Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nanomedicine (Lond) ; 18(26): 1907-1919, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38078434

RESUMO

Aim: We investigated our previous finding of increased retention of poly(lactic-co-glycolic) acid nanoparticles (PLGA-NPs) with metabolic inhibitors (MI) and studied the effect of some small molecule inhibitors on PLGA-NP assimilation. Materials & methods: Intracellular PLGA-NP colocalization in the presence of MI was investigated by confocal microscopy. Intracellular retention of PLGA-NPs by some small molecules was estimated by fluorescence microscopy and flow cytometry after Pulse/Chase experiments. Results: MI caused PLGA-NP colocalization in intracellular membranous structures, mainly endosomes and lysosomes. Some small molecule inhibitors demonstrated increased intracellular PLGA-NP accumulation. Conclusion: This study elucidates the movement of PLGA-NP in cells and suggests that clinically used small molecules can reduce their extrusion by enhancing their stay within intracellular vesicles, with possible clinically beneficial consequences.


Nanoparticles are increasingly being used to carry drugs for treatment of cancer. We wish to decrease their movement out of the cells. This may give time for them to unload their drugs. Cells were treated with nanoparticles for 30 min and observed. Then the nanoparticles were washed off. Cells were again observed after 30 min. Various intracellular trafficking inhibitors were also added. Nanoparticle retention and subcellular localization were measured. We found that nanoparticles are trapped in some membranous compartments within the cells after energy depletion. We also discovered some commonly used clinical molecules that can decrease the excretion of nanoparticles from the cells. These inhibitors can be utilized for increasing the intracellular stay of the drug-loaded nanoparticles.


Assuntos
Nanopartículas , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ácido Poliglicólico/química , Ácido Láctico/química , Glicóis , Nanopartículas/química , Portadores de Fármacos/química
2.
J Exp Clin Cancer Res ; 42(1): 231, 2023 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-37670323

RESUMO

BACKGROUND: Acute lymphoblastic leukemia (ALL) is the most common pediatric hematological malignancy, with ETV6::RUNX1 being the most prevalent translocation whose exact pathogenesis remains unclear. IGF2BP1 (Insulin-like Growth Factor 2 Binding Protein 1) is an oncofetal RNA binding protein seen to be specifically overexpressed in ETV6::RUNX1 positive B-ALL. In this study, we have studied the mechanistic role of IGF2BP1 in leukemogenesis and its synergism with the ETV6::RUNX1 fusion protein. METHODS: Gene expression was analyzed from patient bone marrow RNA using Real Time RT-qPCR. Knockout cell lines were created using CRISPR-Cas9 based lentiviral vectors. RNA-Seq and RNA Immunoprecipitation sequencing (RIP-Seq) after IGF2BP1 pulldown were performed using the Illumina platform. Mouse experiments were done by retroviral overexpression of donor HSCs followed by lethal irradiation of recipients using a bone marrow transplant model. RESULTS: We observed specific overexpression of IGF2BP1 in ETV6::RUNX1 positive patients in an Indian cohort of pediatric ALL (n=167) with a positive correlation with prednisolone resistance. IGF2BP1 expression was essential for tumor cell survival in multiple ETV6::RUNX1 positive B-ALL cell lines. Integrated analysis of transcriptome sequencing after IGF2BP1 knockout and RIP-Seq after IGF2BP1 pulldown in Reh cell line revealed that IGF2BP1 targets encompass multiple pro-oncogenic signalling pathways including TNFα/NFκB and PI3K-Akt pathways. These pathways were also dysregulated in primary ETV6::RUNX1 positive B-ALL patient samples from our center as well as in public B-ALL patient datasets. IGF2BP1 showed binding and stabilization of the ETV6::RUNX1 fusion transcript itself. This positive feedback loop led to constitutive dysregulation of several oncogenic pathways. Enforced co-expression of ETV6::RUNX1 and IGF2BP1 in mouse bone marrow resulted in marrow hypercellularity which was characterized by multi-lineage progenitor expansion and strong Ki67 positivity. This pre-leukemic phenotype confirmed their synergism in-vivo. Clonal expansion of cells overexpressing both ETV6::RUNX1 and IGF2BP1 was clearly observed. These mice also developed splenomegaly indicating extramedullary hematopoiesis. CONCLUSION: Our data suggest a combined impact of the ETV6::RUNX1 fusion protein and RNA binding protein, IGF2BP1 in activating multiple oncogenic pathways in B-ALL which makes IGF2BP1 and these pathways as attractive therapeutic targets and biomarkers.


Assuntos
Leucemia-Linfoma Linfoblástico de Células Precursoras B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animais , Camundongos , Subunidade alfa 2 de Fator de Ligação ao Core , Camundongos Knockout , Fosfatidilinositol 3-Quinases , Variante 6 da Proteína do Fator de Translocação ETS
3.
Int J Appl Basic Med Res ; 13(2): 106-112, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37614837

RESUMO

Background: Chemokine receptor CXCR4 is frequently present in cells of various cancers. Hence, targeted therapy using CXCR4 ligands, such as DV1 peptide, on drug-loaded nanoparticles, has the potential to enhance the efficiency of cancer treatment. Aim: The present study created a CXCR4-targeting drug delivery system using avidin-poly (lactic-co-glycolic acid) (PLGA) nanoparticle surface tagged with biotinylated DV1 peptide ligand. Materials and Methods: A double-emulsion solvent evaporation technique was employed to prepare avidin-PLGA nanoparticles and characterized by transmission electron microscopy (TEM) and dynamic light scattering. Uptake was studied by confocal microscopy after incorporating fluorescein isothiocyanate (FITC)-labeled albumin inside the nanoparticles during their synthesis. Peptide-biotin-avidin-PLGA nanoparticles were tested in vitro on CXCR4-expressing U87MG cells. Photomicroscopy was done by a Nikon A1 Confocal Microscope, and pictures were analyzed by Nikon NIS-Elements BR software. Results: Experimental results confirmed the specificity of DV1 peptide-tagged avidin-PLGA nanoparticles for cells expressing CXCR4 receptors. The avidin-PLGA nanoparticles were successfully synthesized and the same was confirmed by tagging them with FITC-labeled biotin. Conclusion: Avidin-PLGA nanoparticle surface tagged with biotinylated DV1 peptide ligand has potential clinical application in the treatment of various cancers as targeted therapy for CXCR4-expressing cancer cells.

4.
Mol Carcinog ; 62(12): 1817-1831, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37606187

RESUMO

The atypical cadherin FAT1 function either as a pro or antitumorigenic in tumors of different tissue origins. Our group previously demonstrated the protumorigenic nature of FAT1 signaling in glioblastoma (GBM). In this study, we investigated how FAT1 influences the expression of clustered oncomiRs (miR-221-3p/miR-222-3p) and their downstream effects in GBM. Through several experiments involving the measurement of specific gene/microRNA expression, gene knockdowns, protein and cellular assays, we have demonstrated a novel oncogenic signaling pathway mediated by FAT1 in glioma. These results have been verified using antimiRs and miR-mimic assays. Initially, in glioma-derived cell lines (U87MG and LN229), we observed FAT1 as a novel up-regulator of the transcription factor NFκB-RelA. RelA then promotes the expression of the clustered-oncomiRs, miR-221-3p/miR-222-3p, which in turn suppresses the expression of the tumor suppressor gene (TSG), PDCD10 (Programmed cell death protein10). The suppression of PDCD10, and other known TSG targets (PTEN/PUMA), by miR-221-3p/miR-222-3p, leads to increased clonogenicity, migration, and invasion of glioma cells. Consistent with our in-vitro findings, we observed a positive expression correlation of FAT1 and miR-221-3p, and an inverse correlation of FAT1 and the miR-targets (PDCD10/PTEN/PUMA), in GBM tissue-samples. These findings were also supported by publicly available GBM databases (The Cancer Genome Atlas [TCGA] and The Repository of Molecular Brain Neoplasia Data [Rembrandt]). Patients with tumors displaying high levels of FAT1 and miR-221-3p expression (50% and 65% respectively) experienced shorter overall survival. Similar results were observed in the TCGA-GBM database. Thus, our findings show a novel FAT1/RelA/miR-221/miR-222 oncogenic-effector pathway that downregulates the TSG, PDCD10, in GBM, which could be targeted therapeutically in a specific manner.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , MicroRNAs , Humanos , Glioblastoma/metabolismo , Caderinas/genética , Caderinas/metabolismo , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Glioma/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Proliferação de Células/genética , Movimento Celular/genética , Proteínas de Membrana/genética , Proteínas Proto-Oncogênicas/genética
5.
Front Immunol ; 13: 813888, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35720420

RESUMO

FAT atypical cadherin 1 (FAT1) promotes glioblastoma (GBM) by promoting protumorigenic inflammatory cytokine expression in tumor cells. However, tumors also have an immunosuppressive microenvironment maintained by mediators such as transforming growth factor (TGF)-ß cytokines. Here, we have studied the role of FAT1 in tumor immune suppression. Our preliminary TIMER2.0 analysis of The Cancer Genome Atlas (TCGA) database revealed an inverse correlation of FAT1 expression with infiltration of tumor-inhibiting immune cells (such as monocytes and T cells) and a positive correlation with tumor-promoting immune cells [such as myeloid-derived suppressor cells (MDSCs)] in various cancers. We have analyzed the role of FAT1 in modulating the expression of TGF-ß1/2 in resected human gliomas, primary glioma cultures, and other cancer cell lines (U87MG, HepG2, Panc-1, and HeLa). Positive correlations of gene expression of FAT1 and TGF-ß1/2 were observed in various cancers in TCGA, Glioma Longitudinal Analysis Consortium (GLASS), and Chinese Glioma Genome Atlas (CGGA) databases. Positive expression correlations of FAT1 were also found with TGF-ß1/2 and Serpine1 (downstream target) in fresh-frozen GBM samples using q-PCR. siRNA-mediated FAT1 knockdown in cancer cell lines and in primary cultures led to decreased TGF-ß1/2 expression/secretion as assessed by q-PCR, Western blotting, and ELISA. There was increased chemotaxis (transmigration) of THP-1 monocytes toward siFAT1-transfected tumor cell supernatant as a consequence of decreased TGF-ß1/2 secretion. Reduced TGF-ß1 expression was also observed in THP-1 cultured in conditioned media from FAT1-depleted glioma cells, thus contributing to immune suppression. In U87MG cells, decreased TGF-ß1 upon FAT1 knockdown was mediated by miR-663a, a known modulator. FAT1 expression was also observed to correlate positively with the expression of surrogate markers of MDSCs [programmed death ligand-1 (PD-L1), PD-L2, and interleukin (IL)-10] in glioma tumors, suggesting a potential role of FAT1 in MDSC-mediated immunosuppression. Hence, our findings elaborate contributions of FAT1 to immune evasion, where FAT1 enables an immunosuppressive microenvironment in GBM and other cancers via TGF-ß1/2.


Assuntos
Caderinas , Glioblastoma , Glioma , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Glioblastoma/patologia , Glioma/genética , Glioma/metabolismo , Humanos , Fator de Crescimento Transformador beta1/metabolismo , Microambiente Tumoral , Regulação para Cima
6.
Genes (Basel) ; 13(3)2022 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-35328060

RESUMO

Hypoxic ischemic injury to the fetal and neonatal brain is a leading cause of death and disability worldwide. Although animal and culture studies suggest that glutamate excitotoxicity is a primary contributor to neuronal death following hypoxia, the molecular mechanisms, and roles of various neural cells in the development of glutamate excitotoxicity in humans, is not fully understood. In this study, we developed a culture model of human fetal neural stem cell (FNSC)-derived astrocytes and examined their glutamate uptake in response to hypoxia. We isolated, established, and characterized cultures of FNSCs from aborted fetal brains and differentiated them into astrocytes, characterized by increased expression of the astrocyte markers glial fibrillary acidic protein (GFAP), excitatory amino acid transporter 1 (EAAT1) and EAAT2, and decreased expression of neural stem cell marker Nestin. Differentiated astrocytes were exposed to various oxygen concentrations mimicking normoxia (20% and 6%), moderate and severe hypoxia (2% and 0.2%, respectively). Interestingly, no change was observed in the expression of the glutamate transporter EAAT2 or glutamate uptake by astrocytes, even after exposure to severe hypoxia for 48 h. These results together suggest that human FNSC-derived astrocytes can maintain glutamate uptake after hypoxic injury and thus provide evidence for the possible neuroprotective role of astrocytes in hypoxic conditions.


Assuntos
Astrócitos , Ácido Glutâmico , Células-Tronco Neurais , Astrócitos/metabolismo , Hipóxia Celular , Células Cultivadas , Transportador 1 de Aminoácido Excitatório/genética , Transportador 1 de Aminoácido Excitatório/metabolismo , Ácido Glutâmico/metabolismo , Humanos , Células-Tronco Neurais/metabolismo
7.
Iran Biomed J ; 2022 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-37980558

RESUMO

Background: Acquired aplastic anemia is an autoimmune disease in which auto-aggressive T cells destroy hematopoietic progenitors. T-cell differentiation is controlled by transcription factors that interact with NOTCH-1, which influences the respective T-cell lineages. Notch signaling also regulates the BM microenvironment. The present study aimed to assess the gene expressions of NOTCH-1 and T helper cell transcription factors in the acquired aplastic anemia patients. Methods: Using quantitative real-time PCR, we studied the mRNA expression level for NOTCH-1, its ligands (DLL-1 and JAG-1), and T helper cell transcription factors (T-BET, GATA-3, and ROR-γt) in both PB and BM of aAA patients and healthy controls. Further, patients of aplastic anemia were stratified by their disease severity as per the standard criteria. Results: The mRNA expression level of NOTCH-1, T-BET, GATA-3, and ROR-γT genes increased in aAA patients compared to healthy controls. There was no significant difference in the mRNA expression of Notch ligands between patients and controls. The mRNA expression level of the above-mentioned genes was found to be higher in SAA and VSAA than NSAA patients. In addition, NOTCH-1 and T helper cell-specific transcription factors enhanced in aAA. We also observed a significant correlation between the genes and hematological parameters in patients. Conclusion: The interaction between NOTCH-1, T-BET, GATA-3, and ROR-γT might lead to the activation, proliferation, and polarization of T helper cells and subsequent BM destruction. The mRNA expression levels of genes varied with disease severity, which may contribute to pathogenesis of aAA.

8.
Front Oncol ; 11: 588101, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33708624

RESUMO

Around 85% of childhood Acute Lymphoblastic Leukemia (ALL) are of B-cell origin and characterized by the presence of different translocations including BCR-ABL1, ETV6-RUNX1, E2A-PBX1, and MLL fusion proteins. The current clinical investigations used to identify ETV6-RUNX1 translocation include FISH and fusion transcript specific PCR. In the current study we assessed the utility of IGF2BP1, an oncofetal RNA binding protein, that is over expressed specifically in ETV6-RUNX1 translocation positive B-ALL to be used as a diagnostic marker in the clinic. Further, public transcriptomic and Crosslinked Immunoprecipitation (CLIP) datasets were analyzed to identify the putative targets of IGF2BP1. We also studied the utility of using the mRNA expression of two such targets, MYC and EGFL7 as potential diagnostic markers separately or in conjunction with IGF2BP1. We observed that the expression of IGF2BP1 alone measured by RT-qPCR is highly sensitive and specific to be used as a potential biomarker for the presence of ETV6-RUNX1 translocation in future.

9.
Nanomedicine (Lond) ; 15(23): 2229-2239, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32924855

RESUMO

Background: Poly(lactic-co-glycolic) acid nanoparticle (PLGA-NP) trafficking across cell membranes was investigated to confirm preliminary results that contradicted existing studies. Materials & methods: Uptake and retention of PLGA-NPs at 37 and 4°C in the presence and absence of metabolic inhibitors in various cell lines was estimated. Results: Pulse experiments with metabolic inhibitors and culturing at 4°C demonstrated the predominantly passive nature of PLGA-NP uptake. Chase experiments with metabolic inhibitors indicated the role of active exocytosis in the extrusion of these NPs. PLGA-NPs with ionic or nonionic hydrophilic coats with highly positive or negative ζ-potential also showed similar results. Conclusion: Our study opens up the possibility of modulation of active exocytosis to increase intracellular retention of NPs for an extended period of drug delivery.


Assuntos
Nanopartículas , Neoplasias , Linhagem Celular , Portadores de Fármacos , Ácido Láctico , Neoplasias/tratamento farmacológico , Tamanho da Partícula , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
10.
BMC Cancer ; 20(1): 62, 2020 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-31992226

RESUMO

BACKGROUND: Overexpression of FAT1 gene and its oncogenic effects have been reported in several cancers. Previously, we have documented upregulation of FAT1 gene in glioblastoma (GBM) tumors which was found to increase the expression of proinflammatory markers, HIF-1α, stemness genes and EMT markers in glioma cells. Here, we reveal NFкB (RelA)/RelA/p65 as the transcriptional regulator of FAT1 gene in GBM cells. METHODS: In-silico analysis of FAT1 gene promoter was performed using online bioinformatics tool Promo alggen (Transfac 8.3) to identify putative transcription factor(s) binding motifs. A 4.0 kb FAT1 promoter (- 3220 bp to + 848 bp w.r.t. TSS + 1) was cloned into promoter less pGL3Basic reporter vector. Characterization of FAT1 promoter for transcriptional regulation was performed by in-vitro functional assays using promoter deletion constructs, site directed mutagenesis and ChIP in GBM cells. RESULTS: Expression levels of NFкB (RelA) and FAT1 were found to be increased and positively correlated in GBM tumors (n = 16), REMBRANDT GBM-database (n = 214) and TCGA GBM-database (n = 153). In addition to glioma, positive correlation between NFкB (RelA) and FAT1 expression was also observed in other tumors like pancreatic, hepatocellular, lung and stomach cancers (data extracted from TCGA tumor data). A 4.0 kb FAT1-promoter-construct [- 3220 bp/+ 848 bp, transcription start site (TSS) + 1, having 17 NFкB (RelA) motifs] showed high FAT1 promoter luciferase-activity in GBM cells (U87MG/A172/U373MG). FAT1 promoter deletion-construct pGL3F1 [- 200 bp/+ 848 bp, with 3-NFкB (RelA)-motifs] showed the highest promoter activity. Exposure of GBM cells to known NFкB (RelA)-activators [severe-hypoxia/TNF-α/ectopic-NFкB (RelA) + IKBK vectors] led to increased pGL3F1-promoter activity and increased endogenous-FAT1 expression. Conversely, siRNA-mediated NFкB (RelA) knockdown led to decreased pGL3F1-promoter activity and decreased endogenous-FAT1 expression. Deletion of NFкB (RelA)-motif at - 90 bp/- 80 bp [pGL3F1δ1-construct] showed significant decrease in promoter activity. Site directed mutagenesis at -90 bp/- 80 bp and ChIP assay for endogenous-NFкB (RelA) confirmed the importance of this motif in FAT1 expression regulation. Significant reduction in the migration, invasion as well as colony forming capacity of the U87MG glioma cells was observed on siRNA-mediated knockdown of NFкB (RelA). CONCLUSION: Since FAT1 and NFкB (RelA) are independently known to promote pro-tumorigenic inflammation and upregulate the expression of HIF-1α/EMT/stemness in tumors, targeting the NFкB (RelA)-FAT1 axis may attenuate an important tumor-promoting pathway in GBM. This may also be applicable to other tumors.


Assuntos
Neoplasias Encefálicas/metabolismo , Caderinas/genética , Glioma/metabolismo , Fator de Transcrição RelA/metabolismo , Sítios de Ligação , Neoplasias Encefálicas/genética , Caderinas/química , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Clonagem Molecular , Simulação por Computador , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Humanos , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , Transdução de Sinais
11.
Neuroscience ; 408: 293-307, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-31026564

RESUMO

Brain-derived neurotrophic factor (BDNF) is known to mediate activity-dependent changes in the developing auditory system. Its expression in the brainstem auditory nuclei, auditory cortex and hippocampus of neonatal chicks (Gallus gallus domesticus) in response to in ovo high intensity sound exposure at 110 dB (arrhythmic sound: recorded traffic noise, 30-3000 Hz with peak at 2700 Hz, rhythmic sound: sitar music, 100-4000 Hz) was examined to understand the previously reported altered volume and neuronal number in these regions. In the brainstem auditory nuclei, no mature BDNF, but proBDNF at the protein level was detected, and no change in its levels was observed after in ovo sound stimulation (music and noise). Increased ProBDNF protein levels were found in the auditory cortex in response to arrhythmic sound, along with decreased levels of one of the BDNF mRNA transcripts, in response to both rhythmic and arrhythmic sound stimulation. In the hippocampus, increased levels of mature BDNF were found in response to music. Expression microarray analysis was performed to understand changes in gene expression in the hippocampus in response to music and noise, followed by gene ontology analysis showing enrichment of probable signaling pathways. Differentially expressed genes like CAMK1 and STAT1 were found to be involved in downstream signaling on comparing music versus noise-exposed chicks. In conclusion, we report that BDNF is differentially regulated in the auditory cortex at the transcriptional and post-translational level, and in the hippocampus at the post-translational level in response to in ovo sound stimulation.


Assuntos
Córtex Auditivo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hipocampo/metabolismo , Estimulação Acústica , Animais , Animais Recém-Nascidos , Tronco Encefálico/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Galinhas , Neurônios/metabolismo
12.
Int J Cancer ; 142(4): 805-812, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28994107

RESUMO

Glioblastoma (GBM) is characterized by the presence of hypoxia, stemness and local invasiveness. We have earlier demonstrated that FAT1 promotes invasiveness, inflammation and upregulates HIF-1α expression and its signaling in hypoxic GBM. Here, we have identified the role of FAT1 in regulating EMT (epithelial-mesenchymal transition) and stemness characteristics in GBM. The expression of FAT1, EMT (Snail/LOX/Vimentin/N-cad), stemness (SOX2/OCT4/Nestin/REST) and hypoxia markers (HIF-1α/VEGF/PGK1/CA9) was upregulated in ≥39% of GBM tumors (n = 31) with significant positive correlation (p ≤ 0.05) of the expression of FAT1 with LOX/Vimentin/SOX2/HIF-1α/PGK1/VEGF/CA9. Furthermore, positive correlation (p ≤ 0.01) of FAT1 with Vimentin/N-cad/SOX2/REST/HIF-1α has been observed in TCGA GBM-dataset (n = 430). Analysis of cells (U87MG/A172) exposed to severe hypoxia (0.2%O2 ) revealed elevated mRNA expression of FAT1, EMT (Snail/LOX/Vimentin/N-cad), stemness (SOX2/OCT4/Nestin/REST) and hypoxia markers (HIF-1α/PGK1/VEGF/CA9) as compared to their normoxic (20%O2 ) counterparts. FAT1 knockdown in U87MG/A172 maintained in severe hypoxia and in normoxic primary glioma cultures led to significant reduction of EMT/stemness markers as compared to controls. HIF-1α knockdown in U87MG cells markedly reduced the expression of all the EMT/stemness markers studied except for Nestin and SOX2 which were more under the influence of FAT1. This indicates FAT1 has a novel regulatory effect on EMT/stemness markers both via or independent of HIF-1α. The functional relevance of our study was corroborated by significant reduction in the number of soft-agar colonies formed in hypoxic-siFAT1 treated U87MG cells. Hence, our study for the first time reveals FAT1 as a novel regulator of EMT/stemness in hypoxic GBM and suggests FAT1 as a potential therapeutic candidate.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Caderinas/genética , Glioblastoma/genética , Glioblastoma/patologia , Células-Tronco Neoplásicas/patologia , Neoplasias Encefálicas/metabolismo , Caderinas/biossíntese , Caderinas/metabolismo , Hipóxia Celular/genética , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glioblastoma/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Neoplásicas/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Células Tumorais Cultivadas
13.
Methods Mol Biol ; 1543: 245-257, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28349432

RESUMO

Double-stranded RNA-mediated transcriptional gene silencing (TGS) has shown promising results over posttranscriptional gene silencing (PTGS) due to its long term and heritable nature. Various research groups have shed light on different mechanisms by which TGS operate. Some of these include histone modification, DNA methylation, or restriction of RNA polymerase binding onto the target gene's promoter. This serves as an added advantage since permanent c-Myc inactivation is critical for suppressing hepatocellular carcinoma (HCC). Inability to target cancer cells specifically, without affecting the normal cells, has been one of the biggest drawbacks of an effective cancer therapy. Therefore, we aimed to overcome this barrier by first generating tumor-specific transcriptional units expressing TGS inducing shRNAs against c-Myc's P2 promoter only in neoplastic liver cells. Secondly, we coupled this TGS inducing system with Sendai fusion virosomes for liver-specific delivery to minimize nonspecific side effects in vitro.


Assuntos
Carcinoma Hepatocelular/genética , Inativação Gênica , Técnicas de Transferência de Genes , Genes myc , Neoplasias Hepáticas/genética , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Linhagem Celular Tumoral , Células Cultivadas , Ilhas de CpG , Metilação de DNA , Humanos , RNA Interferente Pequeno/administração & dosagem , Transfecção/métodos , Virossomos
14.
Int J Cancer ; 139(11): 2570-82, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27536856

RESUMO

The hypoxic microenvironment is an important contributor of glioblastoma (GBM) aggressiveness via HIF1α, while tumour inflammation is profoundly influenced by FAT Atypical Cadherin (FAT1). This study was designed to explore the functional interaction and significance of FAT1 and HIF1α under severe hypoxia-mimicking tumour microenvironment in primary human tumours. We first identified a positive correlation of FAT1 with HIF1α and its target genes in GBM tumour specimens, revealing the significance of the FAT1-HIF1α axis in glioma cells. We found that severe hypoxia leads to an increased expression of FAT1 and HIF1α in U87MG and U373MG cells. To reveal the relevance of FAT1 under hypoxic conditions, we depleted endogenous FAT1 under hypoxia and found a substantial reduction in the expression of HIF1α and its downstream target genes like CA9, GLUT1, VEGFA, MCT4, HK2, BNIP3 and REDD1, as well as a significant reduction in the invasiveness in GBM cells. At the molecular level, under hypoxia the FAT1 depletion-associated reduction in HIF1α was due to compromised EGFR-Akt signaling as well as increased VHL-dependent proteasomal degradation of HIF1α. In brief, for the first time, these results reveal an upstream master regulatory role of FAT1 in the expression and role of HIF1α under hypoxic conditions and that FAT1-HIF1α axis controls the invasiveness of GBM. Hence, FAT1 represents a novel potential therapeutic target for GBM.


Assuntos
Caderinas/metabolismo , Glioblastoma/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Caderinas/biossíntese , Caderinas/genética , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Técnicas de Silenciamento de Genes , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Gradação de Tumores , Invasividade Neoplásica , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Transdução de Sinais , Proteína Supressora de Tumor Von Hippel-Lindau/biossíntese
15.
Int J Biochem Cell Biol ; 78: 194-205, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27448842

RESUMO

The current treatment therapies available for malignant gliomas are inadequate. There is an urgent need to develop more effective therapies by characterizing the molecular pathogenesis of the disease. Over expression of platelet-derived growth factor (PDGF) ligands and receptors have been reported in malignant gliomas. Platelet-derived growth factor associated protein-1 (PDAP-1) is reported to modulate the mitogenic activity of PDGF ligands, but to date, there is no information concerning its role in PDGF-mediated glioma cell proliferation. This study aimed to characterize the role of PDAP-1 in PDGF-mediated glioma proliferation. The expression of PDAP-1 was observed to be significantly increased (p<0.05) in grade IV glioma tissue and cell lines compared to grade III. siRNA-mediated knockdown of PDAP-1 reduced the expression of PDGF-B and its downstream genes (Akt1/Protein kinase B (PKB) and phosphoinositide-dependent kinase-1 (PDK1) by up to 50%. In PDAP-1 knockdown glioma cells, more than a twofold reduction was also observed in the level of phosphorylated Akt. Interestingly, knockdown of PDAP-1 in combination with PDGF-B antibody inhibited glioma cell proliferation through activation of Caspase 3/7 and 9. We also demonstrate that PDAP-1 co-localizes with PDGF-B in the cytoplasm of glioma cells, and an interaction between both of the proteins was established. Collectively, these findings suggest that the expression of PDAP-1 is associated with disease malignancy, and its inhibition reduced the proliferation of malignant glioma cells through down-regulation of PDGF-B/Akt/PDK1 signaling. Thus, this study establishes PDAP-1 as an effecter of PDGF signaling in glioma cells and suggests that it could also be a promising therapeutic target.


Assuntos
Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Apoptose , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Técnicas de Silenciamento de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Cinética , Potencial da Membrana Mitocondrial , Gradação de Tumores , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-sis/genética , Transdução de Sinais
16.
Int J Biochem Cell Biol ; 74: 60-71, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26923292

RESUMO

Hypoxia is a salient feature of most solid tumors and plays a central role in tumor progression owing to its multiple contributions to therapeutic resistance, metastasis, angiogenesis and stemness properties. Reports exist in literature about hypoxia increasing stemness characteristics and invasiveness potential of malignant cells. In order to delineate molecular crosstalk among factors driving glioma progression, we used knockdown and overexpression strategies. We have demonstrated that U87MG and A172 glioma cells inherently have a subset of cells with high migratory potential due to migration-inducing Mena transcripts. These cells also have elevated stemness markers (Sox-2 and Oct-4). There was a significant increase of number in this subset of migratory cells on exposure to hypoxia with corresponding elevation (over 1000 fold) in migration-inducing Mena transcripts. We were able to demonstrate that a HIF-2α-Sox-2/Oct-4-Mena (INV) axis that is strongly activated in hypoxia and markedly increases the migratory potential of the cells. Such cells also formed tumor spheres with greater efficiency. We have correlated our in-vitro results with human glioblastoma samples and found that hypoxia, invasiveness and stemness markers correlated well in native tumor samples. This study identifies a novel signaling mechanism mediated by HIF-2α in regulating invasiveness and stemness characteristics, suggesting that under hypoxic conditions, some tumor cells acquire more migratory potential by increased Pan Mena and Mena INV expression as a consequence of this HIF-2α mediated increase in Oct-4 and Sox-2. These properties would help the cells to form a new nidus after local invasion or metastasis.


Assuntos
Glioma , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Adulto , Idoso , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Células Cultivadas , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Glioma/fisiopatologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Masculino , Pessoa de Meia-Idade , Fator 3 de Transcrição de Octâmero/genética , Fatores de Transcrição SOXB1/genética
17.
Biol Trace Elem Res ; 172(1): 120-126, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26660891

RESUMO

Copper is an essential microelement required for maintaining normal cell physiology. Copper transporter CutC is one of the six members of Cut family proteins, involved in prokaryotic copper homeostasis. Human homolog of CutC (hCutC) is an intracellular copper-binding protein with unknown physiological function. In the present study using HepG2 cells, we report the effects of hCutC knockdown on copper sensitivity and morphology of cells that ultimately leads to apoptosis. We silenced hCutC using specific small interfering RNA (siRNA), and its downregulation was confirmed by quantitative real-time PCR. Though there was no significant variation in total cellular copper as estimated by inductively coupled plasma-atomic emission spectrometry (ICP-AES), knockdown of hCutC caused an increase in sensitivity of HepG2 cells to copper loads when compared to control cells (studied by MTT-based cell viability assay). Morphological analysis by transmission electron microscopy (TEM) indicated onset of apoptosis in hCutC-silenced cells which was exacerbated upon copper treatment. Mitochondrial transmembrane potential (ΔΨm) assay and DNA fragmentation assay further ensured apoptosis occurring in cells upon hCutC silencing. The present study reveals copper induced damage in cells upon hCutC silencing and provides evidence for the role of hCutC protein in intracellular copper homeostasis.


Assuntos
Apoptose/efeitos dos fármacos , Apoptose/genética , Proteínas de Transporte de Cátions/deficiência , Proteínas de Transporte de Cátions/genética , Cobre/metabolismo , Cobre/farmacologia , Inativação Gênica , Proteínas de Transporte de Cobre , Relação Dose-Resposta a Droga , Células Hep G2 , Humanos , Relação Estrutura-Atividade
19.
J Transl Med ; 13: 254, 2015 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-26242403

RESUMO

BACKGROUND: Placental like alkaline phosphatase (PLAP), an oncofetal antigen, is highly expressed in germ cell, cervical, ovarian and several other tumour types but minimally in normal tissues [corrected]. The expression of a PLAP promoter based transcriptional unit following antigen mediated cell specific delivery is a possible approach for tumour targeting. METHODS: PLAP promoter alone or in combination with NFκB DNA response elements was used for expressing shRNA targeting the long control region (LCR) of human papillomavirus (HPV)-16 oncogenes E6 and E7 via transcriptional gene silencing in PLAP expressing cervical cancer cell lines, SiHa and CaSki. This was packaged in a Sendai virus envelope incorporating a single chain variable fragment antibody (scFv) for antibody mediated targeting. Specificity and efficacy of the shRNA was assessed by studying the heterochromatization, down regulation of the HPV-16 E6/E7 genes and subsequent effects on their targets and cell growth properties. RESULTS: Reduction of HPV-16 E6 and E7 expression by TGS led to the activation of the previously suppressed target genes of p53 (PUMA and NOXA) and Rb (cyclins A2 and E). Cell death was seen only in PLAP expressing HPV-16 infected SiHa and CaSki cells but not in the HPV-18 integrated HeLa and non-PLAP CHO cells. There was reduction in the enhancer associated transcripts of the long control region (LCR) of HPV-16 E6/E7 genes. Also, an increase in the enrichment of dimethylated histone three lysine nine (H3K9Me2) and trimethylated histone three lysine twenty-seven (H3K27Me3) was observed by ChIP assay, which decreased upon trichostatin A treatment, indicating a possible mechanism for the heterochromatization of the target LCR region. CONCLUSION: A combination of novel PLAP promoter and antibody based specificities has the potential for being developed as a possible therapeutic strategy for PLAP positive neoplasia.


Assuntos
Fosfatase Alcalina/genética , Inativação Gênica , Técnicas de Transferência de Genes , Isoenzimas/genética , Neoplasias/metabolismo , Regiões Promotoras Genéticas , Anticorpos de Cadeia Única/metabolismo , Virossomos/metabolismo , Apoptose , Caspases/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Metilação de DNA/genética , Fator de Transcrição E2F1/metabolismo , Elementos Facilitadores Genéticos/genética , Regulação Neoplásica da Expressão Gênica , Papillomavirus Humano 16/metabolismo , Humanos , Cinética , Proteínas Oncogênicas Virais/metabolismo , Proteínas E7 de Papillomavirus/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas Repressoras/metabolismo , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo
20.
PLoS One ; 10(6): e0128416, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26047143

RESUMO

BACKGROUND: Recently various studies have demonstrated the role of promoter associated non-coding RNAs (pRNA) in dsRNA induced transcriptional gene silencing and activation. However the exact mechanistic details of these processes with respect to the orientation of pRNAs are poorly defined. METHODOLOGY/PRINCIPAL FINDINGS: We have identified novel sense and antisense long control region (LCR) associated RNAs (pRNAs) in HPV18 positive cervical cancer cell lines HeLa, C-4 I and C-4 II. Using dsRNAs against these pRNAs, we were able to achieve upregulation or downregulation of the sense and antisense pRNAs and the downstream E6 and E7 oncogenes. We present evidence that knockdown of the sense pRNA is associated with reduction in E6 and E7 oncogenes and an upregulation of antisense pRNA. Conversely upregulation of sense pRNA is accompanied by an induction of the oncogenes and a concomitant reduction in antisense pRNA. Moreover, the exact role of sense and antisense pRNAs in dsRNA mediated gene modulation was confirmed by their selective degradation using antisense phosphorothioate oligodeoxynucleotides (ODN). Degradation of sense pRNA with antisense ODN led to loss of dsRNA induced silencing and activation, suggesting that dsRNA mediated gene modulation requires sense pRNA. Both processes were accompanied with congruent changes in the methylation pattern of activating and repressive histones. CONCLUSION/SIGNIFICANCE: Thus this data identifies and demonstrates the role of previously unknown important regulatory transcripts in HPV18 gene expression which can prove valuable targets in cervical cancer therapeutics. This mode of gene regulation by bidirectional transcription could be operational in other promoters as well and serve as a mechanism of regulating gene expression.


Assuntos
Inativação Gênica , Papillomavirus Humano 18/genética , RNA de Cadeia Dupla/metabolismo , Ativação Viral/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Epigênese Genética , Células HeLa , Humanos , Oligonucleotídeos Antissenso/metabolismo , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/metabolismo , Regiões Promotoras Genéticas , RNA de Cadeia Dupla/análise , RNA não Traduzido/antagonistas & inibidores , RNA não Traduzido/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...