Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Mol Biol Rep ; 50(11): 9497-9509, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37731028

ABSTRACT

Oestrogen, the primary female sex hormone, plays a significant role in tumourigenesis. The major pathway for oestrogen is via binding to its receptor [oestrogen receptor (ERα or ß)], followed by nuclear translocation and transcriptional regulation of target genes. Almost 70% of breast tumours are ER + , and endocrine therapies with selective ER modulators (tamoxifen) have been successfully applied. As many as 25% of tamoxifen-treated patients experience disease relapse within 5 years upon completion of chemotherapy. In such cases, the ER-independent oestrogen actions provide a plausible explanation for the resistance, as well as expands the existing horizon of available drug targets. ER-independent oestrogen signalling occurs via one of the following pathways: signalling through membrane receptors, oxidative catabolism giving rise to genotoxic metabolites, effects on mitochondria and redox balance, and induction of inflammatory cytokines. The current review focuses on the non-classical oestrogen signalling, its role in cancer, and its clinical significance.


Subject(s)
Neoplasm Recurrence, Local , Receptors, Estrogen , Humans , Female , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Neoplasm Recurrence, Local/drug therapy , Estrogens/metabolism , Tamoxifen/pharmacology , Signal Transduction
2.
Ann Clin Biochem ; 60(5): 339-348, 2023 09.
Article in English | MEDLINE | ID: mdl-37158306

ABSTRACT

BACKGROUND: Several studies have demonstrated the analytical sensitivity of MALDI-TOF mass spectrometry (MALDI-TOF MS) by immunoenrichment for M-protein analysis. We report the results of a novel, low-cost, reagent-based extraction process using acetonitrile (ACN) precipitation to enrich for κ and λ light chains which can be analysed by MALDI-TOF MS. METHODS: Institutional Ethics committee approval was obtained. Serum samples from patients with monoclonal gammopathy of undetermined significance (MGUS), multiple myeloma (MM), plasmacytoma, AL amyloidosis and Waldenström macroglobulinemia (WM) underwent ACN precipitation. The images obtained were overlaid on apparently healthy donor serum samples to confirm the presence of M-protein. A sample was considered positive for M-protein if there was a sharp or broad peak within the κ or λ mass/charge (m/z) range: m/z- [M + 2H]2+: 11,550-12,300 Da and λ m/z- [M + 2H]2+: 11,100-11,500 Da. Images were acquired at a m/z range of 10,000-29,000 Da. Corresponding serum protein electrophoresis (SPEP), serum immunofixation electrophoresis (IFE) and serum free light chain (sFLC) assay by nephelometry were performed for all the samples. RESULTS: Two-hundred-and-two serum samples were included in the study: MM- 184 (91%); AL amyloidosis- 2 (1%); plasmacytoma- 8 (4%); MGUS- 6 (3%) and WM- 2 (1%). All the SPEP positive samples were identified by MALDI-TOF MS. Out of 179 samples positive for M-protein by IFE, MALDI-TOF MS was positive in 176 samples (98%). Compared to IFE, the sensitivity and specificity of M-protein identification by MALDI-TOF MS were 98.3% and 52.2%, respectively. CONCLUSIONS: This study demonstrates the feasibility of qualitatively identifying M-protein without the need for antibody-based immunoenrichment, making the technique cost-effective.


Subject(s)
Immunoglobulin Light-chain Amyloidosis , Multiple Myeloma , Paraproteinemias , Plasmacytoma , Humans , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Immunoglobulin Light Chains , Acetonitriles , Paraproteinemias/diagnosis
3.
Breast Cancer Res Treat ; 194(2): 207-220, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35597840

ABSTRACT

BACKGROUND: Therapeutic response predictors like age, nodal status, and tumor grade and markers, like ER/PR, HER2, and Ki67, are not reliable in predicting the response to a specific form of chemotherapy. The current study aims to identify and validate reliable markers that can predict pathological complete response (pCR) in fluorouracil, epirubicin, and cyclophosphamide (FEC)-based neoadjuvant therapy with (NACT/RT) and without concurrent radiation (NACT). MATERIALS AND METHODS: Tandem mass tag (TMT) quantitative liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to identify differentially expressed proteins from core needle breast biopsy between pCR (n = 4) and no-pCR (n = 4). Immunoblotting of shortlisted proteins with the tissue lysates confirmed the differential expression of the markers. Further, immunohistochemistry (IHC) was performed on formalin-fixed paraffin-embedded sections of treatment-naive core needle biopsies. In the NACT, 29 pCR and 130 no-pCR and in NACT/RT, 32 pCR and 71 no-pCR were used. RESULTS: 733 and 807 proteins were identified in NACT and NACT/RT groups, respectively. Ten proteins were shortlisted for validation as potential pCR-predictive markers. THBS1, TNC, and DCN were significantly overexpressed in no-pCR in both the groups. In NACT, CPA3 was significantly upregulated in the no-pCR. In NACT/RT, HnRNPAB was significantly upregulated and HMGB1 significantly downregulated in the no-pCR. HMGB1 was the only marker to show prognostic significance. CONCLUSION: Quantitative proteomics followed by IHC identified and validated potential biomarkers for predicting patient response to therapy. These markers can be used, following larger-scale validation, in combination with routine histological analysis providing vital indications of treatment response.


Subject(s)
Breast Neoplasms , HMGB1 Protein , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Chromatography, Liquid , Female , HMGB1 Protein/therapeutic use , Humans , Neoadjuvant Therapy , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Tandem Mass Spectrometry , Treatment Outcome
4.
Gene ; 815: 146137, 2022 Mar 20.
Article in English | MEDLINE | ID: mdl-35007686

ABSTRACT

The extracellular matrix (ECM) is composed of a mesh of proteins, proteoglycans, growth factors, and other secretory components. It constitutes the tumor microenvironment along with the endothelial cells, cancer-associated fibroblasts, adipocytes, and immune cells. The proteins of ECM can be functionally classified as adhesive proteins and matricellular proteins (MCP). In the tumor milieu, the ECM plays a major role in tumorigenesis and therapeutic resistance. The current review encompasses thrombospondins, osteonectin, osteopontin, tenascin C, periostin, the CCN family, laminin, biglycan, decorin, mimecan, and galectins. The matrix metalloproteinases (MMPs) are also discussed as they are an integral part of the ECM with versatile functions in the tumor stroma. In this review, the role of these proteins in tumor initiation, growth, invasion and metastasis have been highlighted, with emphasis on their contribution to tumor therapeutic resistance. Further, their potential as biomarkers and therapeutic targets based on existing evidence are discussed. Owing to the recent advancements in protein targeting, the possibility of agents to modulate MCPs in cancer as therapeutic options are discussed.


Subject(s)
Biomarkers, Tumor , Extracellular Matrix Proteins/physiology , Neoplasms/etiology , Neoplasms/therapy , Cell Adhesion Molecules/physiology , Extracellular Matrix Proteins/analysis , Humans , Matrix Metalloproteinases/physiology , Osteonectin/analysis , Osteonectin/physiology , Osteopontin/physiology , Tenascin/physiology , Thrombospondin 1/physiology , Treatment Outcome
5.
Am J Transl Res ; 13(11): 12181-12194, 2021.
Article in English | MEDLINE | ID: mdl-34956445

ABSTRACT

EWS-FLI1 is a master regulator of Ewing sarcoma (ES) oncogenesis. Although EWS-FLI1 represents a clear therapeutic target, targeted therapeutic inhibitors are lacking. Scientific literature has indicated accumulating information pertaining to EWS-FLI1 translocation, pathogenesis, function, oncogenic partnerships, and potential clinical relevance. However, attempts to develop EWS-FLI1-driven human-like ES mouse models or in vivo systems ended up with limited success. Establishing such models as preclinical screening tools may accelerate the development of EWS-FLI1 targeted therapeutic inhibitors. This review summarizes the current scenario, which focuses on the limitations, challenges, and possible reasons for past failures in model development and also plausible interim alternatives.

6.
J Biochem ; 169(6): 675-692, 2021 Sep 07.
Article in English | MEDLINE | ID: mdl-34492114

ABSTRACT

Mitochondrial ribosomal small subunit (MRPS) group of proteins is structural constituents of the small subunit of mitoribosomes involved in translation. Recent studies indicate role in tumourigenic process, however, unlike cytosolic ribosomal proteins, knowledge on the role of MRPS proteins in alternate cellular processes is very limited. Mapping protein-protein interactions (PPIs) onto known cellular processes can be a valuable tool to identify novel protein functions. In this study, to identify PPIs of MRPS proteins, we have constructed 31 glutathione-S-transferase (GST)/MRPS fusion clones. GST/MRPS fusion proteins were confirmed by MALDI-TOF analysis. GST pull-downs were performed using eight GST/MRPS proteins (MRPS9, MRPS10, MRPS11, MRPS18B, MRPS31, MRPS33, MRPS38 and MRPS39), GST alone as pull-down control and HEK293 cell lysate as the source for anchor proteins followed by nLC/MS/MS analysis and probable PPIs of eight MRPS proteins were identified. Three PPIs from GST pull-downs and interaction between six MRPS proteins and p53 previously reported in PPI database were validated. The PPI network analysis revealed putative role in cellular processes with implications for tumourigenesis. Gene expression screening of a cancer cell line panel indicated overexpression of MRPS10 and MRPS31 in breast cancer. Co-expression module identification tool analysis of breast cancer gene expression and MRPS10 and MRPS31 PPIs revealed putative role for PPI with acyl-CoA dehydrogenase in fatty acid oxidation process regulated by brain-derived neurotrophic factor signalling pathway.


Subject(s)
Breast Neoplasms/pathology , Mitochondrial Proteins/metabolism , Protein Interaction Maps , Recombinant Fusion Proteins/metabolism , Ribosomal Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Chromatography, Affinity , Female , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/isolation & purification , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Ribosomal Proteins/genetics , Ribosomal Proteins/isolation & purification , Tumor Cells, Cultured
7.
Naunyn Schmiedebergs Arch Pharmacol ; 394(2): 361-371, 2021 02.
Article in English | MEDLINE | ID: mdl-33015747

ABSTRACT

Osteosarcoma has been reported with treatment failure in up to 40% of cases. Our laboratory had identified genes involved in the PPARγ pathway to be associated with doxorubicin (DOX) resistance. We hence used PPARγ agonist pioglitazone (PIO) to modulate DOX resistance. DOX-resistant cell line (143B-DOX) was developed by gradient exposure to DOX. The cytotoxicity to PIO and in combination with DOX was assayed in vitro, followed by HPLC to estimate the metabolites of PIO in the presence of microsomes (HLMs). Gene expression studies revealed the mechanism behind the cytotoxicity of PIO. Further, the effects were evaluated in mice bearing 143B-DOX tumors treated either with PIO (20 mg/kg/p.o or 40 mg/kg/p.o Q1D) alone or in combination with DOX (0.5 mg/kg/i.p Q2W). 143B-DOX was 50-fold resistant over parental cells. While PIO did not show any activity on its own, the addition of HLMs to the cells in culture showed over 80% cell kill within 24 h, possibly due to the metabolites of PIO as determined by HPLC. In combination with DOX, PIO had shown synergistic activity. Additionally, cytotoxicity assay in the presence of HLMs revealed that PIO on its own showed promising activity compared to its metabolites-hydroxy pioglitazone and keto pioglitazone. In vivo studies demonstrated that treatment with 40 mg/kg/p.o PIO alone showed significant activity, followed by a combination with DOX. Gene expression studies revealed that PIO could modulate drug resistance by downregulating MDR1 and IL8. Our study suggests that PIO can modulate DOX resistance in osteosarcoma cells.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Bone Neoplasms/drug therapy , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Hypoglycemic Agents/therapeutic use , Osteosarcoma/drug therapy , Pioglitazone/therapeutic use , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Antibiotics, Antineoplastic/pharmacology , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Doxorubicin/pharmacology , Humans , Hypoglycemic Agents/pharmacology , Interleukin-8/genetics , Male , Mice, Nude , Osteosarcoma/genetics , Osteosarcoma/pathology , Pioglitazone/pharmacology , Xenograft Model Antitumor Assays
8.
J Proteomics ; 226: 103893, 2020 08 30.
Article in English | MEDLINE | ID: mdl-32634479

ABSTRACT

Diagnosis of Ovarian cancer (OC) has been a challenge, the purpose, therefore is to identify plasma proteins differentially expressed in epithelial ovarian cancer patients. Human plasma samples from patients with OC (n = 138), benign tumors (n = 20) and controls (n = 238) were used. Tandem Mass Tag (TMT) based quantitative analysis by high resolution mass spectrometry, was followed by validation using Quantibody array and ELISA techniques. 507 plasma proteins showed differential protein levels in OC plasma samples. 21 proteins were validated using Quantibody array. Further, nine proteins (CA125, CFD, CST3, ICAM1, IGFBP2, IGFBP3, SPP1, TSP1 and VEGFA) which showed significant differences in protein levels in Quantibody array analysis were validated using ELISA. In ELISA, the levels of CA125, IGFBP2, ICAM1 and SPP1 were significantly increased and levels of Adipsin and TSP1 were decreased in tumors compared to controls and benign group. Epithelial ovarian cancer diagnosis model combining five markers (CA125, IGFBP2, SPP1, TSP1 and ADI) showed 90.24% sensitivity and 94.87% specificity. In conclusion a panel of 5 plasma proteins has been found to be useful in distinguishing plasma samples from epithelial ovarian cancers from patients with benign tumors and healthy normal subjects. This has the potential as a diagnostic assay for epithelial ovarian cancer. SIGNIFICANCE: The significance of this case-control study is based on the large and well defined ovarian cancer patient population (epithelial ovarian cancers including serous and mucinous subtypes), age matched controls and benign ovarian tumors. This study incorporates a discovery phase involving quantitative proteomic analysis of immune-depleted plasma followed by two levels of validation studies involving a selected list of proteins using antibody arrays and ELISA. The validations were performed on an independent set of samples comprising of epithelial ovarian cancer subtypes, controls and benign tumors. The multiple marker combination comprising of Adipsin, CA125, IGFBP2, SPP1 and TSP1 identified in the study by ELISA could enable rapid translation to a larger screening study.


Subject(s)
Ovarian Neoplasms , Proteomics , Biomarkers, Tumor , Blood Proteins , Carcinoma, Ovarian Epithelial/diagnosis , Case-Control Studies , Female , Humans , Ovarian Neoplasms/diagnosis
9.
Gastric Cancer ; 23(5): 796-810, 2020 09.
Article in English | MEDLINE | ID: mdl-32333232

ABSTRACT

BACKGROUND: Phosphorylation is an important regulatory mechanism of protein activity in cells. Studies in various cancers have reported perturbations in kinases resulting in aberrant phosphorylation of oncoproteins and tumor suppressor proteins. METHODS: In this study, we carried out quantitative phosphoproteomic analysis of gastric cancer tissues and corresponding xenograft samples. Using these data, we employed bioinformatics analysis to identify aberrant signaling pathways. We further performed molecular inhibition and silencing of the upstream regulatory kinase in gastric cancer cell lines and validated its effect on cellular phenotype. Through an ex vivo technology utilizing patient tumor and blood sample, we sought to understand the therapeutic potential of the kinase by recreating the tumor microenvironment. RESULTS: Using mass spectrometry-based high-throughput analysis, we identified 1,344 phosphosites and 848 phosphoproteins, including differential phosphorylation of 177 proteins (fold change cut-off ≥ 1.5). Our data showed that a subset of differentially phosphorylated proteins belonged to splicing machinery. Pathway analysis highlighted Cdc2-like kinase (CLK1) as upstream kinase. Inhibition of CLK1 using TG003 and CLK1 siRNA resulted in a decreased cell viability, proliferation, invasion and migration as well as modulation in the phosphorylation of SRSF2. Ex vivo experiments which utilizes patient's own tumor and blood to recreate the tumor microenvironment validated the use of CLK1 as a potential target for gastric cancer treatment. CONCLUSIONS: Our data indicates that CLK1 plays a crucial role in the regulation of splicing process in gastric cancer and that CLK1 can act as a novel therapeutic target in gastric cancer.


Subject(s)
Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proteome/metabolism , Stomach Neoplasms/pathology , Animals , Apoptosis , Biomarkers, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, SCID , Neoplasm Invasiveness , Phosphorylation , Prognosis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Proteome/analysis , RNA, Small Interfering/genetics , Stomach Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Hum Antibodies ; 27(1): 69-83, 2019.
Article in English | MEDLINE | ID: mdl-30282350

ABSTRACT

BACKGROUND AND OBJECTIVE: CD99/MIC2 gene product is a heavily glycosylated transmembrane protein which plays a major role in homotypic cell adhesion, apoptosis of double positive T cells and vesicular protein trafficking. It is over expressed in various cancers and has been considered as an ideal therapeutic target. The present study focused at developing monoclonal antibodies against the extracellular domain (ECD) of CD99 using hybridoma technology. MATERIALS AND METHODS: In order to generate monoclonal antibodies, the recombinant ECD of CD99 was used for immunizing the mice. Resulting hybridomas were screened through indirect ELISA. Clones which gave high absorbance values were sub cloned by limiting dilution followed by isotype determination, IP, WB and FACS. The monoclonal antibody 547F2 4F12 was purified from culture supernatant using FPLC and further screened using IF. Finally, the antibodies were validated for specificity using siRNA knock-down. RESULTS: We were able to establish stable hybridoma clones secreting CD99 antibodies. The antibodies reacted with both the recombinant ECD as well as the wild type CD99 and their isotype's were determined as IgM. CONCLUSION: Based on these results, we propose that the purified monoclonal antibody 547F2 4F12 could be possibly used for targeting tumors which over express CD99.


Subject(s)
12E7 Antigen/immunology , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Extracellular Matrix/immunology , Vaccines, Synthetic/immunology , Animals , Antibodies, Monoclonal/isolation & purification , Antibody Specificity , Cloning, Molecular , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Gene Knockdown Techniques , Humans , Hybridomas , Mice , Mice, Inbred BALB C , Molecular Targeted Therapy , RNA, Small Interfering/pharmacology , Recombinant Proteins/immunology
11.
Comput Biol Chem ; 77: 36-43, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30219714

ABSTRACT

Synovial sarcoma (SS) is characterized by a tumour specific chromosomal translocation t(X;18) (p11;q11) which results in the formation of SYT-SSX1 fusion protein. This fusion protein represents a clear therapeutic target and molecules specifically targeting SYT-SSX1 fusion protein are currently not available. In this study, SYT-SSX1 fusion protein sequence was retrieved from Uniprot and 3D structure was generated using I-TASSER modeling program. A structure based computational screening approach has been employed using Glide docking software to identify potential SYT-SSX1 small molecule inhibitors that bind to the junction region of the fusion protein. The obtained inhibitors were further filtered based on the docking score and ADME/T properties. Ten best fit compounds were chosen for in vitro studies. The anti-proliferative activities of these 10 compounds were screened in Yamato, ASKA (carries SYT-SSX1 fusion protein) and other sarcoma cell lines such as A673, 143B to understand the specificity of inhibition of the chosen compounds. The in vitro activity was compared against HEK293 cell lines. The compound 5-fluoro-3-(1-phenyl-1H-tetraazol-5-yl)-1H-indole (FPTI) was found to be selectively cytotoxic in synovial sarcoma cell lines (Yamato and ASKA) and this compound also showed insignificant anti proliferative activity on other cell lines. Further, target gene expression study confirmed that FPTI treatment down-regulated SYT-SSX1 and modulated its downstream target genes. Cell cycle analysis revealed the involvement of an apoptotic mechanism of cell death. Further experimental validations may elucidate the therapeutic potentials of FPTI against SYT-SSX1 fusion protein.


Subject(s)
Oncogene Proteins, Fusion/antagonists & inhibitors , Sarcoma, Synovial/drug therapy , Small Molecule Libraries/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Computer Simulation , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Models, Molecular , Oncogene Proteins, Fusion/chemistry , Sarcoma, Synovial/pathology , Small Molecule Libraries/chemistry
12.
J Chromatogr Sci ; 56(6): 488-497, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-29608650

ABSTRACT

A combination of 5-fluorouracil (FU), epirubicin (EP) and cyclophosphamide (CP) is routinely employed in the treatment of breast cancer. The objective of this study was to develop a reverse phase high-performance liquid chromatography (HPLC)-UV method for simultaneous quantitative analysis of the triple-drug and their metabolites in plasma. RP-HPLC system with a C18 column and a diode array detector was employed. The plasma samples were precipitated with acetonitrile and the supernatant was dried under a flow of nitrogen gas. The mobile phase comprised of two combinations, water (pH 4.0) and methanol (98:2 v/v), and water (pH 4.0):methanol:acetonitrile (70:13:17 v/v/v). The retention times for the compounds were determined and the parameters of validation established in plasma indicated the robustness and reliability. The corresponding HPLC peaks were confirmed using electron spray ionization mass spectrometry. FU and metabolites had a recovery of >93%; EP, epirubicinol and CP were >78% from plasma. Stability at 28-30°C in water (pH 4.0) of FU, 5,6-dihydro-5-fluorouracil and EP were higher followed by CP, EPol, fluorodeoxyuridine and fluorouridine (FUR). Storage of the drug-spiked plasma at -80°C assessed for 72 h showed a small but significant (P < 0.05) change in the recovery of FUR and EP. The method was validated in patient's plasma samples (n = 6).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/blood , Chromatography, High Pressure Liquid/methods , Chromatography, Reverse-Phase/methods , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/metabolism , Cyclophosphamide/blood , Cyclophosphamide/chemistry , Cyclophosphamide/metabolism , Drug Stability , Epirubicin/blood , Epirubicin/chemistry , Epirubicin/metabolism , Fluorouracil/blood , Fluorouracil/chemistry , Fluorouracil/metabolism , Humans , Limit of Detection , Linear Models , Reproducibility of Results , Spectrophotometry, Ultraviolet
13.
Sci Rep ; 7(1): 7172, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28775288

ABSTRACT

The EWS-FLI1 chimeric protein uniquely expressed in Ewing's sarcoma has an obligate role in its aetiology. In our previous report we showed that ectopic expression of the DNA sequences form the junction region (a.a 251-280) can inhibit Ewing's sarcoma cell growth. In the present report, we introduced a peptide (TAT/NLS/EWS-PEP) comprising of thirty amino acids spanning the junction in conjunction with HIV-1-trans-activating (TAT) and nuclear localization signal sequence (NLS). Peptide uptake and localization studies revealed presence of peptide in ~99% of transduced cells and in the nucleus. Peptide transfection induced cytotoxicity relative to untreated and TAT-NLS peptide treated Ewing's sarcoma cells. The peptide inhibited clonogenicity, cell cycle, bromo-deoxy uridine (BrdU) uptake and invasion capacity of treated cells. The treatment also affected epithelial to mesenchymal transition (EMT) markers and EWS-FLI1 target gene expression levels. Co-immunoprecipitation experiments involving ectopically expressed full-length EWS-FLI1 protein and the peptide revealed an interaction. Additionally, we found that peptide interaction also occurs with the protein-GGAA microsatellite sequences complex known to contain EWS-FLI1. Further, in the pull-down assay, the peptide was found to interact with proteins known to potentially interact with EWS-FLI1. Based on these results we conclude that peptide could be applied in targeting EWS-FLI1 protein.


Subject(s)
Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics , Cell Cycle , Cell Line , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/metabolism , Peptides/genetics , Peptides/metabolism , Proto-Oncogene Protein c-fli-1/chemistry , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/chemistry , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/metabolism , Trans-Activators , Transfection
14.
Gene ; 589(1): 27-35, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27170550

ABSTRACT

Mitochondria are prominently understood as power houses producing ATP the primary energy currency of the cell. However, mitochondria are also known to play an important role in apoptosis and autophagy, and mitochondrial dysregulation can lead to pathological outcomes. Mitochondria are known to contain 1500 proteins of which only 13 are coded by mitochondrial DNA and the rest are coded by nuclear genes. Protein synthesis in mitochondria involves mitochondrial ribosomes which are 55-60S particles and are composed of small 28S and large 39S subunits. A feature of mammalian mitoribosome which differentiate it from bacterial ribosomes is the increased protein content. The human mitochondrial ribosomal protein (MRP) gene family comprises of 30 genes which code for mitochondrial ribosomal small subunit and 50 genes for the large subunit. The present review focuses on the mitochondrial ribosomal small subunit genes (MRPS), presents an overview of the literature and data gleaned from publicly available gene and protein expression databases. The survey revealed aberrations in MRPS gene expression patterns in varied human diseases indicating a putative role in their etiology.


Subject(s)
Mitochondria/genetics , Mitochondrial Proteins/genetics , Ribosomal Proteins/genetics , Ribosome Subunits, Small, Eukaryotic/genetics , Animals , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Gene Expression Regulation , Humans , Mammals , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Ribosome Subunits, Small, Eukaryotic/chemistry , Ribosome Subunits, Small, Eukaryotic/metabolism , Signal Transduction , Spondylitis, Ankylosing/genetics , Spondylitis, Ankylosing/metabolism , Spondylitis, Ankylosing/pathology
15.
Biomarkers ; 20(5): 292-8, 2015.
Article in English | MEDLINE | ID: mdl-26329525

ABSTRACT

OBJECTIVE: To differentiate plasma from ovarian cancer and healthy individuals using MALDI-TOF mass spectroscopy. MATERIALS AND METHODS: MALDI-TOF was used to generate profiles of immuno-depleted plasma samples (89 cancers and 199 healthy individuals) that were fractionated using three types of magnetic beads (HIC8, WCX and IMAC-Cu). RESULTS: Differentially expressed mass ranges showing >1.5-2-fold change in expression from HIC8 (30), WCX (12) and IMAC-Cu (6) fractions were identified. Cross validation and recognition capability scores for the models indicated discrimination between the classes. CONCLUSIONS: Spectral profiles can differentiate plasma samples of ovarian cancer patients from healthy individuals.


Subject(s)
Adenocarcinoma/blood , Biomarkers, Tumor/blood , Ovarian Neoplasms/blood , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Blood Proteins/analysis , Electrophoresis, Polyacrylamide Gel , Female , Humans , Molecular Weight
16.
Cell Oncol (Dordr) ; 37(6): 409-19, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25338303

ABSTRACT

PURPOSE: We and others show that SOSTDC1 is down-regulated in breast cancer tissues compared to matched normal tissues. Previously, we found that epigenetic mechanisms underlie the down-regulation of SOSTDC1 in gastric cancer cells. The aim of this study was to assess the putative epigenetic regulation of SOSTDC1 expression in breast cancer cells. METHODS: Microarray-based expression profiling was performed in a series of primary breast cancers and matched normal tissues. Real-time PCR was performed to assess SOSTDC1 and E4BP4 mRNA levels in MCF7, BT549, MBMDA231, T47D (breast cancer) and HEK293T (normal kidney) cell lines. Methylation-specific PCR (MSP) and bisulfite sequencing PCR (BSP) were performed to assess the methylation level of the SOSTDC1 gene promoter, and 5-Aza 2-deoxycytidine (5'-Aza-dC) treatment was used to induce its demethylation. A luciferase assay was used to measure SOSTDC1 promoter activity in vitro. Stable shRNA-mediated knockdown of E4BP4 was carried out in MCF7 cells and confirmed by Western blotting. Finally, MCF7 cell proliferation and survival were measured by MTS assay. RESULTS: We found that SOSTDC1 is frequently down-regulated in primary breast cancers (98.2%) and in all breast cancer cell lines tested. MSP and BSP analyses revealed SOSTC1 promoter hypermethylation at CpG sites. 5'-Aza-dC treatment induced a striking down-regulation of SOSTDC1 gene expression, whereas BSP analysis showed demethylation of its promoter. Subsequent in silico SOSTDC1 promoter analysis indicated the presence of putative transcriptional repressor E4BP4 binding sites, and promoter deletion studies indeed revealed repressor binding regions encompassing these E4BP4 binding sites. Relative quantification of E4BP4 expression showed an inverse correlation to SOSTDC1 expression in the breast cancer cell lines tested. Exogenous over-expression of E4BP4 in HEK-293 and BT549 cells reduced SOSTDC1 expression and its promoter activity, respectively. Stable shRNA-mediated E4BP4 BT549 and MCF7 knock-down cells treated with 5'-Aza-dC exhibited up-regulation of SOSTDC1 expression and a concomitant inhibition of cell proliferation and survival. CONCLUSION: From our results we conclude that the transcriptional repressor E4BP4 plays a role in repressing epigenetically regulated SOSTDC1 expression in breast cancer cells, which can be reverted by E4BP4 silencing.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Breast Neoplasms/metabolism , Epigenesis, Genetic/physiology , Proteins/metabolism , Adaptor Proteins, Signal Transducing , Basic-Leucine Zipper Transcription Factors/genetics , Breast Neoplasms/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , MCF-7 Cells , Proteins/genetics
17.
Int J Radiat Biol ; 88(9): 629-34, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22694363

ABSTRACT

PURPOSE: To find the radiation sensitivity of human cervical carcinoma cell lines and to investigate the effect of the dominant negative-Ubiquitin-conjugating enzyme E2C (DN-UBE2C) on cell proliferation and radiation response. MATERIALS AND METHODS: Radiation sensitivities of human cervical cell lines (SiHa, HeLa, BU25TK, ME 180, and C33A) were analyzed by assessing their cell survival after irradiation by MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) assay. Soft agar cloning assay, growth curve and radiation response of DN-UBE2C stably transfected SiHa and HeLa cell lines were assessed by MTS assay and Clonogenic assay. RESULTS: Difference in sensitivity to radiation was observed among the cervical cancer cell lines studied. SiHa was found to be the most resistant cell line whereas C33A cells were the most sensitive. The growth rate of SiHa and HeLa transfected with DN-UBE2C was significantly reduced compared to vector control. Furthermore, DN-UBE2C-mediated radiosensitivity was correlated with a significant decrease in resistance to radiation by SiHa and HeLa cells after transfection with the DN-UBE2C when compared to control cultures. CONCLUSION: These results suggested that the Ubiquitin-conjugating enzyme E2C (UBE2C) gene is a potential therapeutic target for cervical cancer treatment.


Subject(s)
Ubiquitin-Conjugating Enzymes/physiology , Uterine Cervical Neoplasms/radiotherapy , Cell Line, Tumor , Cell Proliferation/radiation effects , Female , Humans , Radiation Tolerance , Uterine Cervical Neoplasms/enzymology
18.
Genes Chromosomes Cancer ; 49(11): 981-90, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20665731

ABSTRACT

Glioblastoma is the most frequent and malignant brain tumor, characterized by an elevated capacity for cellular proliferation and invasion. Recently, it was demonstrated that podoplanin membrane sialo-glycoprotein encoded by PDPN gene is over-expressed and related to cellular invasion in astrocytic tumors; however the mechanisms of regulation are still unknown. MicroRNAs are noncoding RNAs that regulate gene expression and several biological processes and diseases, including cancer. Nevertheless, their roles in invasion, proliferation, and apoptosis of glioblastoma are not completely understood. In this study, we focused on miR-29b and miR-125a, which were predicted to regulate PDPN, and demonstrated that these microRNAs directly target the 3' untranslated region of PDPN and inhibit invasion, apoptosis, and proliferation of glioblastomas. Furthermore, we report that miR-29b and miR-125a are downregulated in glioblastomas and also in CD133-positive cells. Taken together, these results suggest that miR-29b and miR-125a represent potential therapeutic targets in glioblastoma.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation/physiology , Glioblastoma/genetics , Membrane Glycoproteins/genetics , MicroRNAs/physiology , Neoplasm Invasiveness/prevention & control , Apoptosis , Blotting, Western , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Glioblastoma/pathology , Humans , Reverse Transcriptase Polymerase Chain Reaction
19.
Cancer Res ; 69(4): 1527-35, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19190346

ABSTRACT

Defects in apoptotic pathway contribute to uncontrolled proliferation of cancer cells and confer resistance to chemotherapy. Growth arrest and DNA damage inducible, alpha (GADD45alpha) is up-regulated on docetaxel treatment and may contribute to docetaxel-mediated cytotoxicity. We examined the mechanism of regulation of GADD45alpha in prostate cancer cells and the effect of its up-regulation on sensitivity to docetaxel chemotherapy. Expression of GADD45alpha in PC3 cells was higher than that in Du145 and LNCaP cells (17- and 12-fold, respectively; P < 0.05). Although the proximal promoter region was unmethylated in all three cell lines, methylation of a 4 CpG region upstream of the proximal promoter correlated inversely with gene expression levels. Methylation was reversed by treatment of Du145 and LNCaP cells with DNA methyltransferase inhibitors, leading to reactivation of GADD45alpha expression in these cells. The 5' 4 CpG region was also frequently methylated in prostate cancer tissues. Methylation of this region correlated inversely with gene expression in prostate cancer and benign prostate tissues. The methyl binding protein MeCP2 was associated with the methylated 4 CpGs in Du145 cells, and knockdown of MeCP2 in these cells (Du145 MeCP2(-)) led to a significantly increased expression of GADD45alpha (3-fold; P = 0.035) without affecting the methylation status of the gene. Enhanced sensitivity to docetaxel was observed by up-regulation of GADD45alpha in Du145 cells by recombinant expression of GADD45alpha or pretreatment with 5-azacytidine. Our results show that GADD45alpha is epigenetically repressed and is a potential target for treatment of prostate cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Cycle Proteins/genetics , Nuclear Proteins/genetics , Prostatic Neoplasms/pathology , Taxoids/therapeutic use , Azacitidine/therapeutic use , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , DNA Methylation , DNA Primers , DNA, Neoplasm/genetics , Docetaxel , Gene Expression Regulation, Neoplastic , Genome , Humans , Male , Methylation , Nuclear Proteins/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
20.
Blood ; 113(16): 3744-53, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-19096009

ABSTRACT

Chronic lymphocytic leukemia (CLL) is characterized by cells that exhibit dysfunctional apoptosis. Here, we show that deacetylase inhibition led to the E2F1- and myc-mediated transcriptional activation of the microRNA miR106b in primary CLL cells. Induction of miR106b was associated with a down-regulation in the levels of the E3-ubiquitin ligase Itch. Decreases in Itch protein levels were associated with a reciprocal accumulation of its proapoptotic substrate, TAp73 (p73), and induction of p53 up-regulated modulator of apoptosis (PUMA) mRNA and protein. This event was accompanied by mitochondrial dysfunction, processing of caspase-9, and apoptosis of CLL cells. Ectopic expression of miR106b in CLL cells demonstrated that Itch was a direct target of miR106b such that miR106b-induced decreases in Itch resulted in an accumulation of p73. Thus, our results identify a novel regulatory mechanism wherein microRNA regulate cell survival by mediating the posttranscriptional down-regulation of an ubiquitin ligase, leading to the induction of a proapoptotic regulator in malignant cells. Silencing of miRNA expression in CLL may selectively suppress proapoptotic pathways, providing such tumors with a survival advantage. Consequently, chemotherapeutic drugs that activate miR106b could initiate a p53-independent mechanism that targets CLL cells.


Subject(s)
Apoptosis , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , MicroRNAs/metabolism , Nuclear Proteins/metabolism , RNA, Neoplasm/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Survival/genetics , DNA-Binding Proteins/genetics , Female , Gene Silencing , HeLa Cells , Humans , K562 Cells , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Male , MicroRNAs/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , Repressor Proteins/genetics , Tumor Protein p73 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...