Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Int J Cancer ; 134(4): 755-64, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23907728

RESUMO

Studies on the low-abundance transcriptome are of paramount importance for identifying the intimate mechanisms of tumor progression that can lead to novel therapies. The aim of the present study was to identify novel markers and targetable genes and pathways in advanced human gastric cancer through analyses of the low-abundance transcriptome. The procedure involved an initial subtractive hybridization step, followed by global gene expression analysis using microarrays. We observed profound differences, both at the single gene and gene ontology levels, between the low-abundance transcriptome and the whole transcriptome. Analysis of the low-abundance transcriptome led to the identification and validation by tissue microarrays of novel biomarkers, such as LAMA3 and TTN; moreover, we identified cancer type-specific intracellular pathways and targetable genes, such as IRS2, IL17, IFNγ, VEGF-C, WISP1, FZD5 and CTBP1 that were not detectable by whole transcriptome analyses. We also demonstrated that knocking down the expression of CTBP1 sensitized gastric cancer cells to mainstay chemotherapeutic drugs. We conclude that the analysis of the low-abundance transcriptome provides useful insights into the molecular basis and treatment of cancer.


Assuntos
Adenocarcinoma/genética , Biomarcadores Tumorais/genética , Mucosa Gástrica/metabolismo , Perfilação da Expressão Gênica , Neoplasias Gástricas/genética , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Oxirredutases do Álcool/antagonistas & inibidores , Oxirredutases do Álcool/genética , Oxirredutases do Álcool/metabolismo , Antineoplásicos/farmacologia , Biomarcadores Tumorais/metabolismo , Western Blotting , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Conectina/genética , Conectina/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Técnicas Imunoenzimáticas , Laminina/genética , Laminina/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia , Técnica de Subtração , Análise Serial de Tecidos , Células Tumorais Cultivadas
2.
J Cell Biochem ; 105(2): 381-90, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18553460

RESUMO

Survivin is recognized as an attractive target in cancer therapy because of its selective overexpression in the majority of tumors. Upregulated expression of this protein correlates with increased tumor grade, recurrence risk and decreased cancer patients survival. In this study, we assessed the efficacy of two survivin-specific small interfering RNA (siRNA) constructs to inhibit T47D human breast cancer cell growth. After siRNA transfection, T47D cells showed a significant reduction in proliferation and survival exhibiting clear signs of apoptosis. pSil_1 that targeted exon 1 exhibited a stronger inhibitory effect on cell growth, and increased cell apoptosis compared to pSil_30 that targeted exon 4. Cell apoptosis was found to be mediated by translocation of the mitochondrial apoptosis inducing factor (AIF), while no changes were observed in caspase-3 activation and Bid cleavage. Thus, silencing survivin expression using siRNA strategies represents a suitable therapeutic approach to selectively modulate the survival and growth of human breast cancer cells.


Assuntos
Apoptose , Neoplasias da Mama/patologia , Inativação Gênica/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/genética , Proteínas de Neoplasias/genética , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Caspases , Feminino , Humanos , Proteínas Inibidoras de Apoptose , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/farmacologia , Survivina , Transfecção
3.
Int J Cancer ; 122(7): 1465-75, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18059024

RESUMO

Cell interaction with the extracellular matrix (ECM) has profound influence in cancer progression. The secreted protein, acidic and rich in cysteine (SPARC) a component of the ECM, impairs the proliferation of different cell types and modulates tumor cell aggressive features. This apparent paradox might result either from the biochemical properties of the different SPARC sources or from differential responses of malignant and stromal cells to SPARC. To test these hypotheses, we purified SPARC secreted by melanoma cells (hMel-SPARC) and compared its activity with different recombinant SPARC preparations, including a new one produced in insect cells. All 5 SPARC species were effective in inhibiting bovine aortic endothelial cell proliferation, adhesion and migration. We then used the melanoma-derived protein to assess SPARC effect on additional cell types. hMel-SPARC greatly impaired the proliferation of both normal and transformed human endothelial cells and exerted a moderate biphasic effect on human fetal fibroblasts proliferation, irrespective of their endogenous SPARC levels. However, SPARC had no effect on the proliferation of several human cancer cell lines regardless of their endogenous levels of SPARC expression. Importantly, downregulation of SPARC levels in melanoma cells using either an antisense RNA or a shRNA against SPARC sensitized them to hMel-SPARC addition in proliferation and migration assays, suggesting that malignant cells developed a SPARC-resistance mechanism. This was not a general resistance to growth suppressing agents, as melanoma cells with restricted SPARC expression were more resistant to chemotherapeutic agents. Thus, malignant cells expressing or not expressing SPARC developed alternative mechanisms that, in contrary to stromal cells, rendered them SPARC-insensitive.


Assuntos
Matriz Extracelular/metabolismo , Melanoma/metabolismo , Osteonectina/metabolismo , Células Estromais/metabolismo , Aorta , Movimento Celular , Proliferação de Células , Regulação para Baixo , Endotélio Vascular/metabolismo , Fibroblastos , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/patologia , Osteonectina/genética , RNA Antissenso , RNA Interferente Pequeno , Proteínas Recombinantes/metabolismo , Células Estromais/patologia
4.
J Gene Med ; 10(9): 993-1004, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18615449

RESUMO

BACKGROUND: The interaction between fibrogenic cells and extracellular matrix plays a role in liver fibrosis, yet the mechanisms are largely unknown. Secreted protein, acidic and rich in cysteine (SPARC) is a matricellular glycoprotein that is expressed by hepatic stellate cells and is overexpressed in fibrotic livers. We investigated the in vivo role of SPARC in experimentally induced liver fibrosis in rats. METHODS: A recombinant adenovirus carrying antisense SPARC was constructed (AdasSPARC). Advanced liver fibrosis was induced in Sprague-Dawley rats by prolonged intraperitoneal administration of thioacetamide. Animals received injections of AdasSPARC or Ad beta gal (control adenovirus) via the tail vein and directly into the liver 1 week after the first dose. The pathological changes in liver tissues and indices of fibrosis were assessed at eight weeks. Expression of SPARC, transforming growth factor (TGF)-beta and alpha-smooth muscle actin were evaluated by quantitative real-time polymerase chain reaction, western blotting, enzyme-linked immunosorbent assay and immunohistochemistry. RESULTS: Hepatic SPARC expression significantly increased during the development of liver fibrosis. AdasSPARC markedly attenuated the development of hepatic fibrosis in rats treated with thiocetamide, as assessed by decreased collagen deposition, lower hepatic content of hydroxyproline and less advanced morphometric stage of fibrosis. AdasSPARC treatment reduced inflammatory activity (Knodell score) and suppressed transdifferentiation of hepatic stellate cell to the myofibroblasts like phenotype in vivo. Furthermore, in vitro inhibition of SPARC on hepatic stellate cells decreases the production of TGF-beta. CONCLUSIONS: This is the first study to demonstrate that knockdown of hepatic SPARC expression ameliorates thioacetamide-induced liver fibrosis in rats with chronic liver injury. SPARC is a potential target for gene therapy in liver fibrosis.


Assuntos
Adenoviridae/genética , Cirrose Hepática Experimental/terapia , Osteonectina/antagonistas & inibidores , Actinas/genética , Actinas/metabolismo , Adenoviridae/metabolismo , Animais , Células Cultivadas , Terapia Genética , Humanos , Imuno-Histoquímica , Cirrose Hepática Experimental/genética , Cirrose Hepática Experimental/metabolismo , Neoplasias de Tecido Muscular/metabolismo , Osteonectina/genética , Osteonectina/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Tioacetamida/toxicidade , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
5.
Cancer Res ; 65(12): 5123-32, 2005 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15958556

RESUMO

The expression of secreted protein acidic and rich in cysteine (SPARC) has been associated with the malignant progression of different types of human cancer. SPARC was associated with tumor cell capacity to migrate and invade, although its precise role in tumor progression is still elusive. In the present study, we show that SPARC produced by melanoma cells modulates the antitumor activity of polymorphonuclear leukocytes (PMN). Administration to nude mice of human melanoma cells in which SPARC expression was transiently or stably knocked down by antisense RNA (SPARC-sup cells) promoted PMN recruitment and obliterated tumor growth even when SPARC-sup cells accounted for only 10% of injected malignant cells. In addition, SPARC-sup cells stimulated the in vitro migration and triggered the antimelanoma cytotoxic capacity of human PMN, an effect that was reverted in the presence of SPARC purified from melanoma cells or by reexpressing SPARC in SPARC-sup cells. Leukotrienes, interleukin 8, and growth-related oncogene, in combination with Fas ligand and interleukin 1, mediated SPARC effects. These data indicate that SPARC plays an essential role in tumor evasion from immune surveillance through the inhibition of the antitumor PMN activity.


Assuntos
Proteínas de Transporte/imunologia , Melanoma/imunologia , Neutrófilos/imunologia , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Proteína Ligante Fas , Humanos , Interleucina-1/imunologia , Melanoma/metabolismo , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , RNA Antissenso/genética , Transplante Heterólogo
6.
Redox Biol ; 11: 38-50, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27863297

RESUMO

Infection with oncogenic human papillomavirus induces deregulation of cellular redox homeostasis. Virus replication and papillomavirus-induced cell transformation require persistent expression of viral oncoproteins E7 and E6 that must retain their functionality in a persistent oxidative environment. Here, we dissected the molecular mechanisms by which E7 oncoprotein can sense and manage the potentially harmful oxidative environment of the papillomavirus-infected cell. The carboxy terminal domain of E7 protein from most of the 79 papillomavirus viral types of alpha genus, which encloses all the tumorigenic viral types, is a cysteine rich domain that contains two classes of cysteines: strictly conserved low reactive Zn+2 binding and degenerate reactive cysteine residues that can sense reactive oxygen species (ROS). Based on experimental data obtained from E7 proteins from the prototypical viral types 16, 18 and 11, we identified a couple of low pKa nucleophilic cysteines that can form a disulfide bridge upon the exposure to ROS and regulate the cytoplasm to nucleus transport. From sequence analysis and phylogenetic reconstruction of redox sensing states we propose that reactive cysteine acquisition through evolution leads to three separate E7s protein families that differ in the ROS sensing mechanism: non ROS-sensitive E7s; ROS-sensitive E7s using only a single or multiple reactive cysteine sensing mechanisms and ROS-sensitive E7s using a reactive-resolutive cysteine couple sensing mechanism.


Assuntos
Cisteína/metabolismo , Neoplasias/genética , Estresse Oxidativo/genética , Proteínas E7 de Papillomavirus/metabolismo , Nucléolo Celular/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Cisteína/genética , Citoplasma/metabolismo , Dissulfetos/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Oxirredução , Proteínas E7 de Papillomavirus/genética , Transporte Proteico/genética , Replicação Viral/genética
7.
Mol Cancer Res ; 15(3): 304-316, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28031408

RESUMO

Understanding the mechanism of metastatic dissemination is crucial for the rational design of novel therapeutics. The secreted protein acidic and rich in cysteine (SPARC) is a matricellular glycoprotein which has been extensively associated with human breast cancer aggressiveness although the underlying mechanisms are still unclear. Here, shRNA-mediated SPARC knockdown greatly reduced primary tumor growth and completely abolished lung colonization of murine 4T1 and LM3 breast malignant cells implanted in syngeneic BALB/c mice. A comprehensive study including global transcriptomic analysis followed by biological validations confirmed that SPARC induces primary tumor growth by enhancing cell cycle and by promoting a COX-2-mediated expansion of myeloid-derived suppressor cells (MDSC). The role of SPARC in metastasis involved a COX-2-independent enhancement of cell disengagement from the primary tumor and adherence to the lungs that fostered metastasis implantation. Interestingly, SPARC-driven gene expression signatures obtained from these murine models predicted the clinical outcome of patients with HER2-enriched breast cancer subtypes. In total, the results reveal that SPARC and its downstream effectors are attractive targets for antimetastatic therapies in breast cancer.Implications: These findings shed light on the prometastatic role of SPARC, a key protein expressed by breast cancer cells and surrounding stroma, with important consequences for disease outcome. Mol Cancer Res; 15(3); 304-16. ©2016 AACR.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Osteonectina/metabolismo , Receptor ErbB-2/metabolismo , Animais , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , Células MCF-7 , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Osteonectina/genética , Prognóstico , Receptor ErbB-2/genética , Resultado do Tratamento
8.
PLoS One ; 10(8): e0134714, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26248315

RESUMO

Cell transition to a more aggressive mesenchymal-like phenotype is a hallmark of cancer progression that involves different steps and requires tightly regulated cell plasticity. SPARC (Secreted Protein Acidic and Rich in Cysteine) is a matricellular protein that promotes this transition in various malignant cell types, including melanoma cells. We found that suppression of SPARC expression in human melanoma cells compromised cell migration, adhesion, cytoskeleton structure, and cell size. These changes involved the Akt/mTOR pathway. Re-expression of SPARC or protein addition restored all the cell features. Suppression of SPARC expression was associated with increased Rac1-GTP levels and its membrane localization. Expression of the dominant negative mutant of Rac1 counteracted almost all the changes observed in SPARC-deficient cells. Overall, these data suggest that most of the SPARC-mediated effects occurred mainly through the blockade of Rac1 activity.


Assuntos
Plasticidade Celular , Osteonectina/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Citoesqueleto de Actina/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Regulação para Baixo , Humanos , Integrinas/química , Integrinas/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Osteonectina/química , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Proteínas rac1 de Ligação ao GTP/química
9.
Clin Cancer Res ; 21(7): 1665-74, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25573380

RESUMO

PURPOSE: We decided to construct a novel oncolytic adenovirus whose replication was driven by the CDC25B promoter for its use in preclinical models of pancreatic cancer. EXPERIMENTAL DESIGN: We placed the essential E1A gene under control of the CDC25B promoter. Based on preliminary data, we pseudotyped the adenovirus with a chimeric fiber of serotypes 5/3. We investigated the in vitro lytic effect and the in vivo therapeutic efficacy in combination with gemcitabine on human pancreatic tumor xenografts orthotopically growing in nude mice and in tumors growing in Syrian hamsters. We also assessed biochemical markers of hepatic toxicity and CA19.9 levels. RESULTS: AV25CDC exhibited a strong in vitro lytic effect on pancreatic cancer cells. In vivo administration of AV25CDC combined with gemcitabine in mice harboring subcutaneously growing SW1990 pancreatic tumors almost abrogated tumor growth. Nude mice harboring 15-day-old orthotopic tumors, treated intratumorally or systemically with AV25CDC combined with gemcitabine, exhibited 70% to 80% reduction in tumor size compared with control mice that lasted for at least 60 days. Chemovirotherapy treatment induced a return to normal levels of biochemical parameters of hepatic toxicity; these mice exhibited more than 90% reduction in CA19.9 serum levels compared with control. Chemovirotherapy efficacy was confirmed in mice harboring Mia PaCa-2 tumors and in Syrian hamster harboring HaP-T1 tumors. We observed that viral treatment disrupted tumor architecture and induced an increase in MMP-9 activity that might facilitate gemcitabine penetrability. CONCLUSION: These data demonstrate that AV25CDC is an effective oncolytic agent candidate for pancreatic cancer chemovirotherapy combination.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Fosfatases cdc25/genética , Adenoviridae , Animais , Antimetabólitos Antineoplásicos/farmacologia , Cricetinae , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Humanos , Mesocricetus , Camundongos , Camundongos Nus , Regiões Promotoras Genéticas , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
10.
Cell Stress Chaperones ; 18(2): 243-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22907762

RESUMO

Hsp27 (HSPB1) is usually overexpressed in breast cancers affecting the disease outcome and the sensitivity of tumors to chemotherapy and radiotherapy. Hsp27 interacts with other proteins such as ß-catenin, histone deacetylase HDAC6, transcription factor STAT2 and procaspase-3. Phosphatase and tensin homologue (PTEN) is a tumor suppressor gene that is deleted in many human tumors. The PI3K/Akt signaling pathway is negatively regulated by PTEN. Hsp27 is described as a key component of the Akt signaling cascade: Akt, BAD, Forkhead transcription factors, Hsp27, mitogen-activated protein kinase kinase-3 and -6. Here, we have examined whether the downregulation of Hsp27 by siHsp27 affects the PTEN levels in the MCF-7 human breast cancer cell line. PTEN was detected with two different antibodies using western blots and immunocytochemistry. p-Akt was also evaluated by western blot. In addition, Hsp27 and PTEN were immunoprecipitated to know whether these proteins interact. Intracellular colocalization studies were carried out by confocal microscopy. A significant reduction in the Hsp27 levels was noted in the siHsp27 transfected cells. These Hsp27 downregulated cells showed a significant increased expression of PTEN. The MW 76 and 55 kDa PTEN forms were upregulated as revealed by two different antibodies. The phosphatase activity of PTEN seems to be active because p-Akt levels were reduced. Hsp27 immunoprecipitation was bringing PTEN and vice versa, these two proteins seem to interact at cytoplasmic level by FRET. Downregulation of Hsp27 stabilized PTEN protein levels. Chaperone-assisted E3 ligase C terminus of Hsc70-interacting protein (CHIP) levels were not significantly influenced by Hsp27 downregulation. In conclusion, we report a novel function of Hsp27 modulating the PTEN levels in human breast cancer cells suggesting an interaction between these two molecules.


Assuntos
Proteínas de Choque Térmico HSP27/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Regulação para Baixo , Feminino , Transferência Ressonante de Energia de Fluorescência , Proteínas de Choque Térmico HSP27/antagonistas & inibidores , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Imuno-Histoquímica , Imunoprecipitação , Células MCF-7 , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Regulação para Cima , beta Catenina/metabolismo
11.
Cancer Metastasis Rev ; 27(4): 691-705, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18542844

RESUMO

Tumor growth is essentially the result of an evolving cross-talk between malignant and surrounding stromal cells (fibroblasts, endothelial cells and inflammatory cells). This heterogeneous mass of extracellular matrix and intermingled cells interact through cell-cell and cell-matrix contacts. Malignant cells also secrete soluble proteins that reach neighbor stromal cells, forcing them to provide the soil on which they will grow and metastasize. Different studies including expression array analysis identified the matricellular protein SPARC as a marker of poor prognosis in different cancer types. Further evidence demonstrated that high SPARC levels are often associated with the most aggressive and highly metastatic tumors. Here we describe the most recent evidence that links SPARC with human cancer progression, the controversy regarding its role in certain human cancers and the physiological processes in which SPARC is involved: epithelial-mesenchymal transition, immune surveillance and angiogenesis. Its relevance as a potential target in cancer therapy is also discussed.


Assuntos
Neoplasias/metabolismo , Osteonectina/metabolismo , Transdução de Sinais/fisiologia , Animais , Progressão da Doença , Humanos
12.
Cancer Metastasis Rev ; 27(3): 523-37, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18459035

RESUMO

Tumor growth is essentially the result of an evolving cross-talk between malignant and surrounding stromal cells (fibroblasts, endothelial cells and inflammatory cells). This heterogeneous mass of extracellular matrix and intermingled cells interact through cell-cell and cell-matrix contacts. Malignant cells also secrete soluble proteins that reach neighbor stromal cells, forcing them to provide the soil on which they will grow and metastasize. Different studies including expression array analysis identified the matricellular protein SPARC as a marker of poor prognosis in different cancer types. Further evidence demonstrated that high SPARC levels are often associated with the most aggressive and highly metastatic tumors. Here we describe the most recent evidence that links SPARC with human cancer progression, the controversy regarding its role in certain human cancers and the physiological processes in which SPARC is involved: epithelial-mesenchymal transition, immune surveillance and angiogenesis. Its relevance as a potential target in cancer therapy is also discussed.


Assuntos
Comunicação Celular/fisiologia , Neoplasias/metabolismo , Neoplasias/patologia , Osteonectina/fisiologia , Animais , Progressão da Doença , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Humanos , Invasividade Neoplásica/patologia , Receptor Cross-Talk/fisiologia
13.
Proteomics ; 7(22): 4123-34, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17994631

RESUMO

Secreted protein, acidic and rich in cysteines (SPARC) is a secreted protein associated with increased aggressiveness of different human cancer types. In order to identify downstream mediators of SPARC activity, we performed a 2-DE proteomic analysis of human melanoma cells following antisense-mediated downregulation of SPARC expression. We found 23/504 differential spots, 15 of which were identified by peptide fingerprinting analysis. Three of the differential proteins (N-cadherin (N-CAD), clusterin (CLU), and HSP27) were validated by immunoblotting, confirming decreased levels of N-CAD and CLU and increased amounts of HSP27 in conditioned media of cells with diminished SPARC expression. Furthermore, transient knock down of SPARC expression in melanoma cells following adenoviral-mediated transfer of antisense RNA confirmed these changes. We next developed two different RNAs against SPARC that were able to inhibit in vivo melanoma cell growth. Immunoblotting of the secreted fraction of RNAi-transfected melanoma cells confirmed that downregulation of SPARC expression promoted decreased levels of N-CAD and CLU and increased secretion of HSP27. Transient re-expression of SPARC in SPARC-downregulated cells reverted extracellular N-CAD, CLU, and HSP27 to levels similar to those in the control. These results constitute the first evidence that SPARC, N-CAD, CLU, and HSP27 converge in a unique molecular network in melanoma cells.


Assuntos
Caderinas/metabolismo , Clusterina/metabolismo , Proteínas de Choque Térmico/metabolismo , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Osteonectina/metabolismo , Caderinas/análise , Linhagem Celular Tumoral , Clusterina/análise , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/análise , Humanos , Chaperonas Moleculares , Proteínas de Neoplasias/análise , Osteonectina/biossíntese , Proteômica/métodos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA