Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
5.
Cells ; 13(11)2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38891080

RESUMO

With the lack of specific signs and symptoms, pancreatic ductal adenocarcinoma (PDAC) is often diagnosed at late metastatic stages, resulting in poor survival outcomes. Among various biomarkers, microRNA-21 (miR-21), a small non-coding RNA, is highly expressed in PDAC. By inhibiting regulatory proteins at the 3' untranslated regions (UTR), miR-21 holds significant roles in PDAC cell proliferation, epithelial-mesenchymal transition, angiogenesis, as well as cancer invasion, metastasis, and resistance therapy. We conducted a systematic search across major databases for articles on miR-21 and pancreatic cancer mainly published within the last decade, focusing on their diagnostic, prognostic, therapeutic, and biological roles. This rigorous approach ensured a comprehensive review of miR-21's multifaceted role in pancreatic cancers. In this review, we explore the current understandings and future directions regarding the regulation, diagnostic, prognostic, and therapeutic potential of targeting miR-21 in PDAC. This exhaustive review discusses the involvement of miR-21 in proliferation, epithelial-mesenchymal transition (EMT), apoptosis modulation, angiogenesis, and its role in therapy resistance. Also discussed in the review is the interplay between various molecular pathways that contribute to tumor progression, with specific reference to pancreatic ductal adenocarcinoma.


Assuntos
Transição Epitelial-Mesenquimal , MicroRNAs , Neoplasias Pancreáticas , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/metabolismo , Transição Epitelial-Mesenquimal/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/metabolismo , Regulação Neoplásica da Expressão Gênica , Proliferação de Células/genética , Apoptose/genética , Animais , Neovascularização Patológica/genética , Biomarcadores Tumorais/metabolismo , Biomarcadores Tumorais/genética , Prognóstico
6.
J Biol Chem ; 287(24): 20576-89, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22511755

RESUMO

Urokinase plasminogen activator receptor (uPAR) is known to promote invasion, migration, and metastasis in cancer cells. In this report, we showed that ionizing radiation (IR)-induced uPAR has a role in WNT-ß-catenin signaling and mediates induction of cancer stem cell (CSC)-like properties in medulloblastoma cell lines UW228 and D283. We observed that IR induced the expression of uPAR and CSC markers, such as Musashi-1 and CD44, and activated WNT-7a-ß-catenin signaling molecules. Overexpression of uPAR alone or with IR treatment led to increased WNT-7a-ß-catenin-TCF/LEF-mediated transactivation, thereby promoting cancer stemness. In contrast, treatment with shRNA specific for uPAR (pU) suppressed WNT-7a-ß-catenin-TCF/LEF-mediated transactivation both in vitro and in vivo. Quercetin, a potent WNT/ß-catenin inhibitor, suppressed uPAR and uPAR-mediated WNT/ß-catenin activation, and furthermore, addition of recombinant human WNT-7a protein induced uPAR, indicating the existence of a mutual regulatory relationship between uPAR and WNT/ß-catenin signaling. We showed that uPAR was physically associated with the WNT effector molecule ß-catenin on the membrane, cytoplasm, and nucleus of IR-treated cells and CSC. Most interestingly, we demonstrated for the first time that localization of uPAR in the nucleus was associated with transcription factors (TF) and their specific response elements. We observed from uPAR-ChIP, TF protein, and protein/DNA array analyses that uPAR associates with activating enhancer-binding protein 2α (AP2a) and mediates ß-catenin gene transcription. Moreover, association of uPAR with the ß-catenin·TCF/LEF complex and various other TF involved during embryonic development and cancer indicates that uPAR is a potent activator of stemness, and targeting of uPAR in combination with radiation has significant therapeutic implications.


Assuntos
Raios gama , Meduloblastoma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/efeitos da radiação , beta Catenina/metabolismo , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/patologia , Citoplasma/genética , Citoplasma/metabolismo , Citoplasma/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Meduloblastoma/genética , Meduloblastoma/patologia , Meduloblastoma/radioterapia , Camundongos , Transplante de Neoplasias , Células-Tronco Neoplásicas/patologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Fatores de Transcrição TCF/genética , Fatores de Transcrição TCF/metabolismo , Ativação Transcricional/genética , Ativação Transcricional/efeitos da radiação , Transplante Heterólogo , Proteínas Wnt/genética , beta Catenina/genética
7.
Angiogenesis ; 16(1): 85-100, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22956186

RESUMO

Despite existing aggressive treatment modalities, the prognosis for advanced stage neuroblastoma remains poor with significant long-term illness in disease survivors. Advance stage disease features are associated with tumor vascularity, and as such, angiogenesis inhibitors may prove useful along with current therapies. The matricellular protein, secreted protein acidic and rich in cysteine (SPARC), is known to inhibit proliferation and migration of endothelial cells stimulated by growth factors. Here, we sought to determine the effect of SPARC on neuroblastoma tumor cell-induced angiogenesis and to decipher the molecular mechanisms involved in angiogenesis inhibition. Conditioned medium from SPARC-overexpressed neuroblastoma cells (pSPARC-CM) inhibited endothelial tube formation, cell proliferation, induced programmed cell death and suppressed expression of pro-angiogenic molecules such as VEGF, FGF, PDGF, and MMP-9 in endothelial cells. Further analyses revealed that pSPARC-CM-suppressed expression of growth factors was mediated by inhibition of the Notch signaling pathway, and cells cultured on conditioned medium from tumor cells that overexpress both Notch intracellular domain (NICD-CM) and SPARC resumed the pSPARC-CM-suppressed capillary tube formation and growth factor expression in vitro. Further, SPARC overexpression in neuroblastoma cells inhibited neo-vascularization in vivo in a mouse dorsal air sac model. Furthermore, SPARC overexpression-induced endothelial cell death was observed by co-localization studies with TUNEL assay and an endothelial marker, CD31, in xenograft tumor sections from SPARC-overexpressed mice. Our data collectively suggest that SPARC overexpression induces endothelial cell apoptosis and inhibits angiogenesis both in vitro and in vivo.


Assuntos
Neovascularização Patológica/metabolismo , Neuroblastoma/irrigação sanguínea , Neuroblastoma/metabolismo , Osteonectina/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Indutores da Angiogênese/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Camundongos , Camundongos Nus , Neuroblastoma/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
8.
Mol Carcinog ; 52(10): 777-90, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22495828

RESUMO

Glioma is a highly complex brain tumor characterized by the dysregulation of proteins and genes that leads to tumor metastasis. Cathepsin B and uPAR are overexpressed in gliomas and they are postulated to play central roles in glioma metastasis. In this study, efficient downregulation of cathepsin B and uPAR by siRNA treatments significantly reduced glioma cell adhesion to laminin as compared to vitronectin, fibronectin, or collagen I in U251 and 4910 glioma cell lines. Brain glioma tissue array analysis showed high expression of CD151 in clinical samples when compared with normal brain tissue. Cathepsin B and uPAR siRNA treatment led to the downregulation of CD151 and laminin-binding integrins α3 and ß1. Co-immunoprecipitation experiments revealed that downregulation of cathepsin B and uPAR decreased the interaction of CD151 with uPAR cathepsin B, and α3ß1 integrin. Studies on the downstream signaling cascade of uPAR/CD151/α3ß1 integrin have shown that phosphorylation of FAK, SRC, paxillin, and expression of adaptor cytoskeletal proteins talin and vinculin were reduced with knockdown of cathepsin B, uPAR, and CD151. Treatment with the bicistronic construct reduced interactions between uPAR and CD151 as well as lowering α3ß1 integrin, talin, and vinculin expression levels in pre-established glioma tumors of nude mice. In conclusion, our results show that downregulation of cathepsin B and uPAR alone and in combination inhibit glioma cell adhesion by downregulating CD151 and its associated signaling molecules in vitro and in vivo. Taken together, the results of the present study show that targeting the uPAR-cathepsin B system has possible therapeutic potential.


Assuntos
Catepsina B/metabolismo , Adesão Celular , Movimento Celular , Glioma/patologia , Integrina alfa3beta1/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Tetraspanina 24/metabolismo , Animais , Apoptose , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Catepsina B/antagonistas & inibidores , Catepsina B/genética , Moléculas de Adesão Celular/metabolismo , Proliferação de Células , Regulação para Baixo , Citometria de Fluxo , Imunofluorescência , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Glioma/genética , Glioma/metabolismo , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Integrina alfa3beta1/antagonistas & inibidores , Integrina alfa3beta1/genética , Camundongos , Camundongos Nus , Fosforilação , RNA Interferente Pequeno/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Tetraspanina 24/antagonistas & inibidores , Tetraspanina 24/genética , Células Tumorais Cultivadas , Calinina
9.
J Neurooncol ; 107(1): 69-80, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21964739

RESUMO

Cathepsin B and urokinase plasminogen activator receptor (uPAR) are postulated to play key roles in glioma invasion. Calcineurin is one of the key regulators of mitochondrial-dependent apoptosis, but its mechanism is poorly understood. Hence, we studied subcellular localization of calcineurin after transcriptional downregulation of uPAR and cathepsin B in glioma. In the present study, efficient downregulation of uPAR and cathepsin B increased the translocation of calcineurin A from the mitochondria to the cytosol, decreased pBAD (S136) expression and its interaction with 14-3-3ζ and increased the interaction of BAD with Bcl-xl. Co-depletion of uPAR and cathepsin B induced mitochondrial translocation of BAD, activation of caspase 3 as well as PARP and cytochrome c and SMAC release. These effects were inhibited by FK506 (10 µM), a specific inhibitor of calcineurin. Calcineurin A was co-localized and also co-immunoprecipitated with Bcl-2. This interaction decreased with co-depletion of uPAR and cathepsin B and also with Bcl-2 inhibitor, HA 14-1 (20 µg/ml). Altered localization and interaction of calcineurin A with Bcl-2 was also observed in vivo when uPAR and cathepsin B were downregulated. In conclusion, downregulation of uPAR and cathepsin B induced apoptosis by targeting calcineurin A to BAD via Bcl-2 in glioma.


Assuntos
Apoptose , Calcineurina/metabolismo , Catepsina B/metabolismo , Glioma/metabolismo , Glioma/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteína de Morte Celular Associada a bcl/metabolismo , Western Blotting , Caspase 3/metabolismo , Catepsina B/antagonistas & inibidores , Linhagem Celular Tumoral , Citocromos c/metabolismo , Regulação para Baixo , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Potencial da Membrana Mitocondrial , Receptores de Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Transdução de Sinais , Frações Subcelulares
10.
Mol Cancer ; 10: 130, 2011 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-22004682

RESUMO

BACKGROUND: uPA/uPAR is a multifunctional system that is over expressed in many cancers and plays a critical role in glioblastoma (GBM) invasion. Previous studies from our lab have also shown that uPA/uPAR down regulation inhibits cancer cell invasion in SNB 19 GBM cells. METHODS: As Notch 1 is known to be over expressed and promotes invasion in glioblastoma, we therefore tested our hypothesis of whether down regulation of uPA/uPAR, singly or in tandem, attenuates GBM invasion via Notch 1 receptor. Targeted down regulation of uPA/uPAR, either singly or simultaneously, inhibited the anchorage independent growth of U251MG and GBM xenograft cell lines 4910 and 5310 as assessed by soft agar colony formation assay. Expression of all four Notch receptors was confirmed in GBM tissue array analysis by immunohistochemistry. RESULTS: Down regulation of uPA/uPAR, either singly or simultaneously, in U251 MG and tumor xenografts inhibited the cleavage of the Notch receptor between the Gly 1743 and Val 1744 positions, thereby suggesting inhibition of activated cytosolic fragment-related Notch gene transcription. Morphological analysis confirmed inhibition of NICD when U251 MG cells were treated with puPA, puPAR or pU2. uPA/uPAR down regulation inhibited Notch 1 mRNA in all three examined cell lines. uPA/uPAR shRNA down regulated nuclear activation of NF-κB subunits and phosphorylation of AKT/mTOR pathway in U251 MG and GBM xenografts. puPA down regulated NICD and HES induced phosphorylation of AKT/ERK and NF-κB. Down regulation of Notch 1 using siRNA inhibited uPA activity as shown by fibrinogen zymography. It also decreased uPA expression levels as shown by western blotting. Exogenous addition of uPA activated Notch 1 in uPAR antisense U251 MG cells and also in uPAR antisense cells transfected with siRNA against Delta and Jagged. The Notch 1 receptor co-localized with LAMP-1, a marker for lysosomes in uPA, uPAR and U2, down regulated U251 MG cells which probably indicates inhibition of Notch 1 receptor trafficking in GBM cells. Notch 1 expression was significantly inhibited in puPA- and pU2-treated pre-established intracranial tumors in mice. CONCLUSIONS: Overall our results show that down regulation of uPA/uPAR, either singly or simultaneously, could be an effective approach to attenuate Notch 1 receptor cleavage, signaling and endosomal trafficking in U251MG cells and xenografts, and ultimately inhibiting GBM invasion.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Transporte Proteico , Receptor Notch1/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Neoplasias Encefálicas/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Expressão Gênica , Técnicas de Silenciamento de Genes , Glioblastoma/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína Jagged-1 , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Invasividade Neoplásica , Transplante de Neoplasias , Estrutura Terciária de Proteína , Interferência de RNA , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Proteínas Serrate-Jagged , Transdução de Sinais , Análise Serial de Tecidos , Ativação Transcricional , Ativador de Plasminogênio Tipo Uroquinase/genética
11.
Oncol Lett ; 21(1): 75, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33365086

RESUMO

Angiogenesis is a tightly regulated biological process by which new blood vessels are formed from pre-existing blood vessels. This process is also critical in diseases such as cancer. Therefore, angiogenesis has been explored as a drug target for cancer therapy. The future of effective anti-angiogenic therapy lies in the intelligent combination of multiple targeting agents with novel modes of delivery to maximize therapeutic effects. Therefore, a novel approach is proposed that utilizes dumbbell RNA (dbRNA) to target pathological angiogenesis by simultaneously targeting multiple molecules and processes that contribute to angiogenesis. In the present study, a plasmid expressing miR-34a-3p and -5p dbRNA (db34a) was constructed using the permuted intron-exon method. A simple protocol to purify dbRNA from bacterial culture with high purity was also developed by modification of the RNASwift method. To test the efficacy of db34a, pancreatic cancer cell lines PANC-1 and MIA PaCa-2 were used. Functional validation of the effect of db34a on angiogenesis was performed on human umbilical vein endothelial cells using a tube formation assay, in which cells transfected with db34a exhibited a significant reduction in tube formation compared with cells transfected with scrambled dbRNA. These results were further validated in vivo using a zebrafish angiogenesis model. In conclusion, the present study demonstrates an approach for blocking angiogenesis using db34a. The data also show that this approach may be used to targeting multiple molecules and pathways.

12.
BMC Cancer ; 10: 647, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21106094

RESUMO

BACKGROUND: uPAR and MMP-9, which play critical roles in tumor cell invasion, migration and angiogenesis, have been shown to be associated with lipid rafts. METHODS: To investigate whether cholesterol could regulate uPAR and MMP-9 in breast carcinoma, we used MßCD (methyl beta cyclodextrin, which extracts cholesterol from lipid rafts) to disrupt lipid rafts and studied its effect on breast cancer cell migration, invasion, angiogenesis and signaling. RESULTS: Morphological evidence showed the association of uPAR with lipid rafts in breast carcinoma cells. MßCD treatment significantly reduced the colocalization of uPAR and MMP-9 with lipid raft markers and also significantly reduced uPAR and MMP-9 at both the protein and mRNA levels. Spheroid migration and invasion assays showed inhibition of breast carcinoma cell migration and invasion after MßCD treatment. In vitro angiogenesis studies showed a significant decrease in the angiogenic potential of cells pretreated with MßCD. MßCD treatment significantly reduced the levels of MMP-9 and uPAR in raft fractions of MDA-MB-231 and ZR 751 cells. Phosphorylated forms of Src, FAK, Cav, Akt and ERK were significantly inhibited upon MßCD treatment. Increased levels of soluble uPAR were observed upon MßCD treatment. Cholesterol supplementation restored uPAR expression to basal levels in breast carcinoma cell lines. Increased colocalization of uPAR with the lysosomal marker LAMP1 was observed in MßCD-treated cells when compared with untreated cells. CONCLUSION: Taken together, our results suggest that cholesterol levels in lipid rafts are critical for the migration, invasion, and angiogenesis of breast carcinoma cells and could be a critical regulatory factor in these cancer cell processes mediated by uPAR and MMP-9.


Assuntos
Neoplasias da Mama/enzimologia , Movimento Celular , Células Endoteliais/enzimologia , Metaloproteinase 9 da Matriz/metabolismo , Microdomínios da Membrana/enzimologia , Neovascularização Patológica/enzimologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Caveolinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Colesterol/deficiência , Meios de Cultivo Condicionados/metabolismo , Relação Dose-Resposta a Droga , Células Endoteliais/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Quinase 1 de Adesão Focal/metabolismo , Regulação da Expressão Gênica , Humanos , Metaloproteinase 9 da Matriz/genética , Microdomínios da Membrana/efeitos dos fármacos , Invasividade Neoplásica , Neovascularização Patológica/patologia , Nistatina/farmacologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Fatores de Tempo , beta-Ciclodextrinas/farmacologia , Quinases da Família src/metabolismo
13.
J Cell Physiol ; 220(2): 285-91, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19391103

RESUMO

In vivo gene silencing using RNAi plays an important role in target validation and is advancing towards the development of RNAi-based therapeutics. RNAs were thought to have just two broad functions in cells as messenger RNAs (mRNAs) and ribosomal RNAs, but recently the relevance of microRNAs is becoming more clearly understood. mRNA molecules transmit information between DNA and protein and, as such, are vital intermediaries for gene expression. Ribosomal and transfer RNAs have structural, catalytic, and information-decoding roles in the process of protein synthesis, whereas microRNAs are regulators of gene expression. This review presents the early and intriguing successes of using siRNAs for in vivo gene silencing and its use as a possible cancer therapeutics.


Assuntos
Inativação Gênica , Técnicas de Transferência de Genes , Terapia Genética/métodos , Neoplasias/terapia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Animais , Ensaios Clínicos como Assunto , Humanos , Lipídeos/química , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias/genética , RNA Interferente Pequeno/genética
14.
Biochem Biophys Res Commun ; 386(2): 386-91, 2009 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-19524549

RESUMO

We have assessed the capacity of human umbilical cord blood (hUCB)-derived stem cells to differentiate into cardiomyocytes and repair angiotensin II induced insult in culture and in mouse hearts when injected. hUCB were able to differentiate into cardiomyocyte-like cells, when induced with 5-azacytidine or co-cultured with rat neonatal cardiomyocytes (NRCM). When co-cultured, hUCB reversed the pathological effects induced by angiotensin II (Ang-II) in NRCM and in mice injected after Ang-II infusion. As assessed by increased heart weight to body mass ratio and Ang-II-induced fibrosis, cardiac hypertrophy was also reduced after hUCB were injected. hUCB also reversed the pathological heart failure markers induced by Ang-II in mice. Further, we observed a shift from pathological hypertrophy towards physiological hypertrophy by hUCB in Ang-II-challenged mice. Our findings support hUCB as a feasible model for experimentation in stem cell therapy and emphasize the relevance of the hUCB in reversing heart failure conditions.


Assuntos
Cardiomegalia/terapia , Sangue Fetal/citologia , Sangue Fetal/transplante , Miócitos Cardíacos/transplante , Transplante de Células-Tronco , Células-Tronco/fisiologia , Angiotensina II/farmacologia , Animais , Apoptose , Biomarcadores/metabolismo , Cardiomegalia/induzido quimicamente , Cardiomegalia/patologia , Diferenciação Celular , Técnicas de Cocultura , Antígenos de Histocompatibilidade Classe I/biossíntese , Humanos , Camundongos , Miócitos Cardíacos/fisiologia , Ratos
15.
Int J Oncol ; 35(2): 257-63, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19578738

RESUMO

The epidermal growth factor receptor (EGFR) family (also known as the ErbB protein family) is comprised of four structurally-related receptor tyrosine kinases. Insufficient ErbB signaling in humans is associated with the development of neurodegenerative diseases, such as multiple sclerosis and Alzheimer's disease. In contrast, excessive ErbB signaling is associated with the development of a wide variety of solid tumors. ErbB-1 and -2 are found in many human cancers and their excessive signaling may be critical factors in the development and malignancy of solid tumors. Several molecular strategies have been developed recently to modulate either EGFR or the downstream signal beyond the cell surface receptor. In the present study, we used human EGFR-overexpressing glioma xenograft cells 4910 and 5310 and targeted MMP-2 expression using an adenoviral RNAi construct. We observed that the RNAi-mediated downregulation of MMP-2 causes the upregulation of ErbB-2 in certain EGFR-overexpressing glioma xenograft cells both in vitro and in vivo. Targeted MMP-2 downregulation was observed in a dose-dependent manner with no apparent off-target effects in these xenograft cells. We also noted that the overexpression of ErbB-2 induced by MMP-2 downregulation is consistent with p50-mediated cell death in 5310 cells but not in 4910 cells. In addition, APAF-1 expression levels increased in correlation with increased ErbB-2 expression after MMP-2 downregulation in vitro and in vivo. Our results suggest that MMP-2 may play a role in a hitherto unknown signaling pathway mediated via ErbB-2 in certain cancer cell types.


Assuntos
Apoptose , Glioma/patologia , Metaloproteinase 2 da Matriz/fisiologia , Receptor ErbB-2/fisiologia , Animais , Fator Apoptótico 1 Ativador de Proteases/metabolismo , Caspase 8/metabolismo , Linhagem Celular Tumoral , Humanos , Inibidores de Metaloproteinases de Matriz , Camundongos , Subunidade p50 de NF-kappa B/metabolismo , Transplante de Neoplasias , Interferência de RNA , Transplante Heterólogo
16.
Methods Mol Biol ; 487: 267-81, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19301652

RESUMO

Targeting proteases and their activators would retard the invasive ability of cancer cells, and has been shown to induce apoptosis in certain instances. Various methods have been developed to specifically target protease molecules in an attempt to retard invasion and migration. Of these methods, RNA interference (RNAi) holds great therapeutic potential. RNAi technology is now being used to target specific molecules for use as potential anti-cancer agents. RNAi-mediated silencing is almost catalytic when compared to anti-sense silencing. Of these targets, the uPAR-uPA system and MMPs holds great promise. Targeting uPA/uPAR may provide additive or synergistic treatment benefits if used in combination with conventional therapeutics such as chemotherapy or radiation. Studies point to the fact that specifically targeting MMP-9 or MMP-2 singly or in combination with other proteases could have specific therapeutic implications in the treatment of cancer. In this chapter we discuss the therapeutic potential of siRNA-mediated targeting of the uPAR-uPA system and MMPs as therapeutic agents for the treatment of cancer.


Assuntos
Metaloproteinases da Matriz/genética , Neoplasias/terapia , Interferência de RNA , RNA Interferente Pequeno/uso terapêutico , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/genética , Animais , Regulação da Expressão Gênica , Terapia Genética , Humanos , Neoplasias/genética
17.
Int J Oncol ; 33(4): 783-90, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18813792

RESUMO

We have previously demonstrated the effectiveness of simultaneous RNA interference (RNAi)-mediated downregulation of urokinase-type plasminogen activator receptor (uPAR) and matrix metalloproteinase-9 (MMP-9) in inhibiting tumor invasion in vitro and in vivo. In particular, we have shown that the downregulation of uPAR and MMP-9 inhibits intracranial tumor growth. The mechanism of the inhibition of tumor growth has not yet been determined. In this study, we have attempted to explain the mechanisms involved in the inhibition of invasiveness and tumor growth in vitro. SNB19 glioma cells were transfected with scrambled vector plasmid (pSV) and a siRNA-expressing plasmid targeting either uPAR (pU) or MMP-9 (pM) singly or in combination (pUM). Untransfected cells were also used as a control. Western blotting and RT-PCR analyses showed the downregulation of uPAR in pU-transfected cells and MMP-9 in pM-transfected cells. In cells transfected with pUM, we observed down-regulation of both uPAR and MMP-9, thereby indicating the specificity of the siRNA-expressing plasmids. An increase in caspase 9 expression was observed in cells transfected with pUM whereas no change in the level of caspase 9 was observed in pU or pM-transfected cells. Additionally, no change in the expression level of caspase 8 was observed. However, an increase in the expression level of cleaved PARP was observed in the case of cells transfected with pU, pM and pUM. Cells transfected with pUM showed the highest levels of cleaved PARP expression. Expression levels of APAF-1 were also higher in pUM-transfected cells with no change in expression levels of controls and in pU and pM-transfected cells. Total CAD expression levels did not change under any of the transfection conditions. However, immunohistochemical studies demonstrated that CAD was translocated to the nucleus, thereby indicating DNA damage. As determined by Western blot analysis of subcellular fractions, cytoplasmic levels of cytochrome c were also increased. We determined the extent of DNA damage using the TUNEL assay (poly-A termination of free -OH ends of degraded nuclear DNA). Based on our results we conclude that the simultaneous downregulation of uPAR and MMP-9 induces apoptosome-mediated apoptosis.


Assuntos
Apoptose , Neoplasias Encefálicas/metabolismo , Caspase 9/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Metaloproteinase 9 da Matriz/biossíntese , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Receptores de Superfície Celular/biossíntese , Citosol/metabolismo , Fragmentação do DNA , Ativação Enzimática , Humanos , Mitocôndrias/metabolismo , Modelos Biológicos , Receptores de Ativador de Plasminogênio Tipo Uroquinase
18.
Int J Oncol ; 33(5): 937-47, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18949356

RESUMO

Meningioma is a well-known tumor of the central nervous system, and is treated by surgical resection and/or radiation. Recently, ionizing radiation has been shown to enhance invasiveness of surviving tumor cells, and several proteolytic enzyme molecules, including urokinase plasminogen activator (uPA), seem to be upregulated after radiation. uPA and its receptor (uPAR) have been strongly implicated in tumor invasion, angiogenesis and progression. Hence, the tumor-associated uPA-uPAR system is considered a potential target for cancer therapy. In the present study, we show that radiation increases uPA levels in the IOMM-Lee meningioma cells, and subsequently, increases tumor invasion, migration and angiogenesis in vitro. Studies with signaling molecule inhibitors AG1478, U0126 and SB203580 (specific inhibitors of EGFR, MEK1/2 and p38 respectively) showed inhibition of uPA levels in both basal and irradiated-IOMM-Lee cells. The PI3K inhibitor (LY294002) and the AKT inhibitor (AKT inhibitor IV) also partially decreased uPA expression, whereas SP600125, a JNK inhibitor, did not affect uPA levels in either radiated or non-radiated cells. Further, a bicistronic plasmid construct with small interfering RNA (siRNA) against uPA and its receptor inhibited tumor invasion, migration and angiogenesis in radiation-treated IOMM-Lee cells. In addition, siRNA against uPA and its receptor inhibited subcutaneous tumor growth in athymic nude mice in combination with radiation in a synergistic manner. Thus, the specific targeting of proteases via RNA interference could augment the therapeutic effect of radiation and prevent the adverse effects resulting from tumor cells that receive sublethal doses of radiation within the tumor mass.


Assuntos
Movimento Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Terapia Genética/métodos , Neoplasias Meníngeas/terapia , Meningioma/terapia , Neovascularização Patológica/prevenção & controle , Interferência de RNA , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Capilares/enzimologia , Capilares/efeitos da radiação , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Neoplasias Meníngeas/enzimologia , Neoplasias Meníngeas/patologia , Neoplasias Meníngeas/radioterapia , Meningioma/enzimologia , Meningioma/patologia , Meningioma/radioterapia , Camundongos , Camundongos Nus , Invasividade Neoplásica , Neovascularização Patológica/enzimologia , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno/metabolismo , Radioterapia/efeitos adversos , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Transfecção , Ativador de Plasminogênio Tipo Uroquinase/genética , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Clin Cancer Res ; 13(12): 3507-17, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17575213

RESUMO

PURPOSE: The induction of apoptotic pathways in cancer cells offers a novel and potentially useful approach to improve patient responses to conventional chemotherapy. Tissue factor pathway inhibitor-2 (TFPI-2) is a protease inhibitor that is abundant in the extracellular matrix and highly expressed in noninvasive cells but absent or undetectable in highly invasive human glioblastoma cells. EXPERIMENTAL DESIGN: Using a recombinant adeno-associated viral vector carrying human TFPI-2 cDNA, we stably expressed TFPI-2 in U-251 cells, a highly invasive human glioblastoma cell line. Our previous studies showed that restoration of TFPI-2 in glioblastomas effectively prevents cell proliferation, angiogenesis, and tumor invasion. In this study, we determined whether TFPI-2 restoration could induce apoptosis through the caspase-mediated signaling pathway. RESULTS: The results from nuclear chromatin staining, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay, and fluorescence-activated cell sorting analysis showed increased apoptosis in U-251 cells after restoration of TFPI-2. Caspase-9 and caspase-3 activity assays showed increased activity, indicating enhanced apoptosis. Immunofluorescence for cleaved caspase-9 and caspase-3 depicted increased expression and colocalization of both molecules. Western blot analysis showed increased transcriptional activities of Fas ligand, tumor necrosis factor-alpha, Bax, Fas-associated death domain, and tumor necrosis factor receptor 1-associated death domain as well as elevated levels of cleaved caspases and poly(ADP-ribose) polymerase. Semiquantitative reverse transcription-PCR depicted increased expression of tumor necrosis factor-alpha and Fas ligand and the related death domains tumor necrosis factor receptor 1-associated death domain and Fas-associated death domain. CONCLUSIONS: Taken together, these results show that restoration of TFPI-2 activates both intrinsic and extrinsic caspase-mediated, proapoptotic signaling pathways and induces apoptosis in U-251 cells. Furthermore, our study suggests that recombinant adeno-associated viral vector-mediated gene expression offers a novel tool for cancer gene therapy.


Assuntos
Apoptose/fisiologia , Neoplasias Encefálicas/metabolismo , Terapia Genética/métodos , Glioblastoma/metabolismo , Glicoproteínas/metabolismo , Adenoviridae/genética , Western Blotting , Neoplasias Encefálicas/genética , Caspases/metabolismo , Linhagem Celular Tumoral , Citometria de Fluxo , Imunofluorescência , Expressão Gênica , Vetores Genéticos , Glioblastoma/genética , Glicoproteínas/genética , Humanos , Marcação In Situ das Extremidades Cortadas , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Clin Cancer Res ; 13(14): 4051-60, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17634529

RESUMO

PURPOSE: The purpose of this study was to evaluate the therapeutic potential of using plasmid-expressed RNA interference (RNAi) targeting urokinase-type plasminogen activator (uPA) receptor (uPAR) and uPA to treat human glioma. EXPERIMENTAL DESIGN: In the present study, we have used plasmid-based RNAi to simultaneously down-regulate the expression of uPAR and uPA in SNB19 glioma cell lines and epidermal growth factor receptor (EGFR)--overexpressing 4910 human glioma xenografts in vitro and in vivo, and evaluate the i.p. route for RNAi-expressing plasmid administered to target intracranial glioma. RESULTS: Plasmid-mediated RNAi targeting uPAR and uPA did not induce OAS1 expression as seen from reverse transcription-PCR analysis. In 4910 EGFR-overexpressing cells, down-regulation of uPAR and uPA induced the down-regulation of EGFR and vascular endothelial growth factor and inhibited angiogenesis in both in vitro and in vivo angiogenic assays. In addition, invasion and migration were inhibited as indicated by in vitro spheroid cell migration, Matrigel invasion, and spheroid invasion assays. We did not observe OAS1 expression in mice with preestablished intracranial tumors, which were given i.p. injections of plasmid-expressing small interfering RNA--targeting uPAR and uPA. Furthermore, the small interfering RNA plasmid targeting uPAR and uPA caused regression of preestablished intracranial tumors when compared with the control mice. CONCLUSION: In conclusion, the plasmid-expressed RNAi targeting uPAR and uPA via the i.p. route has potential clinical applications for the treatment of glioma.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Neoplasias Encefálicas/patologia , Plasmídeos , Interferência de RNA , Receptores de Superfície Celular/genética , Ativador de Plasminogênio Tipo Uroquinase/genética , Animais , Neoplasias Encefálicas/prevenção & controle , Linhagem Celular Tumoral , Glioma/prevenção & controle , Humanos , Injeções Intraperitoneais , Camundongos , Camundongos Nus , Transplante de Neoplasias , RNA Interferente Pequeno/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA