Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 12(5)2023 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-36899894

RESUMO

Fragile X syndrome (FXS) is the most common form of monogenic intellectual disability and autism, caused by the absence of the functional fragile X messenger ribonucleoprotein 1 (FMRP). FXS features include increased and dysregulated protein synthesis, observed in both murine and human cells. Altered processing of the amyloid precursor protein (APP), consisting of an excess of soluble APPα (sAPPα), may contribute to this molecular phenotype in mice and human fibroblasts. Here we show an age-dependent dysregulation of APP processing in fibroblasts from FXS individuals, human neural precursor cells derived from induced pluripotent stem cells (iPSCs), and forebrain organoids. Moreover, FXS fibroblasts treated with a cell-permeable peptide that decreases the generation of sAPPα show restored levels of protein synthesis. Our findings suggest the possibility of using cell-based permeable peptides as a future therapeutic approach for FXS during a defined developmental window.


Assuntos
Síndrome do Cromossomo X Frágil , Células-Tronco Neurais , Humanos , Precursor de Proteína beta-Amiloide/metabolismo , Síndrome do Cromossomo X Frágil/genética , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo
2.
Cell Death Dis ; 13(8): 719, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35982038

RESUMO

Converging evidence indicates that the Fragile X Messenger Ribonucleoprotein (FMRP), which absent or mutated in Fragile X Syndrome (FXS), plays a role in many types of cancers. However, while FMRP roles in brain development and function have been extensively studied, its involvement in the biology of brain tumors remains largely unexplored. Here we show, in human glioblastoma (GBM) biopsies, that increased expression of FMRP directly correlates with a worse patient outcome. In contrast, reductions in FMRP correlate with a diminished tumor growth and proliferation of human GBM stem-like cells (GSCs) in vitro in a cell culture model and in vivo in mouse brain GSC xenografts. Consistently, increased FMRP levels promote GSC proliferation. To characterize the mechanism(s) by which FMRP regulates GSC proliferation, we performed GSC transcriptome analyses in GSCs expressing high levels of FMRP, and in these GSCs after knockdown of FMRP. We show that the WNT signalling is the most significantly enriched among the published FMRP target genes and genes involved in ASD. Consistently, we find that reductions in FMRP downregulate both the canonical WNT/ß-Catenin and the non-canonical WNT-ERK1/2 signalling pathways, reducing the stability of several key transcription factors (i.e. ß-Catenin, CREB and ETS1) previously implicated in the modulation of malignant features of glioma cells. Our findings support a key role for FMRP in GBM cancer progression, acting via regulation of WNT signalling.


Assuntos
Neoplasias Encefálicas , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Glioblastoma , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioblastoma/patologia , Humanos , Camundongos , Células-Tronco Neoplásicas/metabolismo , Ribonucleoproteínas , Via de Sinalização Wnt/genética , beta Catenina/metabolismo
3.
Cell Death Dis ; 8(11): e3169, 2017 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-29144507

RESUMO

The fragile X mental retardation protein (FMRP) is lacking or mutated in patients with the fragile X syndrome (FXS), the most frequent form of inherited intellectual disability. FMRP affects metastasis formation in a mouse model for breast cancer. Here we show that FMRP is overexpressed in human melanoma with high Breslow thickness and high Clark level. Furthermore, meta-analysis of the TCGA melanoma data revealed that high levels of FMRP expression correlate significantly with metastatic tumor tissues, risk of relapsing and disease-free survival. Reduction of FMRP in metastatic melanoma cell lines impinges on cell migration, invasion and adhesion. Next-generation sequencing in human melanoma cells revealed that FMRP regulates a large number of mRNAs involved in relevant processes of melanoma progression. Our findings suggest an association between FMRP levels and the invasive phenotype in melanoma and might open new avenues towards the discovery of novel therapeutic targets.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Proteína do X Frágil da Deficiência Intelectual/genética , Humanos , Invasividade Neoplásica , Transfecção
4.
EMBO Rep ; 17(12): 1872-1889, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27852622

RESUMO

MYC deregulation is common in human cancer and has a role in sustaining the aggressive cancer stem cell populations. MYC mediates a broad transcriptional response controlling normal biological programmes, but its activity is not clearly understood. We address MYC function in cancer stem cells through the inducible expression of Omomyc-a MYC-derived polypeptide interfering with MYC activity-taking as model the most lethal brain tumour, glioblastoma. Omomyc bridles the key cancer stemlike cell features and affects the tumour microenvironment, inhibiting angiogenesis. This occurs because Omomyc interferes with proper MYC localization and itself associates with the genome, with a preference for sites occupied by MYC This is accompanied by selective repression of master transcription factors for glioblastoma stemlike cell identity such as OLIG2, POU3F2, SOX2, upregulation of effectors of tumour suppression and differentiation such as ID4, MIAT, PTEN, and modulation of the expression of microRNAs that target molecules implicated in glioblastoma growth and invasion such as EGFR and ZEB1. Data support a novel view of MYC as a network stabilizer that strengthens the regulatory nodes of gene expression networks controlling cell phenotype and highlight Omomyc as model molecule for targeting cancer stem cells.


Assuntos
Regulação Neoplásica da Expressão Gênica , Genes myc , Glioblastoma/genética , Células-Tronco Neoplásicas/fisiologia , Fragmentos de Peptídeos/genética , Proteínas Proto-Oncogênicas c-myc/genética , Fatores de Transcrição/genética , Inibidores da Angiogênese , Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular , Proliferação de Células , Receptores ErbB/genética , Glioblastoma/fisiopatologia , Humanos , Proteínas Inibidoras de Diferenciação/genética , MicroRNAs/genética , Proteínas do Tecido Nervoso/genética , Fator de Transcrição 2 de Oligodendrócitos , Ligação Proteica , Ativação Transcricional , Microambiente Tumoral/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética
5.
Int J Mol Sci ; 16(8): 18312-27, 2015 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-26258776

RESUMO

Spinal muscular atrophy (SMA) is an inherited neuromuscular disorder and the leading genetic cause of death in infants. Despite the disease-causing gene, survival motor neuron (SMN1), encodes a ubiquitous protein, SMN1 deficiency preferentially affects spinal motor neurons (MNs), leaving the basis of this selective cell damage still unexplained. As neural stem cells (NSCs) are multipotent self-renewing cells that can differentiate into neurons, they represent an in vitro model for elucidating the pathogenetic mechanism of neurodegenerative diseases such as SMA. Here we characterize for the first time neural stem cells (NSCs) derived from embryonic spinal cords of a severe SMNΔ7 SMA mouse model. SMNΔ7 NSCs behave as their wild type (WT) counterparts, when we consider neurosphere formation ability and the expression levels of specific regional and self-renewal markers. However, they show a perturbed cell cycle phase distribution and an increased proliferation rate compared to wild type cells. Moreover, SMNΔ7 NSCs are characterized by the differential expression of a limited number of miRNAs, among which miR-335-5p and miR-100-5p, reduced in SMNΔ7 NSCs compared to WT cells. We suggest that such miRNAs may be related to the proliferation differences characterizing SMNΔ7 NSCs, and may be potentially involved in the molecular mechanisms of SMA.


Assuntos
MicroRNAs/genética , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patologia , Células-Tronco Neurais/patologia , Medula Espinal/patologia , Transcriptoma , Animais , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Camundongos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Mutação , Células-Tronco Neurais/metabolismo , Medula Espinal/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/genética
6.
Oncotarget ; 6(26): 22526-52, 2015 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-26188123

RESUMO

Glioblastoma multiforme (GBM) is the most common and deadliest primary brain tumor, driving patients to death within 15 months after diagnosis (short term survivors, ST), with the exception of a small fraction of patients (long term survivors, LT) surviving longer than 36 months. Here we present deep sequencing data showing that peritumoral (P) areas differ from healthy white matter, but share with their respective frankly tumoral (C) samples, a number of mRNAs and microRNAs representative of extracellular matrix remodeling, TGFß and signaling, of the involvement of cell types different from tumor cells but contributing to tumor growth, such as microglia or reactive astrocytes. Moreover, we provide evidence about RNAs differentially expressed in ST vs LT samples, suggesting the contribution of TGF-ß signaling in this distinction too. We also show that the edited form of miR-376c-3p is reduced in C vs P samples and in ST tumors compared to LT ones. As a whole, our study provides new insights into the still puzzling distinction between ST and LT tumors, and sheds new light onto that "grey" zone represented by the area surrounding the tumor, which we show to be characterized by the expression of several molecules shared with the proper tumor mass.


Assuntos
Neoplasias Encefálicas/genética , Glioblastoma/genética , Adulto , Idoso , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Transdução de Sinais , Transcriptoma
7.
RNA Biol ; 11(4): 334-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24717285

RESUMO

Several cellular microRNAs show substantial changes in expression during HIV-1 infection and their active role in the viral life cycle is progressively emerging. In the present study, we found that HIV-1 infection of Jurkat T cells significantly induces the expression of miR-222. We show that this induction depends on HIV-1 Tat protein, which is able to increase the transcriptional activity of NFkB on miR-222 promoter. Moreover, we demonstrate that miR-222 directly targets CD4, a key receptor for HIV-1, thus reducing its expression. We propose that Tat, by inducing miR-222 expression, complements the CD4 downregulation activity exerted by other viral proteins (i.e., Nef, Vpu, and Env), and we suggest that this represents a novel mechanism through which HIV-1 efficiently represses CD4 expression in infected cells.


Assuntos
Antígenos CD4/genética , Regulação da Expressão Gênica , Infecções por HIV/genética , HIV-1/fisiologia , MicroRNAs/genética , Subpopulações de Linfócitos T/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Antígenos CD4/metabolismo , Linhagem Celular , Infecções por HIV/imunologia , Infecções por HIV/metabolismo , Humanos , NF-kappa B/metabolismo , RNA Mensageiro/genética , Subpopulações de Linfócitos T/imunologia
8.
EMBO Mol Med ; 5(10): 1523-36, 2013 10.
Artigo em Inglês | MEDLINE | ID: mdl-24092663

RESUMO

The role of the fragile X mental retardation protein (FMRP) is well established in brain, where its absence leads to the fragile X syndrome (FXS). FMRP is almost ubiquitously expressed, suggesting that, in addition to its effects in brain, it may have fundamental roles in other organs. There is evidence that FMRP expression can be linked to cancer. FMR1 mRNA, encoding FMRP, is overexpressed in hepatocellular carcinoma cells. A decreased risk of cancer has been reported in patients with FXS while a patient-case with FXS showed an unusual decrease of tumour brain invasiveness. However, a role for FMRP in regulating cancer biology, if any, remains unknown. We show here that FMRP and FMR1 mRNA levels correlate with prognostic indicators of aggressive breast cancer, lung metastases probability and triple negative breast cancer (TNBC). We establish that FMRP overexpression in murine breast primary tumours enhances lung metastasis while its reduction has the opposite effect regulating cell spreading and invasion. FMRP binds mRNAs involved in epithelial mesenchymal transition (EMT) and invasion including E-cadherin and Vimentin mRNAs, hallmarks of EMT and cancer progression.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/metabolismo , RNA Mensageiro/metabolismo , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caderinas/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Forma Celular , Progressão da Doença , Transição Epitelial-Mesenquimal , Feminino , Proteína do X Frágil da Deficiência Intelectual/antagonistas & inibidores , Proteína do X Frágil da Deficiência Intelectual/genética , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Camundongos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Vimentina/metabolismo
9.
J Gen Virol ; 92(Pt 5): 1228-1232, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21289159

RESUMO

The adenosine deaminases acting on RNA (ADAR) enzymes catalyse conversion of adenosine to inosine in dsRNA. A positive effect of ADAR1 on human immunodeficiency virus type 1 (HIV-1) replication has recently been reported. Here, we show that another ADAR enzyme, ADAR2, positively affects the replication process of HIV-1. We found that, analogously to ADAR1, ADAR2 enhances the release of progeny virions by an editing-dependent mechanism. However, differently from the ADAR1 enzyme, ADAR2 does not increase the infectious potential of the virus. Importantly, downregulation of ADAR2 in Jurkat cells significantly impairs viral replication. Therefore, ADAR2 shares some but not all proviral functions of ADAR1. These results suggest a novel role of ADAR2 as a viral regulator.


Assuntos
Adenosina Desaminase/metabolismo , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno , Provírus/patogenicidade , Replicação Viral , Humanos , Células Jurkat , Proteínas de Ligação a RNA
10.
Free Radic Biol Med ; 49(3): 374-82, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20441791

RESUMO

Recent studies suggest that the effects of VEGF-A, the prototype VEGF ligand, may extend to a variety of cell types other than endothelial cells. The expression of VEGF-A and its main receptors, Flt-1/VEGFR-1 and KDR/Flk-1/VEGFR-2, was indeed detected in several cell types, including cardiac myocytes and regenerating myotubes. In addition to its proangiogenic activity, evidence indicates that VEGF-A can sustain skeletal muscle regeneration by enhancing the survival and migration of myogenic cells and by promoting the growth of myogenic fibers. In this study, our aim was to investigate whether VEGF could protect skeletal muscle satellite cells from apoptotic cell death triggered by reactive oxygen species and to identify the main molecular mechanisms. C2C12 mouse myoblasts, cultured in vitro in the presence of exogenous VEGF or stably transfected with a plasmid vector expressing VEGF-A, were subjected to oxidative stress and analyzed for cell growth and survival, induction of apoptosis, and molecular signaling. The results of our study demonstrated that VEGF protects C2C12 myoblasts from apoptosis induced by oxidative or hypoxic-like stress. This protection did not correlate with the modulation of the expression of VEGF receptors, but is clearly linked to the phosphorylation of the KDR/Flk-1 receptor, the activation of NF-kappaB, and/or the overexpression of the antiapoptotic protein alphaB-crystallin.


Assuntos
Mioblastos Esqueléticos/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/fisiologia , Cadeia B de alfa-Cristalina/fisiologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Peróxido de Hidrogênio/farmacologia , Camundongos , Mioblastos Esqueléticos/efeitos dos fármacos , NF-kappa B/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Interferência de RNA , Regeneração , Transdução de Sinais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Cadeia B de alfa-Cristalina/biossíntese , Proteína bcl-X/fisiologia
11.
Mol Cell Neurosci ; 42(4): 341-9, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19698788

RESUMO

In the adult mammalian brain, multipotential neural stem cells (NSC) persist throughout life in areas where neurogenesis is maintained. A distinctive trait of NSCs growing in vitro as neurospheres (NS), is their ability to self-renew, differentiate and migrate to sites of injury, such as gliomas. We have studied the role of Reelin, an extracellular matrix protein involved in brain development, in NSCs derived from normal newborn mice or from reeler, a natural mutant in which Reelin is not expressed. We show that the absence of Reelin negatively affects proliferation, NS-forming ability, and neuronal differentiation. Reeler NSCs are unable to migrate in chains, a migration mode typical of neural precursors homing to injury sites in adult CNS. All these effects are partially rescued by ectopic Reelin supplementation. Finally, we show that reeler NSCs fail to migrate in vivo towards gliomas. Overall, our results indicate that Reelin affects all major features of postnatal NSCs, and that it is required for the proper homing of NSCs to tumor sites in adult brain.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Proteínas da Matriz Extracelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Serina Endopeptidases/metabolismo , Células-Tronco/fisiologia , Animais , Encéfalo/citologia , Encéfalo/fisiologia , Moléculas de Adesão Celular Neuronais/genética , Células Cultivadas , Proteínas da Matriz Extracelular/genética , Feminino , Glioma/metabolismo , Glioma/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes Neurológicos , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Proteína Reelina , Serina Endopeptidases/genética , Células-Tronco/citologia
12.
FASEB J ; 23(12): 4276-87, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19713529

RESUMO

MicroRNAs are a class of sophisticated regulators of gene expression, acting as post-transcriptional inhibitors that recognize their target mRNAs through base pairing with short regions along the 3'UTRs. Several microRNAs are tissue specific, suggesting a specialized role in tissue differentiation or maintenance, and quite a few are critically involved in tumorigenesis. We studied miR-128, a brain-enriched microRNA, in retinoic acid-differentiated neuroblastoma cells, and we found that this microRNA is up-regulated in treated cells, where it down-modulates the expression of two proteins involved in the migratory potential of neural cells: Reelin and DCX. Consistently, miR-128 ectopic overexpression suppressed Reelin and DCX, whereas the LNA antisense-mediated miR-128 knockdown caused the two proteins to increase. Ectopic miR-128 overexpression reduced neuroblastoma cell motility and invasiveness, and impaired cell growth. Finally, the analysis of a small series of primary human neuroblastomas showed an association between high levels of miR-128 expression and favorable features, such as favorable Shimada category or very young age at diagnosis. Thus, we provide evidence for a role for miR-128 in the molecular events modulating neuroblastoma progression and aggressiveness.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica/fisiologia , MicroRNAs/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuroblastoma/tratamento farmacológico , Neuropeptídeos/metabolismo , Serina Endopeptidases/metabolismo , Sequência de Bases , Moléculas de Adesão Celular Neuronais/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Proteínas da Matriz Extracelular/genética , Humanos , MicroRNAs/genética , Proteínas Associadas aos Microtúbulos/genética , Dados de Sequência Molecular , Invasividade Neoplásica , Proteínas do Tecido Nervoso/genética , Neuroblastoma/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Neuropeptídeos/genética , Proteína Reelina , Serina Endopeptidases/genética , Tretinoína/farmacologia
13.
Nucleic Acids Res ; 37(17): 5848-58, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19651874

RESUMO

Adenosine deaminases that act on dsRNA (ADARs) are enzymes that target double-stranded regions of RNA converting adenosines into inosines (A-to-I editing) thus contributing to genome complexity and fine regulation of gene expression. It has been described that a member of the ADAR family, ADAR1, can target viruses and affect their replication process. Here we report evidence showing that ADAR1 stimulates human immuno deficiency virus type 1 (HIV-1) replication by using both editing-dependent and editing-independent mechanisms. We show that over-expression of ADAR1 in HIV-1 producer cells increases viral protein accumulation in an editing-independent manner. Moreover, HIV-1 virions generated in the presence of over-expressed ADAR1 but not an editing-inactive ADAR1 mutant are released more efficiently and display enhanced infectivity, as demonstrated by challenge assays performed with T cell lines and primary CD4(+) T lymphocytes. Finally, we report that ADAR1 associates with HIV-1 RNAs and edits adenosines in the 5' untranslated region (UTR) and the Rev and Tat coding sequence. Overall these results suggest that HIV-1 has evolved mechanisms to take advantage of specific RNA editing activity of the host cell and disclose a stimulatory function of ADAR1 in the spread of HIV-1.


Assuntos
Adenosina Desaminase/metabolismo , HIV-1/genética , Edição de RNA , RNA Viral/metabolismo , Linfócitos T CD4-Positivos/enzimologia , Linhagem Celular , HIV-1/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Humanos , Células Jurkat , Proteínas de Ligação a RNA , Vírion/metabolismo
14.
PLoS One ; 3(12): e4029, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19107213

RESUMO

BACKGROUND: MiR-221 and miR-222 are two highly homologous microRNAs whose upregulation has been recently described in several types of human tumors, for some of which their oncogenic role was explained by the discovery of their target p27, a key cell cycle regulator. We previously showed this regulatory relationship in prostate carcinoma cell lines in vitro, underlying the role of miR-221/222 as inducers of proliferation and tumorigenicity. METHODOLOGY/PRINCIPAL FINDINGS: Here we describe a number of in vivo approaches confirming our previous data. The ectopic overexpression of miR-221 is able, per se, to confer a high growth advantage to LNCaP-derived tumors in SCID mice. Consistently, the anti-miR-221/222 antagomir treatment of established subcutaneous tumors derived from the highly aggressive PC3 cell line, naturally expressing high levels of miR-221/222, reduces tumor growth by increasing intratumoral p27 amount; this effect is long lasting, as it is detectable as long as 25 days after the treatment. Furthermore, we provide evidence in favour of a clinical relevance of the role of miR-221/222 in prostate carcinoma, by showing their general upregulation in patient-derived primary cell lines, where we find a significant inverse correlation with p27 expression. CONCLUSIONS/SIGNIFICANCE: These findings suggest that modulating miR-221/222 levels may have a therapeutic potential in prostate carcinoma.


Assuntos
Carcinoma/patologia , Proliferação de Células/efeitos dos fármacos , MicroRNAs/antagonistas & inibidores , Oligonucleotídeos/farmacologia , Neoplasias da Próstata/patologia , Idoso , Animais , Sequência de Bases , Carcinoma/genética , Progressão da Doença , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Regulação para Baixo/fisiologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Terapia Genética , Humanos , Masculino , Camundongos , Camundongos SCID , MicroRNAs/genética , Pessoa de Meia-Idade , Antígeno Nuclear de Célula em Proliferação/genética , Neoplasias da Próstata/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
15.
EMBO J ; 26(15): 3699-708, 2007 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-17627278

RESUMO

MicroRNAs (miRNAs) are potent post-transcriptional regulators of protein coding genes. Patterns of misexpression of miRNAs in cancer suggest key functions of miRNAs in tumorigenesis. However, current bioinformatics tools do not entirely support the identification and characterization of the mode of action of such miRNAs. Here, we used a novel functional genetic approach and identified miR-221 and miR-222 (miR-221&222) as potent regulators of p27(Kip1), a cell cycle inhibitor and tumor suppressor. Using miRNA inhibitors, we demonstrate that certain cancer cell lines require high activity of miR-221&222 to maintain low p27(Kip1) levels and continuous proliferation. Interestingly, high levels of miR-221&222 appear in glioblastomas and correlate with low levels of p27(Kip1) protein. Thus, deregulated expression of miR-221&222 promotes cancerous growth by inhibiting the expression of p27(Kip1).


Assuntos
Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , MicroRNAs/fisiologia , Neoplasias/patologia , Células 3T3 , Animais , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
J Biol Chem ; 282(32): 23716-24, 2007 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-17569667

RESUMO

MicroRNAs are short regulatory RNAs that negatively modulate protein expression at a post-transcriptional level and are deeply involved in the pathogenesis of several types of cancers. Here we show that miR-221 and miR-222, encoded in tandem on chromosome X, are overexpressed in the PC3 cellular model of aggressive prostate carcinoma, as compared with LNCaP and 22Rv1 cell line models of slowly growing carcinomas. In all cell lines tested, we show an inverse relationship between the expression of miR-221 and miR-222 and the cell cycle inhibitor p27(Kip1). We recognize two target sites for the microRNAs in the 3' untranslated region of p27 mRNA, and we show that miR-221/222 ectopic overexpression directly results in p27 down-regulation in LNCaP cells. In those cells, we demonstrate that the ectopic overexpression of miR-221/222 strongly affects their growth potential by inducing a G(1) to S shift in the cell cycle and is sufficient to induce a powerful enhancement of their colony-forming potential in soft agar. Consistently, miR-221 and miR-222 knock-down through antisense LNA oligonucleotides increases p27(Kip1) in PC3 cells and strongly reduces their clonogenicity in vitro. Our results suggest that miR-221/222 can be regarded as a new family of oncogenes, directly targeting the tumor suppressor p27(Kip1), and that their overexpression might be one of the factors contributing to the oncogenesis and progression of prostate carcinoma through p27(Kip1) down-regulation.


Assuntos
Carcinoma/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs , Oncogenes/genética , Sequência de Bases , Linhagem Celular Tumoral , Sobrevivência Celular , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Progressão da Doença , Humanos , Masculino , Dados de Sequência Molecular , Plasmídeos/metabolismo , Neoplasias da Próstata/metabolismo , Transfecção
17.
Free Radic Res ; 41(4): 391-401, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17454121

RESUMO

Since it was suggested that cobalt chloride (CoCl(2)) could mimic the O(2) sensing role of mitochondria by increasing reactive oxygen species (ROS) generation during normoxia, we studied the correlation between CoCl(2)-generation of free radicals and the induction of a hypoxic cellular response in myogenic cell lines. In both L6C5 and C2C12 cell lines, exposure to CoCl(2) induced an increase of intracellular oxidants, the accumulation of HIF-1alpha protein, and the expression of vascular endothelial growth factor (VEGF) and/or iNOS genes. On the other hand, only ascorbic acid, but not trolox, was effective in lowering the CoCl(2) gene up-regulation. Neither the cytotoxicity nor the apoptosis induced by CoCl(2) in skeletal muscle cells were modified by culture supplementation with either ascorbic acid or trolox. Thus, CoCl(2) treatment of myogenic cell lines may represent a useful and convenient in vitro model to study gene modulation induced by hypoxia in skeletal muscle, although cellular loss induced by this metal may involve mechanisms other than HIF-1alpha stabilization. It is unlikely, however, that ROS would represent the main mediators of CoCl(2) effects on muscle cells.


Assuntos
Apoptose , Cobalto/farmacologia , Radicais Livres , Hipóxia , Músculo Esquelético/patologia , Regulação para Cima , Animais , Gatos , Relação Dose-Resposta a Droga , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Oxigênio/metabolismo , Ratos , Espécies Reativas de Oxigênio , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Vaccine ; 24(21): 4586-91, 2006 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-16154671

RESUMO

The high toll of death among first-week infants is due to infections occurring at the end of pregnancy, during birth or by breastfeeding. This problem significantly concerns industrialized countries also. To prevent the typical "first-week infections", a vaccine would be protective as early as at the birth. In utero DNA immunization has demonstrated the effectiveness in inducing specific immunity in newborns. We have already published results of a 2-year follow-up showing long-term safety, protective antibody titers at birth and long-term immune memory, following intramuscular in utero anti-HBV DNA immunization in 90-days pig fetuses. We have now analyzed further parameters of short-term safety. Two different reporter genes were injected in the thigh muscles of 90-days fetuses. At 8 days following DNA injection, we found high-level of transgenes expression in all injected fetuses. A step gradient of expression from the area of injection was observed with both reporter genes. CMV promoter/enhancer produced higher levels of expression compared to SV40 promoter/enhancer. Moreover, no evidence of local or systemic flogistic alterations or fetal malformations, mortality or haemorrhage following intramuscular injection were observed. A single anti-HBV s-antigen DNA immunization in 90-days fetuses supported protective antibody levels in all immunized newborns, lasting at least up to 4 months after birth. Our report further sustains safety and efficacy of intramuscular in utero naked gene transfer and immunization. This approach may support therapeutic or prophylactic procedure in many early life-threatening pathologic conditions.


Assuntos
Técnicas de Transferência de Genes , Músculos/embriologia , Suínos/embriologia , Transgenes , Vacinas de DNA/administração & dosagem , Animais , Estudos de Viabilidade , Feminino , Plasmídeos , Gravidez , Vacinas de DNA/genética , Vacinas de DNA/imunologia
19.
Cancer Biol Ther ; 5(2): 174-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16340308

RESUMO

Angiogenesis is required for the development and biologic progression of glioblastoma multiform (GBM), which is the most malignant infiltrative astrocytoma. Vascular endothelial growth factor (VEGF) plays a predominant role in the increased vascularity and endothelial cell proliferation in GBMs driven by the expression of pro-angiogenic cytokines. In this study, we employed a vector-encoded VEGF siRNA to impair VEGF secretion from U87 human glioblastoma cells. The direct intra-tumor injection of a siRNA-encoding plasmid complexed with linear polyethylenimine (PEI) efficiently reduced the vascularization of treated tumors in xenografts established in SCID mice by subcutaneous inoculation of U87 cells, but was not able to reduce tumor growth. We then sought to strengthen the in vivo action of our siRNA by coupling it to a well known direct antiangiogenic agent, mouse interleukin 4 (mIL4). We infected U87 cells with a retroviral vector coexpressing the VEGF siRNA and mIL4 and produced stable cell lines that we used for an in vivo experiment of subcutaneous injection in SCID mice. In this setting, the concomitant expression of mIL4 and siRNA totally abolished the growth of subcutaneous tumors. These results suggest that our retroviral vector might be employed as a potential tool in future antiangiogenic gene therapy trials for glioblastoma.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética , Glioblastoma/terapia , Interleucina-4/uso terapêutico , Neovascularização Patológica/terapia , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/tratamento farmacológico , Terapia Combinada , Regulação para Baixo , Glioblastoma/irrigação sanguínea , Glioblastoma/tratamento farmacológico , Humanos , Camundongos , Camundongos SCID , Neovascularização Patológica/tratamento farmacológico , Plasmídeos/administração & dosagem , Plasmídeos/genética , RNA Interferente Pequeno/genética , Retroviridae/genética , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Cancer Gene Ther ; 12(12): 926-34, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15956982

RESUMO

RNA interference technology is emerging as a very potent tool to obtain a cellular knockdown of a desired gene. In this work we used vector-based RNA interference to inhibit vascular endothelial growth factor (VEGF) expression in prostate cancer in vitro and in vivo. We demonstrated that transduction with a plasmid carrying a small interfering RNA targeting all isoforms of VEGF, dramatically impairs the expression of this growth factor in the human prostate cancer cell line PC3. As a consequence, PC3 cells loose their ability to induce one of the fundamental steps of angiogenesis, namely the formation of a tube-like network in vitro. Most importantly, our "therapeutic" vector is able to impair tumor growth rate and vascularization in vivo. We show that a single injection of naked plasmid in developing neoplastic mass significantly decreases microvessel density in an androgen-refractory prostate xenograft and is able to sustain a long-term slowing down of tumor growth. In conclusion, our results confirm the basic role of VEGF in the angiogenic development of prostate carcinoma, and suggest that the use of our vector-based RNA interference approach to inhibit angiogenesis could be an effective tool in view of future gene therapy applications for prostate cancer.


Assuntos
Vetores Genéticos/genética , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/patologia , Interferência de RNA , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Sequência de Bases , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Masculino , Camundongos , Camundongos SCID , Dados de Sequência Molecular , Plasmídeos/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Ratos , Alinhamento de Sequência , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...