Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Drug Dev Res ; 83(2): 525-543, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34569640

RESUMO

New candidates of imidazo[1,2-a]pyridine were designed by combining 2-amino pyridine, TOSMIC and various assorted aldehydes to explore their antioxidant and antifungal potential. The design of these derivatives was based on utilizing the antifungal potential of azoles and TOSMIC moiety. These derivatives were synthesized by adopting multi-component reaction methodology, as it serves as a rapid and efficient tool to target structurally diverse heterocyclic compounds in quantitative yield. The resulting imidazo[1,2-a]pyridine derivatives were structurally verified by 1 HNMR, 13 CNMR, HRMS, and HPLC. The compounds were analyzed for their antioxidant and fluorescent properties and it was observed that compound 15 depicted highest potential. The compounds were evaluated for their antifungal potential to highlight their medical application in the area of Invasive Fungal Infections (IFI). Compound 12 gave the highest antifungal inhibition against Aspergillus fumigatus 3007 and Candida albicans 3018. To elucidate the antifungal mechanism, confocal images of treated fungi were analyzed, which depicted porous nature of fungal membrane. Estimation of fungal membrane sterols by UPLC indicated decrease in ergosterol component of fungal membrane. In silico studies further corroborated with the in vitro results as docking studies depicted interaction of synthesized heterocyclic compounds with amino acids present in the active site of target enzyme (lanosterol 14 alpha demethylase). Absorption, distribution, metabolism, and excretion (ADME) analysis was indicative of drug-likeliness of the synthesized compounds.


Assuntos
Antifúngicos , Antioxidantes , Antifúngicos/química , Antifúngicos/farmacologia , Cianetos , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Piridinas/farmacologia
2.
Cell Physiol Biochem ; 54(4): 748-766, 2020 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-32809300

RESUMO

BACKGROUND/AIMS: The hypoxic microenvironment in NSCLC has been widely accepted as a contributor to both therapeutic resistance and tumor progression. In this study, we have explored Allicin, a key organosulfur compound present in garlic for its previously unreported effectiveness in the heterogeneous hypoxic tumor microenvironment of NSCLC. METHODS: The effect of Allicin on the viability of NSCLC cells was determined by MTT assay. To determine the migration rate of treated cells compared to the control, scratch and transwell migration assays were performed. Flowcytometry was done to explore cell cycle distribution, apoptosis and ROS production in cells. Fluorescence microscopy was used to examine autophagy and DNA damage in cells. Dot blot was done to check genome wide methylation. RNA expression was detected by RT-PCR and protein expression by western blotting. RESULTS: Allicin significantly decreases cell viability, proliferation and migration of NSCLC cells in both normoxia and hypoxia. It elicits both apoptosis and autophagy pathway in A549 cells by ROS accumulation and facilitating S/G2-M phase arrest in both normoxia as well as hypoxia. We suggest that ROS/MAPK and ROS/JNK signaling pathway together govern the cytotoxic effect of allicin in NSCLC cells. Notably, allicin suppresses the expression of HIF-1α and HIF-2α in hypoxic cells, pointing towards a mechanism of its effectiveness in hypoxia. A long term passive demethylation was observed, with decreased mC and no change in TET expression, thereby ruling out active demethylation by allicin. Furthermore, allicin synergistically enhances growth inhibitory activity of low dose cisplatin to effectively overcome hypoxia induced cisplatin resistance in A549 cells. CONCLUSION: Altogether, our results elucidate a potential use of allicin in sensitizing hypoxic and chemoresistant NSCLC to cisplatin-based chemotherapy and provide new, affordable therapeutic strategy with reduced side effects.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Hipóxia Celular/efeitos dos fármacos , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias Pulmonares/tratamento farmacológico , Ácidos Sulfínicos/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dissulfetos , Sinergismo Farmacológico , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases/genética , Metilação , Espécies Reativas de Oxigênio/metabolismo , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos
3.
Stem Cells ; 35(6): 1468-1478, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28376560

RESUMO

Activation of pluripotency regulatory circuit is an important event in solid tumor progression and the hypoxic microenvironment is known to enhance the stemness feature of some cells. The distinct population of cancer stem cells (CSCs)/tumor initiating cells exist in a niche and augment invasion, metastasis, and drug resistance. Previously, studies have reported global hypomethylation and site-specific aberrant methylation in gliomas along with other epigenetic modifications as important contributors to genomic instability during glioma progression. Here, we have demonstrated the role of hypoxia-mediated epigenetic modifications in regulating expression of core pluripotency factors, OCT4 and NANOG, in glioma cells. We observe hypoxia-mediated induction of demethylases, ten-eleven-translocation (TET) 1 and 3, but not TET2 in our cell-line model. Immunoprecipitation studies reveal active demethylation and direct binding of TET1 and 3 at the Oct4 and Nanog regulatory regions. Tet1 and 3 silencing assays further confirmed induction of the pluripotency pathway involving Oct4, Nanog, and Stat3, by these paralogues, although with varying degrees. Knockdown of Tet1 and Tet3 inhibited the formation of neurospheres in hypoxic conditions. We observed independent roles of TET1 and TET3 in differentially regulating pluripotency and differentiation associated genes in hypoxia. Overall, this study demonstrates an active demethylation in hypoxia by TET1 and 3 as a mechanism of Oct4 and Nanog overexpression thus contributing to the formation of CSCs in gliomas. Stem Cells 2017;35:1468-1478.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Epigênese Genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Diferenciação Celular/genética , Hipóxia Celular/genética , Linhagem Celular Tumoral , Desmetilação do DNA , Dioxigenases/metabolismo , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Loci Gênicos , Glioma/patologia , Humanos , Oxigenases de Função Mista/metabolismo , Modelos Biológicos , Células-Tronco Pluripotentes/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Esferoides Celulares/metabolismo
4.
Anal Biochem ; 557: 111-119, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30048629

RESUMO

A label-free electrochemical immunosensor is fabricated using high quality single-walled carbon nanotube for early detection of leukemia cells. It is based on P-glycoprotein (P-gp) expression level detection; by effective surface immune-complex formation with the monoclonal anti-P-glycoprotein antibodies bound to an epoxy modified nanotube surface. The expression level of P-gp on the leukemia cell surface detected by cyclic voltammetry is in good agreement with immunofluorescence microscopy studies. The proposed biosensor could be used for the detection of P-gp expressing cells within a linear range of 1.5 × 103 cells/mL - 1.5 × 107 cells/mL where lowest detection limit is found to be 19 cells/mL. A calibration plot of peak current v/s the logarithm of concentration of leukemia K562 cells is found linear with a regression coefficient of 0.935. This strategy promises high sensitivity, low-cost, fast, and repeatable recognition of cancer cells. The immunosensor was stable for three weeks and showed good precision with the relative standard deviation (RSD) of 3.57% and 2.12% assayed at the cell concentrations of 1.5 × 103 and 1.5 × 105 cells mL-1 respectively. The proposed single-wall carbon nanotube based immunosensor showed better analytical performance in comparison to similar leukemia electrochemical sensors reported.


Assuntos
Técnicas Biossensoriais/métodos , Técnicas Eletroquímicas/instrumentação , Técnicas Eletroquímicas/métodos , Imunoensaio/instrumentação , Imunoensaio/métodos , Leucemia/diagnóstico , Nanotubos de Carbono/química , Técnicas Biossensoriais/instrumentação , Humanos , Células K562
5.
Biochim Biophys Acta ; 1840(1): 350-6, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24041991

RESUMO

BACKGROUND: Allicin has received much attention due to its anti-proliferative activity and not-well elucidated underlying mechanism of action. This work focuses towards determining the cellular toxicity of allicin and understanding its interaction with nucleic acid at molecular level. METHODS: MTT assay was used to assess the cell viability of A549 lung cancer cells against allicin. Fourier transform infrared (FTIR) and UV-visible spectroscopy were used to study the binding parameters of nucleic acid-allicin interaction. RESULTS: Allicin inhibits the proliferation of cancer cells in a concentration dependent manner. FTIR spectroscopy exhibited that allicin binds preferentially to minor groove of DNA via thymine base. Analysis of tRNA allicin complex has also revealed that allicin binds primarily through nitrogenous bases. Some amount of external binding with phosphate backbone was also observed for both DNA and RNA. UV visible spectra of both DNA allicin and RNA allicin complexes showed hypochromic shift with an estimated binding constant of 1.2×10(4)M(-1) for DNA and 1.06×10(3)M(-1)for RNA binding. No major transition from the B-form of DNA and A-form of RNA is observed after their interaction with allicin. CONCLUSIONS: The results demonstrated that allicin treatment inhibited the proliferation of A549 cells in a dose-dependent manner. Biophysical outcomes are suggestive of base binding and helix contraction of nucleic acid structure upon binding with allicin. GENERAL SIGNIFICANCE: The results describe cytotoxic potential of allicin and its binding properties with cellular nucleic acid, which could be helpful in deciphering the complete mechanism of cell death exerted by allicin.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/patologia , DNA/metabolismo , Neoplasias Pulmonares/patologia , Ácidos Sulfínicos/farmacologia , Antioxidantes/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Sobrevivência Celular/efeitos dos fármacos , Dissulfetos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Espectrofotometria Ultravioleta , Espectroscopia de Infravermelho com Transformada de Fourier , Células Tumorais Cultivadas
6.
Cell Cycle ; 22(9): 1135-1153, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36945177

RESUMO

Gliomas are brain tumors associated with high morbidity, relapse and lethality despite improvement in therapeutic regimes. The hypoxic tumor microenvironment is a key feature associated with such poor outcomes in gliomas. The Hypoxia Inducible Factor (HIF) family of transcription factors are master regulators of cellular proliferation, high metabolic rates and angiogenesis via aberrant expression of downstream genes. Recent studies have implicated long non-coding RNAs (lncRNAs) as potential prognostic and diagnostic biomarkers. In this study, identification of hypoxia regulated lncRNA with a bioinformatic pipeline consisting of a newly developed tool "GenOx" was utilized for the identification of Hypoxia Response Element (HRE) and Hypoxia Ancillary Sequence (HAS) motifs in the promoter regions of lncRNAs. This was coupled with molecular, functional and interactome-based analyses of these hypoxia-relevant lncRNAs in primary tumors and cell-line models. We report on the identification of novel hypoxia regulated lncRNAs SNHG12, CASC7 and MF12-AS1. A strong association of RNA splicing mechanisms was observed with enriched lncRNAs. Several lncRNAs have emerged as prognostic biomarkers, of which TP53TG1 and SNHG1 were identified as highly relevant lncRNAs in glioma progression and validated in hypoxia cultured cells. Significantly, we determined that SNHG1 expression in tumor (vs. normal) is different from glioma stem cells, GSC (vs. tumors) and in hypoxia (vs. normoxia), positioning downregulation of SNHG1 to be associated with worsened prognosis.


Assuntos
Glioma , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , Regulação para Baixo , Glioma/patologia , Hipóxia/genética , Biomarcadores , Regulação Neoplásica da Expressão Gênica , Microambiente Tumoral
7.
Biochim Biophys Acta Mol Cell Res ; 1870(2): 119302, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35649481

RESUMO

The question of how lung cancer progresses in never-smokers remains largely unanswered. In our analysis of data from 1727 lung cancer patients, we observed a difference of only 47 days in the overall survival between lung adenocarcinoma patients who were smokers vis-a-vis never-smokers - the disease has a poor prognosis irrespective of the smoking status, or gender. We have investigated the possible collaboration between the nAChR and hypoxia signaling pathway to explicate a mechanism of disease progression in never-smokers using patient-derived tumor cells. We found a previously unidentified increase in both acetylcholine and nAChR-α7 levels in non-small cell lung cancer cells in hypoxia. A similar increase in ubiquitously expressed nAChR-α7 transcripts was also observed in other cancer lines and primary tumor tissues. A direct binding of HIF-1α with the hypoxia-response element (HRE) present at -48 position preceding the transcriptional start site in nAChR-α7 promoter region was established. Crucially, the increased acetylcholine levels in hypoxia drove a feedback loop via modulation of PI3K/AKT pathway to stabilize HIF-1α in hypoxia. Further, hypoxia-mediated metastasis and induction of HIF-1α in these cells was significantly reversed by bungarotoxin, an antagonist of nAChR-α7. The nAChR-AKT-HIF network needs to be further investigated to conclusively prove its mechanism and to explore its therapeutic potential. Our study gives a plausible explanation for the equally worse prognosis of lung adenocarcinoma in never-smokers wherein the nAChR signaling is enhanced in hypoxia by acetylcholine in the absence of nicotine.


Assuntos
Adenocarcinoma de Pulmão , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Receptores Nicotínicos , Humanos , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Nicotina/farmacologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fumantes , Acetilcolina , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Linhagem Celular Tumoral , Adenocarcinoma de Pulmão/genética , Transdução de Sinais
8.
Asian Pac J Cancer Prev ; 23(10): 3553-3561, 2022 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-36308382

RESUMO

OBJECTIVE: Receptor tyrosine kinases (RTK) are relevant therapeutic targets in the treatment of lung cancer. Germline susceptibility variants that influence these RTKs may provide new insights into their regulation.  rs9387478 is located in the genomic interval between two RTK-genes ROS1/DCBLD1, of which ROS1 alterations are implicated in lung carcinogenesis and treatment response while the latter remains poorly understood. MATERIALS AND METHODS: Venous blood was drawn from 100 control and 231 case subjects. Genotype was scored by restriction fragment length polymorphism (RFLP), PCR amplification followed by HindIII digestion. Logistic regression was applied to compare the association between variables. Survival curve was plotted to draw a correlation between the genotype and overall survival. Also, eQTL and chromatin state changes were analyzed and correlated with the survival of patients using available datasets. RESULTS: In our population smoking correlated significantly with lung cancer [OR= 2.607] with the presence of the minor allele 'A' enhancing the nicotine dependence [CA (OR=3.23)]. Individuals with homozygous risk allele 'A' had a higher chance of developing lung cancer [OR=2.65] than individuals with CA/CC implying a recessive model of association. Patients with CC/CA genotype had better overall survival than patients with AA genotype [161 days/142 days vs 54 days, p=0.005]. The homozygous risk allele was significantly associated with increased DCBLD1 and ROS1 expression in lung cancer, with enriched active histone marks due to the polymorphism. Interestingly, increased DCBLD1 expression was associated with poor outcomes in lung cancer. CONCLUSION: Overall, our study provides strong evidence that rs9387478 is significantly associated with both nicotine dependence and lung cancer in our North Indian cohort. The association of the SNP with prognostic genes, DCBLD1 and ROS1 make rs9387478 a promising prognostic marker in the North Indian population. The results obtained are significant, however, the study needs to be performed in a larger sample size.


Assuntos
Neoplasias Pulmonares , Tabagismo , Humanos , Estudos de Casos e Controles , Predisposição Genética para Doença , Genótipo , Pulmão , Neoplasias Pulmonares/genética , Polimorfismo de Nucleotídeo Único , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/genética
9.
3 Biotech ; 11(10): 451, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34631352

RESUMO

Investigating the therapeutic and prognostic potential of genes in the heterogeneous hypoxic niche of glioblastoma. We have analyzed RNA expression of U87MG cells cultured in hypoxia compared to normoxia. Common differentially expressed genes (DEGs) from GSE45301 and GSE18494 and their functional enrichment was performed using MetaScape and PANTHER. Hub genes and their ontology were identified using MCode cytoHubba and ClueGO and validated with GlioVis, Oncomine, HPA and PrognoScan. Using the GEO2R analysis of GSE45301 and GSE18494 datasets, we have found a total of 246 common DEGs (180 upregulated and 66 downregulated) and identified 2 significant modules involved in ribosome biogenesis and TNF signaling. Meta-analysis of key genes of each module in cytoHubba identified 17 hub genes (ATF3, BYSL, DUSP1, EGFR, JUN, ETS1, LYAR, NIP7, NOLC1, NOP2, NOP56, PNO1, RRS1, TNFAIP3, TNFRSF1B, UTP15, VEGFA). Of the 17 hub genes, ATF3, BYSL, EGFR, JUN, NIP7, NOLC1, PNO1, RRS1, TNFAIP3 and VEGFA were identified as hypoxia signatures associated with poor prognosis in Glioma. Ribosome biogenesis emerged as a vital contender of possible therapeutic potential with BYSL, NIP7, NOLC1, PNO1 and RRS1 showing prognostic value. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s13205-021-02987-2.

10.
J Cell Mol Med ; 14(11): 2646-54, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19508390

RESUMO

Hypoxia is an integral part of tumorigenesis and contributes extensively to the neoplastic phenotype including drug resistance and genomic instability. It has also been reported that hypoxia results in global demethylation. Because a majority of the cytosine-phosphate-guanine (CpG) islands are found within the repeat elements of DNA, and are usually methylated under normoxic conditions, we suggested that retrotransposable Alu or short interspersed nuclear elements (SINEs) which show altered methylation and associated changes of gene expression during hypoxia, could be associated with genomic instability. U87MG glioblastoma cells were cultured in 0.1% O2 for 6 weeks and compared with cells cultured in 21% O2 for the same duration. Real-time PCR analysis showed a significant increase in SINE and reverse transcriptase coding long interspersed nuclear element (LINE) transcripts during hypoxia. Sequencing of bisulphite treated DNA as well as the Combined Bisulfite Restriction Analysis (COBRA) assay showed that the SINE loci studied underwent significant hypomethylation though there was patchy hypermethylation at a few sites. The inter-alu PCR profile of DNA from cells cultured under 6-week hypoxia, its 4-week revert back to normoxia and 6-week normoxia showed several changes in the band pattern indicating increased alu mediated genomic alteration. Our results show that aberrant methylation leading to increased transcription of SINE and reverse transcriptase associated LINE elements could lead to increased genomic instability in hypoxia. This might be a cause of genetic heterogeneity in tumours especially in variegated hypoxic environment and lead to a development of foci of more aggressive tumour cells.


Assuntos
Elementos Alu/genética , Metilação de DNA , Instabilidade Genômica , Hipóxia/genética , Apoptose , Western Blotting , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Caspases/metabolismo , Proliferação de Células , Ilhas de CpG/genética , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Hipóxia/patologia , Elementos Nucleotídeos Longos e Dispersos/genética , Osteossarcoma/genética , Osteossarcoma/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Elementos Nucleotídeos Curtos e Dispersos/genética , Transcrição Gênica , Células Tumorais Cultivadas
11.
BMC Cancer ; 9: 5, 2009 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-19126244

RESUMO

BACKGROUND: We had earlier used the comparison of RAPD (Random Amplification of Polymorphic DNA) DNA fingerprinting profiles of tumor and corresponding normal DNA to identify genetic alterations in primary human glial tumors. This has the advantage that DNA fingerprinting identifies the genetic alterations in a manner not biased for locus. METHODS: In this study we used RAPD-PCR to identify novel genomic alterations in the astrocytic tumors of WHO grade II (Low Grade Diffuse Astrocytoma) and WHO Grade IV (Glioblastoma Multiforme). Loss of heterozygosity (LOH) of the altered region was studied by microsatellite and Single Nucleotide Polymorphism (SNP) markers. Expression study of the gene identified at the altered locus was done by semi-quantitative reverse-transcriptase-PCR (RT-PCR). RESULTS: Bands consistently altered in the RAPD profile of tumor DNA in a significant proportion of tumors were identified. One such 500 bp band, that was absent in the RAPD profile of 33% (4/12) of the grade II astrocytic tumors, was selected for further study. Its sequence corresponded with a region of FAT, a putative tumor suppressor gene initially identified in Drosophila. Fifty percent of a set of 40 tumors, both grade II and IV, were shown to have Loss of Heterozygosity (LOH) at this locus by microsatellite (intragenic) and by SNP markers. Semi-quantitative RT-PCR showed low FAT mRNA levels in a major subset of tumors. CONCLUSION: These results point to a role of the FAT in astrocytic tumorigenesis and demonstrate the use of RAPD analysis in identifying specific alterations in astrocytic tumors.


Assuntos
Astrocitoma/genética , Caderinas/genética , Neoplasias do Sistema Nervoso Central/genética , Genes Supressores de Tumor , Perda de Heterozigosidade , Astrocitoma/patologia , Sítios de Ligação , Neoplasias do Sistema Nervoso Central/patologia , Impressões Digitais de DNA/métodos , Primers do DNA , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Polimorfismo de Nucleotídeo Único , Técnica de Amplificação ao Acaso de DNA Polimórfico/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Cancer Res ; 78(11): 2978-2989, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29535220

RESUMO

Inhibin is a heterodimeric TGFß family ligand that is expressed in many cancers and is a selective biomarker for ovarian cancers; however, its tumor-specific functions remain unknown. Here, we demonstrate that the α subunit of inhibin (INHA), which is critical for the functionality of dimeric inhibin A/B, correlates with microvessel density in human ovarian tissues and is predictive of poor clinical outcomes in multiple cancers. We demonstrate that inhibin-regulated angiogenesis is necessary for metastasis. Although inhibin had no direct impact on tumor cell signaling, both tumor cell-derived and recombinant inhibin elicit a strong paracrine response from endothelial cells by triggering SMAD1/5 activation and angiogenesis in vitro and in vivo Inhibin-induced angiogenesis was abrogated via anti-inhibin α antibodies. The endothelial-specific TGFß receptor complex comprising ALK1 and endoglin was a crucial mediator of inhibin signaling, offering a molecular mechanism for inhibin-mediated angiogenesis. These results are the first to define a role for inhibin in tumor metastasis and vascularization and offer an antibody-based approach for targeting inhibin therapeutically.Significance: Inhibin is a predictor of poor patient survival in multiple cancers and is a potential target for antiangiogenic therapies. Cancer Res; 78(11); 2978-89. ©2018 AACR.


Assuntos
Inibinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Metástase Neoplásica/patologia , Neovascularização Patológica/metabolismo , Neoplasias Ovarianas/metabolismo , Receptores de Activinas Tipo II/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Neovascularização Patológica/patologia , Neoplasias Ovarianas/patologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo
13.
Asian Pac J Cancer Prev ; 18(11): 3073-3079, 2017 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-29172281

RESUMO

Background: The 15q24-25 loci contain genes (CHRNA5 and CHRNA3) encoding nicotinic acetylcholine receptor subunits. We here determined for the first time the association of genetic variants rs16969968 and rs3743074 in CHRNA5 and CHRNA3, respectively, on nicotine dependence and lung cancer risk in a North Indian population by a case-control approach. Methods: Venous blood samples were obtained from 324 participants (108 lung cancer patients and 216 healthy individuals). DNA was extracted and PCR amplified with primers flanking the SNPs rs16969968 and rs3743074. Amplicons were subjected to sequencing and logistic regression was used to analyze association between variables. Results: The risk variant SNP rs16969968 in both heterozygous and homozygous forms appeared to exert a significant effect on nicotine dependence [GA (OR=2.77) and AA (OR=2.53)]. As expected, smoking was strongly associated with lung cancer (OR= 2.62). Risk allele rs16969968 in CHRNA5 also showed a significant association with increased lung cancer risk in our cohort, alone (OR= 4.99) and with smoking as a co-variable (OR= 4.28). Comparison of our analysis with other populations suggested that individuals with rs16969968 risk allele in the Indian population are more susceptible to lung cancer. Conclusion: Overall, the results strongly indicated that, in our cohort North Indian population, the genetic variant rs16969968, but not rs3743074, is significantly associated with both nicotine dependence and increased risk of lung cancer. While the results are significant, there is further need to increase the sample size and improve precision of our risk prediction.

14.
Chem Biol Interact ; 254: 221-30, 2016 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-27270449

RESUMO

BACKGROUND: The transition metal oxide nanoparticles are in focus for their anti-cancer potential. In this study we have synthesized and characterized CuO, NiO and Fe2O3 nanoparticles and, investigated their cytotoxic potential in the heterogeneous tumour microenvironment. METHODS: Nanoparticles were synthesized by aqueous precipitation method and characterized with UV-Visible spectrophotometer, Fourier transform infrared spectroscopy (FTIR), Scanning electron microscopy (SEM) and X-ray diffraction (XRD). Cell viability of lung cancer cells (A549) grown in normoxia (18%O2) and hypoxia (1%O2) was determined for all nanoparticles. The mechanism of cell death was assessed by nuclear morphological analysis, flow cytometry analysis and western blotting. Generation of intracellular ROS in treated cells and its contribution to cell viability was determined. RESULTS: The synthesized metal oxide nanoparticles were successfully characterized with SEM, spectroscopy and X-ray diffraction patterns. Cell viability of lung cancer cells was compromised in both normoxia and hypoxia. ROS generation was shown to contribute to cellular toxicity in CuO, but not NiO and Fe2O3. CONCLUSION: We have shown the therapeutic potential of CuO, NiO and Fe2O3 nanoparticles in non small cell lung cancer cells cultured in hypoxia, a relevant feature of solid tumors along with normoxia. The newly synthesized nanoparticles showed efficacy in both conditions. GENERAL SIGNIFICANCE: Hypoxia drives metabolic alterations and epigenetic modifications in the tumor microenvironment. By using conditions that mimic tumour microenvironment, this study expands the possibility of using metal oxide nanoparticles as a therapeutic agent for lung cancer treatment.


Assuntos
Apoptose/efeitos dos fármacos , Nanopartículas Metálicas , Óxidos/farmacologia , Elementos de Transição/farmacologia , Microambiente Tumoral , Células A549 , Antineoplásicos/química , Antineoplásicos/farmacologia , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Citometria de Fluxo , Histonas/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Microscopia Eletrônica de Varredura , Poli(ADP-Ribose) Polimerase-1/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier , Ressonância de Plasmônio de Superfície , Termogravimetria , Elementos de Transição/química , Difração de Raios X
15.
Oncotarget ; 7(44): 71841-71855, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27708247

RESUMO

Non-small cell lung cancer (NSCLC), comprising 85% of lung cancer cases, has been associated with resistance to chemo/radiotherapy. The hypoxic tumor micro-environment, where insufficient vasculature results in poor drug penetrance and sub-optimal chemotherapy in the tumor interiors contributes heavily to this resistance. Additionally, epigenetic changes in tumorigenic cells also change their response to different forms of therapy. In our study, we have investigated the effectiveness of a combination of cisplatin with scriptaid [a pan-Histone Deacetylase inhibitor (HDACi)] in a model that mimics the tumor microenvironment of hypoxia and sub-lethal chemotherapy. Scriptaid synergistically increases the efficacy of cisplatin in normoxia as well as hypoxia, accompanied with reduced metastasis and enhanced DNA damage. Addition of scriptaid also overcomes the cisplatin resistance exhibited in lung cancer cells with stabilized hypoxia inducible factor 1 (HIF1)-α (mutant) and mutant p53. Molecular studies showed that the combination treatment increased apoptotic cell death in both normoxia and hypoxia with a dual role of p38MAPK. Together, our results suggest that the combination of low dose cisplatin and scriptaid is cytotoxic to NSCLC lines, can overcome hypoxia induced resistance and mutant p53- induced instability often associated with this cancer, and has the potential to be an effective therapeutic modality.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Contagem de Células , Cisplatino/farmacologia , Genes p53 , Inibidores de Histona Desacetilases/imunologia , Hidroxilaminas/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Mutação , Quinolinas/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Cisplatino/administração & dosagem , Resistencia a Medicamentos Antineoplásicos , Inibidores de Histona Desacetilases/farmacologia , Humanos , Hidroxilaminas/administração & dosagem , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Neoplasias Pulmonares/genética , Quinolinas/administração & dosagem , Microambiente Tumoral
16.
Expert Rev Neurother ; 15(8): 935-46, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26118735

RESUMO

Glioblastomas are the most common form of brain tumor with a very dismal prognosis. While a standard treatment regimen of surgery followed by chemo/radiotherapy is currently used, this has only marginally improved the survival time of patients with little benefit on tumor recurrence. Although many molecular targets have already been identified and tested in clinical trials, very few are approved for use in clinics. Efforts are ongoing to target newer molecules that could be used for drug development. This review provides up-to-date information on the drugs and their molecular targets, which are currently in different stages of clinical trials. Since multiple signaling pathways are deregulated, it appears that the use of combination drugs along with personalized targeting approach would provide better therapy in the future.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Terapia Combinada , Humanos
17.
Int J Biochem Cell Biol ; 34(2): 148-57, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11809417

RESUMO

Generation of reactive oxygen species (ROS) is an important mode of action of many chemotherapeutic agents. Hydrogen peroxide (H(2)O(2)) is a model oxidant that has been used to study the response of cells to oxidative stress. The role of p53 in ROS induced cell death has not been consistent and has been shown to be cell type dependent. Study of cellular and molecular parameters and mechanisms involved in H(2)O(2) induced cell death in glioma cells will contribute to the understanding of response of these cells to oxidative stress. We investigated induction of cell death by H(2)O(2), and its relation to p53 in two human glial tumor derived cell lines U87MG (wild type p53) and U373MG (mutated p53). We observed that H(2)O(2) was able to induce apoptosis (as shown by morphology, flow cytometry and DNA fragmentation studies) in U87MG in a dose dependent manner. Dimethyl sulfoxide (DMSO), a known ROS scavenger, was protective to the cells. H(2)O(2) induced cell death was significantly reduced by antisense p53 oligonucleotide. Pretreatment with pyrrolidine dithiocarbamate (PDTC), an inhibitor of the redox sensitive transcription factor NF-kappa B, abrogated the increased expression of p53 protein in response to H(2)O(2), and enhanced cell survival. The U373MG cell line, having mutated p53, was comparatively resistant to H(2)O(2) induced cell death. We conclude from the study that p53, activated by NF-kappa B, is essential for H(2)O(2) induced apoptosis in glioma cells.


Assuntos
Apoptose/efeitos dos fármacos , Glioma/metabolismo , Peróxido de Hidrogênio/farmacologia , Estresse Oxidativo , Proteína Supressora de Tumor p53/metabolismo , Antioxidantes/farmacologia , Apoptose/fisiologia , Neoplasias Encefálicas/metabolismo , Sobrevivência Celular , Dimetil Sulfóxido/farmacologia , Citometria de Fluxo , Humanos , NF-kappa B/antagonistas & inibidores , Oligonucleotídeos Antissenso/farmacologia , Oxidantes/farmacologia , Pirrolidinas/farmacologia , Solventes/farmacologia , Tiocarbamatos/farmacologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética
18.
Int J Biochem Cell Biol ; 35(12): 1639-44, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12962703

RESUMO

We had earlier shown that higher concentration of hydrogen peroxide (H(2)O(2)) induced p53-dependent apoptosis in glioma cell line with wild type p53 but had minimal effect on cells with mutated p53. Here we show a potentiating effect of hydroxylamine (HA), an inhibitor of catalase, on a nontoxic dose of H(2)O(2) in glioma cells. HA sensitized both p53 wild type and mutated glioma cells to 0.25 mM H(2)O(2). Potentiating effect of HA was independent of p53. Higher levels of reactive oxygen species (ROS) generation were observed in cells treated with HA+H(2)O(2) as compared to cells treated with each component alone in both the cell lines. Dimethyl sulfoxide (DMSO) protected cells. Cytosolic cytochrome c and activated caspase 3 were detected at 4h. The results suggest that higher levels of intracellular ROS, generated by HA+H(2)O(2) act as a molecular switch in activating a rapidly acting p53-independent mitochondrial apoptotic pathway.


Assuntos
Glioma/metabolismo , Peróxido de Hidrogênio/metabolismo , Hidroxilamina/metabolismo , Oxidantes/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Caspase 3 , Caspases/metabolismo , Morte Celular , Linhagem Celular Tumoral , Citocromos c/metabolismo , Dimetil Sulfóxido/metabolismo , Ativação Enzimática , Glioma/patologia , Humanos , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Taxa de Sobrevida
19.
Cancer Gene Ther ; 11(8): 525-31, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15167899

RESUMO

Most gene therapy strategies related to p53 concentrate on the restoration of the activity of mutant p53, as several observations indicate that tumors and cell lines having the mutant gene are resistant to chemotherapy. However, as there is also some evidence to the contrary, we studied the relationship of the p53 status to the cellular response of glioma cells that were exposed to cisplatin. At a concentration of 2.5 microg/ml (which is about half the peak pharmacological blood level reached during chemotherapy), U373MG glioma cells, which had a mutant p53 gene, were more sensitive to the drug as compared to U87MG glioma cells (with normal p53). The U373MG cells responded with apoptosis while U87MG cells responded with a G2-M arrest. In U87MG cells, blocking the p53 response by antisense oligonucleotides also sensitized the cells to 2.5 microg/ml cisplatin, and shifted the cellular response from arrest to caspase 3-mediated apoptosis. A sensitive, p53-independent, mechanism for chemotherapy-induced apoptosis suggests that, in some cases, p53 abrogation by gene therapy or small molecule-based strategies could be a viable therapeutic strategy.


Assuntos
Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Glioma/tratamento farmacológico , Oligorribonucleotídeos Antissenso/uso terapêutico , Proteína Supressora de Tumor p53/antagonistas & inibidores , Antineoplásicos/farmacologia , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Fase G2/efeitos dos fármacos , Genes p53 , Glioma/genética , Humanos , Mutação/genética , Oligorribonucleotídeos Antissenso/farmacologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
20.
Cancer Genet Cytogenet ; 198(2): 126-34, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20362227

RESUMO

There are few reports of loss of heterozygosity (LOH) of 1p and 19q in astrocytic tumors, especially glioblastoma multiforme (GBM). We evaluated 1p and 19q (either or both) heterozygosity status in 71 astrocytomas, including 6 pediatric cases: 20 diffuse astrocytomas (DA), 9 anaplastic astrocytomas (AA), and 42 GBM. In the GBMs, p53 protein expression was assessed by immunohistochemistry and epidermal growth factor receptor (EGFR) gene amplification by fluorescence in situ hybridization; TP53 sequencing was done in 15 of the GBMs. In adults, LOH of 1p or 19q was detected in 16% of DAs and 50% of GBMs; none of the AAs showed this alteration. In GBMs, LOH of 19q was most common (26%), followed by combined 1p and 19q LOH (13%) and 1p LOH (10%). Pediatric GBMs also harbored isolated 1p and 19q LOH (50%). Notably, LOH of 1p or 19q LOH was more frequent in p53 immunopositive secondary GBMs (61%) than in primary GBMs (17%). This suggests that LOH of 1p and 19q may be acquired during progression to secondary GBMs. Thus, 1p and 19q LOH can occur in astrocytic tumors, most commonly in secondary GBMs without morphological correlation with an oligodendroglial histology. The clinical significance of recognition of this subset of GBMs is based on several recent reports of association with better prognosis, although long-term follow-up studies are required.


Assuntos
Astrocitoma/genética , Neoplasias Encefálicas/genética , Cromossomos Humanos Par 19 , Cromossomos Humanos Par 1 , Amplificação de Genes , Genes erbB-1 , Proteína Supressora de Tumor p53/genética , Adolescente , Adulto , Idoso , Astrocitoma/metabolismo , Astrocitoma/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Estudos de Casos e Controles , Criança , Feminino , Amplificação de Genes/fisiologia , Heterozigoto , Humanos , Índia , Perda de Heterozigosidade , Masculino , Pessoa de Meia-Idade , Proteína Supressora de Tumor p53/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa