Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncotarget ; 9(7): 7567-7581, 2018 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-29484133

RESUMO

Our previous data supported a role for the Stearoyl-CoA desaturase (SCD5) in protection against malignancy, whereby it appears to functionally modify tumor stroma impairing tumor spread. SCD5 is significantly expressed in primary melanoma, but becomes barely detectable at tumor advanced stages. Looking for the regulatory mechanisms underlying SCD5 reduced expression during melanoma progression, we demonstrated a significantly lower stability of SCD5 protein as well as the direct targeting of SCD5 mRNA by the oncogenic miR-221&222 in metastatic cell lines. Moreover, our results indicated the existence of a negative feedback loop between SCD5 and miR-221&222, in good agreement with their opposite functions. Also, we showed how SCD5 re-expression and the direct supplementation of its main product oleic acid (OA) can drive advanced melanoma cell lines toward differentiation and reversion of the epithelial-mesenchymal (EMT)-like process, eventually inducing a less malignant phenotype. Indeed, SCD5 re-established the sensitivity to all-trans retinoic acid in A375M metastatic melanoma, associated with increased levels of Tyrosinase, melanin production and reduced proliferation. As evidenced by the correct modulation of some key transcription factors, SCD5 managed by favoring a partial mesenchymal-to-epithelial (MET) transition in in vitro studies. Interestingly, a more complete MET, including E-cadherin re-expression correctly localized at cell membranes, was obtained in in vivo xenograft models, thus indicating the requirement of direct contacts between tumor cells and the surrounding microenvironment as well as the presence of some essential factors for SCD5 complete function.

2.
Hum Mutat ; 38(7): 798-804, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28390077

RESUMO

RASopathies are a group of rare, clinically related conditions affecting development and growth, and are caused by germline mutations in genes encoding signal transducers and modulators with a role in the RAS signaling network. These disorders share facial dysmorphia, short stature, variable cognitive deficits, skeletal and cardiac defects, and a variable predisposition to malignancies. Here, we report on a de novo 10-nucleotide-long deletion in HRAS (c.481_490delGGGACCCTCT, NM_176795.4; p.Leu163ProfsTer52, NP_789765.1) affecting transcript processing as a novel event underlying a RASopathy characterized by developmental delay, intellectual disability and autistic features, distinctive coarse facies, reduced growth, and ectodermal anomalies. Molecular and biochemical studies demonstrated that the deletion promotes constitutive retention of exon IDX, which is generally skipped during HRAS transcript processing, and results in a stable and mildly hyperactive GDP/GTP-bound protein that is constitutively targeted to the plasma membrane. Our findings document a new mechanism leading to altered HRAS function that underlies a previously unappreciated phenotype within the RASopathy spectrum.


Assuntos
Deficiências do Desenvolvimento/genética , Regulação Neoplásica da Expressão Gênica , Genes ras , Proteínas Proto-Oncogênicas p21(ras)/genética , Animais , Transtorno Autístico/genética , Células COS , Membrana Celular/metabolismo , Criança , Pré-Escolar , Chlorocebus aethiops , Éxons , Fácies , Deleção de Genes , Mutação em Linhagem Germinativa , Humanos , Deficiência Intelectual/genética , Masculino , Fenótipo , RNA Mensageiro/metabolismo , Transdução de Sinais
3.
J Pathol ; 236(3): 315-25, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25802234

RESUMO

A proper balance between saturated and unsaturated fatty acids (FAs) is required for maintaining cell homeostasis. The increased demand of FAs to assemble the plasma membranes of continuously dividing cancer cells might unbalance this ratio and critically affect tumour outgrowth. We unveiled the role of the stearoyl-CoA desaturase SCD5 in converting saturated FAs into mono-unsaturated FAs during melanoma progression. SCD5 is down-regulated in advanced melanoma and its restored expression significantly reduced melanoma malignancy, both in vitro and in vivo, through a mechanism governing the secretion of extracellular matrix proteins, such as secreted protein acidic and rich in cysteine (SPARC) and collagen IV and of their proteases, such as cathepsin B. Enforced expression of SCD5 or supplementation of its enzymatic product, oleic acid, reduced the intracellular pH (pHe > pHi) and, in turn, vesicular trafficking across plasma membranes as well as melanoma dissemination. This intracellular acidification appears also to depend on SCD5-induced reduction of the C2 subunit of the vacuolar H(+) -ATPase, a proton pump whose inhibition changes the secretion profile of cancer cells. Our data support a role for SCD5 and its enzymatic product, oleic acid, in protection against malignancy, offering an explanation for the beneficial Mediterranean diet. Furthermore, SCD5 appears to functionally connect tumour cells and the surrounding stroma toward modification of the tumour microenvironment, with consequences on tumour spread and resistance to treatment.


Assuntos
Catepsina B/metabolismo , Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Ácido Oleico/metabolismo , Osteonectina/metabolismo , Estearoil-CoA Dessaturase/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Regulação para Baixo , Ácidos Graxos/análise , Ácidos Graxos/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Espaço Intracelular/metabolismo , Melanócitos/metabolismo , Melanócitos/patologia , Melanoma/patologia , Ácido Oleico/análise
4.
Hum Mol Genet ; 23(16): 4315-27, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24705357

RESUMO

RASopathies, a family of disorders characterized by cardiac defects, defective growth, facial dysmorphism, variable cognitive deficits and predisposition to certain malignancies, are caused by constitutional dysregulation of RAS signalling predominantly through the RAF/MEK/ERK (MAPK) cascade. We report on two germline mutations (p.Gly39dup and p.Val55Met) in RRAS, a gene encoding a small monomeric GTPase controlling cell adhesion, spreading and migration, underlying a rare (2 subjects among 504 individuals analysed) and variable phenotype with features partially overlapping Noonan syndrome, the most common RASopathy. We also identified somatic RRAS mutations (p.Gly39dup and p.Gln87Leu) in 2 of 110 cases of non-syndromic juvenile myelomonocytic leukaemia, a childhood myeloproliferative/myelodysplastic disease caused by upregulated RAS signalling, defining an atypical form of this haematological disorder rapidly progressing to acute myeloid leukaemia. Two of the three identified mutations affected known oncogenic hotspots of RAS genes and conferred variably enhanced RRAS function and stimulus-dependent MAPK activation. Expression of an RRAS mutant homolog in Caenorhabditis elegans enhanced RAS signalling and engendered protruding vulva, a phenotype previously linked to the RASopathy-causing SHOC2(S2G) mutant. Overall, these findings provide evidence of a functional link between RRAS and MAPK signalling and reveal an unpredicted role of enhanced RRAS function in human disease.


Assuntos
Carcinogênese/genética , Mutação/fisiologia , Fenótipo , Proteínas ras/genética , Animais , Caenorhabditis elegans , Estudos de Coortes , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mielomonocítica Juvenil/genética , MAP Quinase Quinase Quinases/metabolismo , Síndrome de Noonan/genética , Proteína Oncogênica v-akt/metabolismo , Transdução de Sinais/genética , Proteínas ras/química , Proteínas ras/metabolismo
5.
Cancer Cell Int ; 13(1): 101, 2013 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-24148231

RESUMO

BACKGROUND: Homeobox (HOX) genes deregulation has been largely implicated in the development of human leukemia. Among the HOXB cluster, HOXB1 was silent in a number of analyzed acute myeloid leukemia (AML) primary cells and cell lines, whereas it was expressed in normal terminally differentiated peripheral blood cells. METHODS: We evaluated the biological effects and the transcriptome changes determined by the retroviral transduction of HOXB1 in the human promyelocytic cell line HL60. RESULTS: Our results suggest that the enforced expression of HOXB1 reduces cell growth proliferation, inducing apoptosis and cell differentiation along the monocytic and granulocytic lineages. Accordingly, gene expression analysis showed the HOXB1-dependent down-regulation of some tumor promoting genes, paralleled by the up-regulation of apoptosis- and differentiation-related genes, thus supporting a tumor suppressor role for HOXB1 in AML. Finally, we indicated HOXB1 promoter hypermethylation as a mechanism responsible for HOXB1 silencing. CONCLUSIONS: We propose HOXB1 as an additional member of the HOX family with tumour suppressor properties suggesting a HOXB1/ATRA combination as a possible future therapeutic strategy in AML.

6.
Int J Cancer ; 133(4): 879-92, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23400877

RESUMO

Cutaneous melanoma is the fastest increasing cancer worldwide. Although several molecular abnormalities have been associated with melanoma progression, the underlying mechanisms are still largely unknown and few targeted therapies are under evaluation. Here we show that the HOXB7/PBX2 dimer acts as a positive transcriptional regulator of the oncogenic microRNA-221 and -222. In addition, demonstrating c-FOS as a direct target of miR-221&222, we identify a HOXB7/PBX2→miR-221&222 →c-FOS regulatory link, whereby the abrogation of functional HOXB7/PBX2 dimers leads to reduced miR-221&222 transcription and elevated c-FOS expression with consequent cell death. Taking advantage of the treatment with the peptide HXR9, an antagonist of HOX/PBX dimerization, we recognize miR-221&222 as effectors of its action, in turn confirming the HXR9 efficacy in the treatment of human melanoma malignancy, whilst sparing normal human melanocytes. Our findings, besides suggesting the potential therapeutic of HXR9 or its derivatives in malignant melanoma, suggest the disruption of the HOXB7/PBX2 complexes, miR-221&222 inhibition or even better their combination, as innovative therapeutic approaches.


Assuntos
Apoptose/fisiologia , Proteínas de Homeodomínio/genética , Melanoma/patologia , MicroRNAs/fisiologia , Proteínas Proto-Oncogênicas c-fos/fisiologia , Proteínas Proto-Oncogênicas/genética , Neoplasias Cutâneas/patologia , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Dimerização , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/fisiologia , Humanos , MicroRNAs/genética , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/fisiologia , RNA Interferente Pequeno , Transcrição Gênica
7.
PLoS One ; 8(2): e56824, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23437250

RESUMO

The abnormal expression of several microRNAs has a causal role in tumorigenesis with either antineoplastic or oncogenic functions. Here we demonstrated that miR-126 and miR-126* play a tumor suppressor role in human melanoma through the direct or indirect repression of several key oncogenic molecules. The expression levels of miR-126&126* were elevated in normal melanocytes and primary melanoma cell lines, whereas they markedly declined in metastatic cells. Indeed, the restored expression of miR-126&126* in two advanced melanoma cell lines was accompanied by a significant reduction of proliferation, invasion and chemotaxis in vitro as well as of growth and dissemination in vivo. In accordance, the reverse functional effects were obtained by knocking down miR-126&126* by transfecting antisense LNA oligonucleotides in melanoma cells. Looking for the effectors of these antineoplastic functions, we identified ADAM9 and MMP7, two metalloproteases playing a pivotal role in melanoma progression, as direct targets of miR-126&126*. In addition, as ADAM9 and MMP7 share a role in the proteolytic cleavage of the HB-EGF precursor, we looked for the effectiveness of this regulatory pathway in melanoma, confirming the decrease of HB-EGF activation as a consequence of miR-126&126*-dependent downmodulation of ADAM9 and MMP7. Finally, gene profile analyses showed that miR-126&126* reexpression was sufficient to inactivate other key signaling pathways involved in the oncogenic transformation, as PI3K/AKT and MAPK, and to restore melanogenesis, as indicated by KIT/MITF/TYR induction. In view of this miR-126&126* wide-ranging action, we believe that the replacement of these microRNAs might be considered a promising therapeutic approach.


Assuntos
Proteínas ADAM/genética , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Metaloproteinase 7 da Matriz/genética , Melanoma/genética , Proteínas de Membrana/genética , MicroRNAs/genética , Neoplasias Cutâneas/genética , Proteínas ADAM/metabolismo , Animais , Pareamento de Bases , Sequência de Bases , Linhagem Celular Tumoral , Progressão da Doença , Perfilação da Expressão Gênica , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Melanócitos/metabolismo , Melanoma/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Osteopontina/genética , Osteopontina/metabolismo , Proteólise , Interferência de RNA , Neoplasias Cutâneas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Pigment Cell Melanoma Res ; 24(5): 953-65, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21711453

RESUMO

MicroRNAs-221 and -222 are highly upregulated in several solid tumors, including melanomas. We demonstrate that the proto-oncogene ETS-1, involved in the pathogenesis of cancers of different origin, is a transcriptional regulator of miR-222 by direct binding to its promoter region. Differently from 293FT cells or early stage melanomas, where unphosphorylated ETS-1 represses miR-222 transcription, in metastatic melanoma the constitutively Thr-38 phosphorylated fraction of ETS-1 induces miR-222. Despite its stepwise decreased expression along with melanoma progression, the oncogenic activity of ETS-1 relies on its RAS/RAF/ERK-dependent phosphorylation status more than on its total amount. To close the loop, we demonstrate ETS-1 as a direct target of miR-222, but not miR-221, showing the novel option of their uncoupled functions. In addition, a spatial redistribution of ETS-1 protein from the nucleus to the cytoplasm is also evidenced in advanced melanoma cells. Finally, in vivo studies confirmed the contribution of miR-222 to the increased invasive potential obtained by ETS- silencing.


Assuntos
Regulação Neoplásica da Expressão Gênica , Melanoma/genética , Melanoma/patologia , MicroRNAs/genética , Proteína Proto-Oncogênica c-ets-1/genética , Transdução de Sinais/genética , Neoplasias Cutâneas/genética , Animais , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Melanócitos/metabolismo , Melanócitos/patologia , Camundongos , Camundongos Nus , MicroRNAs/metabolismo , Metástase Neoplásica , Fosforilação/efeitos dos fármacos , Fosfotreonina/metabolismo , Proto-Oncogene Mas , Proteína Proto-Oncogênica c-ets-1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias Cutâneas/patologia , Acetato de Tetradecanoilforbol/farmacologia , Transcrição Gênica/efeitos dos fármacos
9.
Int J Cancer ; 125(7): 1514-22, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19521982

RESUMO

Caveolin-1 (Cav-1), a member of the caveolin family, regulates caveolae-associated signaling proteins, which are involved in many biological processes, including cancer development. Cav-1 was found to exert a complex and ambiguous role as oncogene or tumor suppressor depending on the cellular microenvironment. Here we investigated Cav-1 expression and function in a panel of melanomas, finding its expression in all the cell lines. The exception was the primary vertical melanoma cell line, WM983A, characterized by the lack of Cav-1, and then utilized as a recipient for Cav-1 gene transduction to address a series of functional studies. The alleged yet controversial role of phospho (Ph)-Cav-1 on cell regulation was also tested by transducing the nonphosphorylatable Cav-1Y14A mutant. Wild-type Cav-1, but not mutated Cav-1Y14A, increased tumorigenicity as indicated by enhanced proliferation, migration, invasion and capacity of forming foci in semisolid medium. Accordingly, Cav-1 silencing inhibited melanoma cell growth reducing some of the typical traits of malignancy. Finally, we detected a secreted fraction of Cav-1 associated with cell released microvesicular particles able to stimulate in vitro anchorage independence, migration and invasion in a paracrine/autocrine fashion and, more important, competent to convey metastatic asset from the donor melanoma to the less aggressive recipient cell line. A direct correlation between Cav-1 levels, the amount of microvesicles released in the culture medium and MMP-9 expression was also observed.


Assuntos
Biomarcadores Tumorais/metabolismo , Caveolina 1/metabolismo , Melanoma/metabolismo , Neoplasias Cutâneas/metabolismo , Western Blotting , Caveolina 1/genética , Linhagem Celular Tumoral , Movimento Celular , Progressão da Doença , Imunofluorescência , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Melanoma/patologia , Invasividade Neoplásica , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Cutâneas/patologia
10.
Cancer Res ; 68(8): 2745-54, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18417445

RESUMO

The incidence of cutaneous melanoma is steadily increasing. Although several molecular abnormalities have been associated with melanoma progression, the mechanisms underlying the differential gene expression are still largely unknown and targeted therapies are not yet available. Noncoding small RNAs, termed microRNAs (miR), have been recently reported to play important roles in major cellular processes, including those involved in cancer development and progression. We have identified the promyelocytic leukemia zinc finger (PLZF) transcription factor as a repressor of miR-221 and miR-222 by direct binding to their putative regulatory region. Specifically, PLZF silencing in melanomas unblocks miR-221 and miR-222, which in turn controls the progression of the neoplasia through down-modulation of p27Kip1/CDKN1B and c-KIT receptor, leading to enhanced proliferation and differentiation blockade of the melanoma cells, respectively. In vitro and in vivo functional studies, including the use of antisense "antagomir" oligonucleotides, confirmed the key role of miR-221/-222 in regulating the progression of human melanoma; this suggests that targeted therapies suppressing miR-221/-222 may prove beneficial in advanced melanoma.


Assuntos
Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição Kruppel-Like/metabolismo , Melanoma Experimental/genética , MicroRNAs/genética , Neoplasias Cutâneas/genética , Dedos de Zinco , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Inibidor de Quinase Dependente de Ciclina p27 , Progressão da Doença , Regulação para Baixo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Nus , Oligonucleotídeos Antissenso/farmacologia , Proteína com Dedos de Zinco da Leucemia Promielocítica , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo
11.
Oncogene ; 23(26): 4567-76, 2004 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-15077196

RESUMO

The promyelocytic leukemia zinc finger (PLZF) protein has been described as a transcriptional repressor of homeobox (HOX)-containing genes during embryogenesis. As we previously demonstrated a functional link between overexpression of HOXB7 and melanoma progression, we investigated the lack of PLZF as the possible cause of HOXB7 constitutive activation in these neoplastic cells. Accordingly, we found PLZF expression in melanocytes, but not in melanoma cells, a pattern inversely related to that of HOXB7. PLZF retroviral gene transduction was then performed in a panel of melanoma cell lines, and tumorigenicity was compared with that of empty vector-transduced control cell lines. Evaluation of in vitro migration, invasion and adhesion indicated that PLZF gene transduction induced a less malignant phenotype, as confirmed through in vivo studies performed in athymic nude mice. This reduced tumorigenicity was not coupled with HOXB7 repression. In order to find more about the molecular targets of PLZF, the gene expression profiles of PLZF- and empty vector-transduced A375 melanoma cells were analysed by Atlas Cancer macroarray. Among several genes modulated by PLZF enforced expression, of particular interest were integrin alphavbeta3, osteonectin/SPARC and matrix metalloprotease-9 that were downmodulated, and the tyrosinase-related protein-1 that was upregulated in all the analysed samples. This profile confirms the reduced tumorigenic phenotype with reversion to a more differentiated, melanocyte like, pattern, thus suggesting a suppressor role for PLZF in solid tumors. Moreover, these results indicate that PLZF and HOXB7 are functionally independent and that their coupled deregulation may account for most of the alterations described in melanomas.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Melanoma/patologia , Glicoproteínas de Membrana , Oxirredutases , Fatores de Transcrição/fisiologia , Animais , Testes de Carcinogenicidade , Divisão Celular/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Integrina alfaVbeta3/genética , Fatores de Transcrição Kruppel-Like , Metaloproteinase 9 da Matriz/genética , Melanócitos/metabolismo , Melanoma/genética , Camundongos , Camundongos Nus , Invasividade Neoplásica , Osteonectina/genética , Regiões Promotoras Genéticas , Proteína com Dedos de Zinco da Leucemia Promielocítica , Proteínas/genética , Valores de Referência , Células Tumorais Cultivadas
12.
Biochim Biophys Acta ; 1626(1-3): 1-9, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12697323

RESUMO

Products of HOX genes are transcription factors responsible for developmental regulation and postnatal tissue homeostasis. Besides their well-established function played during embryonic development, we had previously demonstrated the direct role of HOXB7 in tumor progression through transactivation of several genes involved in the proliferative and angiogenic processes. This role is at first exerted through the deregulated, constitutive expression of this gene. To define the factors possibly responsible for such activation, we studied the molecular regulation of HOXB7 in embryonic and neoplastic cells. In a 1.9-kb 5' promoter region, we identified and functionally tested, at least in vitro, different regulatory sequences showing a direct binding by the NF-Y, YY1, Sp1/Sp3 and upstream stimulatory factor 1 (USF-1) transcription factors. Cell transfection and site-specific mutagenesis demonstrated Sp1/Sp3, NF-Y, YY1 and USF-1 binding to be functional and fundamental in driving HOXB7 expression. Disruption of the corresponding sites reduces gene expression of 65%, 78% and 55%, respectively. Because HOXB7 seems to play an important role in tumor proliferation and progression, the analysis of its regulatory sequences might represent an important step for gene targeting according to a new therapeutic strategy.


Assuntos
Proteínas de Homeodomínio/genética , Fatores de Transcrição/fisiologia , Ativação Transcricional , Sequência de Bases , Sítios de Ligação , Fator de Ligação a CCAAT/fisiologia , Linhagem Celular , Proteínas de Ligação a DNA/fisiologia , Fatores de Ligação de DNA Eritroide Específicos , Proteínas de Homeodomínio/biossíntese , Humanos , Mutação , Regiões Promotoras Genéticas , Fator de Transcrição Sp1/fisiologia , Sítio de Iniciação de Transcrição , Células Tumorais Cultivadas , Fatores Estimuladores Upstream , Fator de Transcrição YY1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...