6 and 7 in the 5-kb upstream region of HSPB1. Fli-1 has been linked to oncogenic transformation with upregulation in radio/TMZR GBM. Overexpression of Fli-1 in GBM promotes resistance, whereas Fli-1 knockdown in radio/TMZR GBM cells suppresses resistance. We identify the underlying molecular mechanisms of Fli-1-mediated regulation of HSPB1 that drive extracellular matrix remodeling and epithelial to mesenchymal transition in radio/TMZR GBM cells. This study uncovers Fli-1 as a potential therapeutic target for combating radiation and temozolomide resistance in GBM.

5.
Biochem Pharmacol ; 164: 1-16, 2019 06.
Article in English | MEDLINE | ID: mdl-30885764

ABSTRACT

Glioblastoma (GBM) is the most malignant form of brain tumor posing a major threat to cancer amelioration. Temozolomide (TMZ) resistance is one of the major hurdles towards GBM prognosis. Oxidative stress and ECM remodeling are the two important processes involved in gaining chemo-resistance. Here, we established NFE2L2, an important member of oxidative stress regulation elevated in resistant cells, to be playing a transcriptional regulatory role on MMP-2, an ECM remodeling marker. This link led us to further explore targeted molecules to inhibit NFE2L2, thus affecting MMP-2, an important member promoting chemo-resistance. Thus, diosgenin was proposed as a novel NFE2L2 inhibitor acting as an alternative strategy to prevent the high dose administration of TMZ. Combinatorial therapy of diosgenin and TMZ significantly reduced the dosage regimen of TMZ and also showed affectivity in hitherto TMZ resistant GBM cells. GBM cells underwent apoptosis and early cell cycle arrest with significant reduction in MMP-2 levels. Thus preclinical validation of molecular interaction between diosgenin and NFE2L2 down-regulating MMP-2, EMT markers and promoting apoptosis, offers rationale for new therapeutic horizons in the field of glioblastoma management.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Matrix Metalloproteinase 2/metabolism , NF-E2-Related Factor 2/metabolism , Temozolomide/administration & dosage , Animals , Base Sequence , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Matrix Metalloproteinase 2/genetics , Mice, Nude , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , Protein Structure, Tertiary , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Cancer Lett ; 452: 254-263, 2019 06 28.
Article in English | MEDLINE | ID: mdl-30904616

ABSTRACT

Epithelial to mesenchymal transition (EMT) is compulsory for metastatic dissemination and is stimulated by TGF-ß. Although targeting EMT has significant therapeutic potential, very few pharmacological agents have been shown to exert anti-metastatic effects. BI-69A11, a competitive Akt inhibitor, displays anti-tumor activity toward melanoma and colon carcinoma. This study provides molecular and biochemical insights into the effects of BI-69A11 on EMT in colon carcinoma cells in vitro and in vivo. BI-69A11 inhibited metastasis-associated cellular migration, invasion and adhesion by inhibiting the Akt-ß-catenin pathway. The underlying mechanism of BI-69A11-mediated inhibition of EMT included suppression of nuclear transport of ß-catenin and diminished phosphorylation of ß-catenin, which was accompanied by enhanced E-cadherin-ß-catenin complex formation at the plasma membrane. Additionally, BI-69A11 caused increased accumulation of vinculin in the plasma membrane, which fortified focal adhesion junctions leading to inhibition of metastasis. BI-69A11 downregulated activation of the TGF-ß-induced non-canonical Akt/NF-κB pathway and blocked TGF-ß-induced enhanced expression of Snail causing restoration of E-cadherin. Overall, this study enhances our understanding of the molecular mechanism of BI-69A11-induced reversal of EMT in colorectal carcinoma cells in vitro, in vivo and in TGF-ß-induced model systems.


Subject(s)
Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Cadherins/metabolism , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Quinolones/pharmacology , beta Catenin/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Colorectal Neoplasms/drug therapy , Humans , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Vinculin/metabolism
7.
Oncogene ; 37(33): 4546-4561, 2018 08.
Article in English | MEDLINE | ID: mdl-29743594

ABSTRACT

Although there is a strong correlation between multinucleated cells (MNCs) and cancer chemo-resistance in variety of cancers, our understanding of how multinucleated cells modulate the tumor micro-environment is limited. We captured multinucleated cells from triple-negative chemo-resistant breast cancers cells in a time frame, where they do not proliferate but rather significantly regulate their micro-environment. We show that oxidatively stressed MNCs induce chemo-resistance in vitro and in vivo by secreting VEGF and MIF. These factors act through the RAS/MAPK pathway to induce chemo-resistance by upregulating anti-apoptotic proteins. In MNCs, elevated reactive oxygen species (ROS) stabilizes HIF-1α contributing to increase production of VEGF and MIF. Together the data indicate, that the ROS-HIF-1α signaling axis is very crucial in regulation of chemo-resistance by MNCs. Targeting ROS-HIF-1α in future may help to abrogate drug resistance in breast cancer.


Subject(s)
Drug Resistance, Neoplasm/physiology , Reactive Oxygen Species/metabolism , Triple Negative Breast Neoplasms/metabolism , Breast/metabolism , Cell Line, Tumor , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Signal Transduction/physiology , Tumor Microenvironment/physiology , Vascular Endothelial Growth Factor A/metabolism
8.
Cell Signal ; 35: 24-36, 2017 07.
Article in English | MEDLINE | ID: mdl-28347875

ABSTRACT

Tumor angiogenesis and invasion are deregulated biological processes that drive multistage transformation of tumors from a benign to a life-threatening malignant state activating multiple signaling pathways including MD-2/TLR4/NF-κB. Development of potential inhibitors of this signaling is emerging area for discovery of novel cancer therapeutics. In the current investigation, we identified Iturin A (A lipopeptide molecule from Bacillus megaterium) as a potent inhibitor of angiogenesis and cancer invasion by various in vitro and in vivo methods. Iturin A was found to suppress VEGF, a powerful inducer of angiogenesis and key player in tumor invasion, as confirmed by ELISA, western blot and real time PCR. Iturin A inhibited endothelial tube arrangement, blood capillary formation, endothelial sprouting and vascular growth inside the matrigel. In addition, Iturin A inhibited MMP-2/9 expression in MDA-MB-231 and HUVEC cells. Cancer invasion, migration and colony forming ability were significantly hampered by Iturin A. Expressions of MD-2/TLR4 and its downstream MyD88, IKK-α and NF-κB were also reduced in treated MDA-MB-231 and HUVEC cells. Western blot and immunofluorescence study showed that nuclear accumulation of NF-κB was hampered by Iturin A. MD-2 siRNA or plasmid further confirmed the efficacy of Iturin A by suppressing MD-2/TLR4 signaling pathway. The in silico docking study showed that the Iturin A interacted well with the MD-2 in MD-2/TLR4 receptor complex. Conclusively, inhibition of MD-2/TLR4 complex with Iturin A offered strategic advancement in cancer therapy.


Subject(s)
Lymphocyte Antigen 96/genetics , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Peptides, Cyclic/administration & dosage , Toll-Like Receptor 4/genetics , Bacillus megaterium/chemistry , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Human Umbilical Vein Endothelial Cells , Humans , I-kappa B Kinase/genetics , Lymphocyte Antigen 96/chemistry , Myeloid Differentiation Factor 88/genetics , NF-kappa B/genetics , Neoplasm Invasiveness/genetics , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Peptides, Cyclic/chemistry , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Toll-Like Receptor 4/chemistry
9.
Acta Pharmacol Sin ; 38(5): 591-613, 2017 May.
Article in English | MEDLINE | ID: mdl-28317871

ABSTRACT

Glioma accounts for the majority of human brain tumors. With prevailing treatment regimens, the patients have poor survival rates. In spite of current development in mainstream glioma therapy, a cure for glioma appears to be out of reach. The infiltrative nature of glioma and acquired resistance substancially restrict the therapeutic options. Better elucidation of the complicated pathobiology of glioma and proteogenomic characterization might eventually open novel avenues for the design of more sophisticated and effective combination regimens. This could be accomplished by individually tailoring progressive neuroimaging techniques, terminating DNA synthesis with prodrug-activating genes, silencing gliomagenesis genes (gene therapy), targeting miRNA oncogenic activity (miRNA-mRNA interaction), combining Hedgehog-Gli/Akt inhibitors with stem cell therapy, employing tumor lysates as antigen sources for efficient depletion of tumor-specific cancer stem cells by cytotoxic T lymphocytes (dendritic cell vaccination), adoptive transfer of chimeric antigen receptor-modified T cells, and combining immune checkpoint inhibitors with conventional therapeutic modalities. Thus, the present review captures the latest trends associated with the molecular mechanisms involved in glial tumorigenesis as well as the limitations of surgery, radiation and chemotherapy. In this article we also critically discuss the next generation molecular therapeutic strategies and their mechanisms for the successful treatment of glioma.


Subject(s)
Brain Neoplasms/therapy , Brain/pathology , Glioma/therapy , Molecular Targeted Therapy/trends , Antineoplastic Agents/therapeutic use , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/pathology , Glioma/genetics , Glioma/pathology , Humans , Molecular Targeted Therapy/methods
10.
Macromol Biosci ; 17(4)2017 04.
Article in English | MEDLINE | ID: mdl-27879056

ABSTRACT

Stimuli-sensitive polymeric vesicles or polymersomes as self-assembled colloidal nanocarriers have received paramount importance for their integral role as delivery system for therapeutics and biotherapeutics. This work describes spontaneous polymersome formation at pH 7, as evidenced by surface tension, steady state fluorescence, dynamic light scattering, and microscopic studies, by three hydrophilic random cationic copolymers synthesized using N,N-(dimethylamino)ethyl methacrylate (DMAEM) and methoxy poly(ethylene glycol) monomethacrylate in different mole ratios. The results suggest that methoxy poly(ethylene glycol) chains constitute the bilayer membrane of the polymersomes and DMAEM projects toward water constituting the positively charged surface. The polymersomes have been observed to release their encapsulated guest at acidic pH as a result of transformation into polymeric micelles. All these highly biocompatible cationic polymers show successful gene transfection ability as nonviral vector on human cell line with different potential. Thus these polymers prove their utility as a potential delivery system for hydrophilic model drug as well as genetic material.


Subject(s)
Drug Delivery Systems/methods , Gene Transfer Techniques , Polymers/chemistry , Cations , Cell Line, Tumor , Cell Survival , Circular Dichroism , DNA/metabolism , Drug Liberation , Electrophoretic Mobility Shift Assay , Humans , Hydrodynamics , Hydrogen-Ion Concentration , Materials Testing , Microscopy, Electron, Transmission , Particle Size , Plasmids/metabolism , Polymers/chemical synthesis , Proton Magnetic Resonance Spectroscopy , Serum Albumin/metabolism , Spectrometry, Fluorescence , Surface Tension , Temperature , Transfection
12.
Eur J Pharmacol ; 765: 217-27, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26306675

ABSTRACT

BI-69A11, novel Akt inhibitor, is currently drawing much attention due to its intriguing effect in inducing apoptosis in melanoma, breast, prostate and colon cancer. However, earlier reports reveal that PI3K/Akt/mTOR inhibitors promote autophagy at the early stage as a survival mechanism that might affect its apoptotic potential. It is necessary to investigate whether BI-69A11 mediated apoptosis is associated with autophagy for enhancing its therapeutic efficacy. Here, we found that BI-69A11 induced autophagy at earlier time point through the inhibition of Akt/mTOR/p70S6kinase pathway. Dose-dependent and time-dependent conversion of LC3-I to LC3-II, increased accumulation of LC3-GFP dots in cytoplasm and increase in other autophagic markers such as Beclin-1, firmly supported the fact that BI-69A11 induces autophagy. Atg5, Atg7 and Beclin-1 siRNA mediated genetic attenuation and pre-treatment with pharmacological inhibitor 3-MA and CQ diminished the autophagy and increased the propensity of cell death towards apoptosis. It was also suggested that BI-69A11 mediated interaction between Akt, HSP-90 and Beclin-1 maintained the fine balance between autophagy and apoptosis. Interaction between Beclin-1 and HSP90 is one of the prime causes of induction of autophagy. Here, we also generated a novel combination therapy by pretreatment with CQ that inhibited the autophagy and accelerated the apoptotic potential of BI-69A11. In summary; our findings suggest that induction of autophagy lead to the resistance of colon cancer towards BI-69A11 mediated apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Benzimidazoles/pharmacology , Colonic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Quinolones/pharmacology , Cell Cycle/drug effects , Cell Survival/drug effects , Chloroquine/pharmacology , Colonic Neoplasms/metabolism , Drug Synergism , HCT116 Cells , HT29 Cells , Humans , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects , Transfection
13.
Cancer Cell Int ; 15: 74, 2015.
Article in English | MEDLINE | ID: mdl-26225121

ABSTRACT

BACKGROUND: Squamous cell carcinoma of the oral cavity (SCCOC) is the dominant origin of cancer associated mortality. Previous findings by our study reported that acquisition of anoikis resistance has a significant role in tumor progression of oral cavity. Several genes were over-expressed in anoikis-resistant cells under detached conditions which we confirmed earlier by microarray. Normal oral squamous epithelia grow adherent to a basement membrane, and when detached from the extracellular matrix, undergoes programmed cell death. The acquisition of anoikis-resistance is crucial phenomena in oral tumor advancement. In the current study, we have identified S100A7 expression as contributing factor for anoikis resistance and tumorigenicity in human oral cancer cells. Further, we have explored that elevated S100A7 expression in anoikis-sensitive oral keratinocytes and cancer cells reshape them more resistant to anoikis and apoptosis inducers via activation of cellular intrinsic and extrinsic avenue. METHODS: A subset of human cancer cell lines TU167, JMAR, JMARC39, JMARC42 and MDA-MB-468 were utilized for the generation of resistant stable cell lines. Further, immunohistochemistry, western blot and immunoprecipitation, assays of apoptosis, soft agar assay, orthotopic animal model and signaling elucidation were performed to establish our hypothesis. RESULTS: S100A7 gene is found to be responsible for anoikis resistance and tumorigenicity in human oral cancer cells. We have observed up-regulation of S100A7 in anoikis resistant cell lines, orthotropic model and patients samples with head and neck cancer. It is also noticed that secretion of S100A7 protein in conditioned medium by anoikis resistant head & neck cancer cell and in saliva of head and neck cancer patients. Up-regulation of S100A7 expression has triggered enhanced tumorigenicity and anchorage-independent growth of cancer cells through Akt phosphorylation leading to development of aniokis resistance in head and neck cancer cells. CONCLUSIONS: These data have led us to conclude that S100A7 is the major contributing factor in mediating anoikis-resistance of oral cancer cells and local tumor progression, and S100A7 might be useful as diagnostic marker for early detection of primary and recurrent squamous cell cancer.

14.
J Cell Physiol ; 230(3): 620-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25164250

ABSTRACT

Radiotherapy remains a prime approach to adjuvant therapies in patients with early and advanced breast cancer. In spite of therapeutic success, metastatic progression in patients undergoing therapy, limits its application. However, effective therapeutic strategies to understand the cellular and molecular machinery in inhibiting radiation-induced metastatic progression, which is poorly understood so far, need to be strengthened. Ionizing radiation was known to prompt cancer cell's metastatic ability by eliciting Transforming Growth Factor-beta (TGF-ß), a key regulator in epithelial-mesenchymal transdifferentiation and radio-resistance. In this viewpoint, we employed thymoquinone as a radiosensitizer to investigate its migration and invasion reversal abilities in irradiated breast cancer cell lines by assessing their respective attributes. The role of metastasis regulatory molecules like TGF-ß, E-cadherin, and integrin αV and its downstream molecules were determined using RT-PCR, western blotting, immunofluorescence, and extracellular TGF-ß levels affirmed through ELISA assays. These studies affirmed the TGF-ß restoring ability of thymoquinone in radiation-driven migration and invasion. Also, results demonstrated that the epithelial markers E-cadherin and cytokeratin 19 were downregulated whereas mesenchymal markers like integrin αV, MMP9, and MMP2 were upregulated by irradiation treatment; however thymoquinone pre-sensitization has reverted the expression of these proteins back to control proteins expression. Here, paclitaxel was chosen as an apoptosis inducer in TGF-ß restored cells and confirmed its cytotoxic effects in radiation alone and thymoquinone sensitized irradiated cells. We conclude that this therapeutic modality is effective in preventing radiation-induced epithelial-mesenchymal transdifferentiation and concomitant induction of apoptosis in breast cancer.


Subject(s)
Benzoquinones/administration & dosage , Breast Neoplasms/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Transforming Growth Factor beta/biosynthesis , Apoptosis/drug effects , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Cell Line, Tumor , Cell Transdifferentiation/drug effects , Female , Humans , Paclitaxel/administration & dosage , Radiation , Radiotherapy/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...