Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 12(5): 422, 2021 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-33927188

RESUMO

Tumor-associated macrophages (TAMs) in the tumor microenvironment contribute to poor prognosis in gastric cancer (GC). However, the underlying mechanism by which TAMs promote GC progression and metastasis remains elusive. Expression of POU1F1 was detected in 60 matched GC-normal tissue pairs using qRT-PCR and immunohistochemistry (IHC) analysis. The correlation between POU1F1 and the clinical-pathological factors of GC patients were further assessed. Cell proliferation was monitored by CCK-8, colony formation, and 5-Ethynyl-2'-deoxyuridine (EdU) incorporation assays. Cell migration and invasion were assessed by transwell assays. The impact on angiogenesis was evaluated by tube formation assay. Xenograft model was generated to investigate the role of POU1F1 on tumor growth and lung metastasis in vivo. GST pull-down and Co-immunoprecipitation (Co-IP) were used to study the interaction between HMGA1B/2 and POU1F1. Chromatin immunoprecipitation (ChIP) and dual luciferase reporter assays were performed to investigate the transcriptional regulation of POU1F1. Flow cytometry was performed to detect the surface expression of macrophage markers. Upregulated POU1F1 observed both in GC tissues and cell lines was positively correlated with poor prognosis. Knockdown of POU1F1 inhibited cell proliferation, migration, invasion, and angiogenesis in vitro, and suppressed tumor growth in vivo. HMGA1B/2 transcriptionally activated-POU1F1. POU1F1 promoted GC progression via regulating macrophage proliferation, migration, polarization, and angiogenesis in a CXCL12/CXCR4-dependent manner. POU1F1 also promoted GC metastasis in lung by modulating macrophage polarization through CXCL12/CXCR4 axis in vivo. HMGA1B/2-upregulated POU1F1 promoted GC metastasis via regulating macrophage polarization in a CXCL12/CXCR4-dependent manner.


Assuntos
Quimiocina CXCL12/metabolismo , Proteína HMGA1b/metabolismo , Proteína HMGA2/metabolismo , Macrófagos/metabolismo , Receptores CXCR4/metabolismo , Neoplasias Gástricas/metabolismo , Fator de Transcrição Pit-1/metabolismo , Animais , Proliferação de Células/fisiologia , Proteína HMGA1b/genética , Proteína HMGA2/genética , Xenoenxertos , Humanos , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Nus , Metástase Neoplásica , Transdução de Sinais , Neoplasias Gástricas/genética , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/patologia , Análise de Sobrevida , Fator de Transcrição Pit-1/genética , Transfecção , Microambiente Tumoral
2.
Oncotarget ; 7(19): 28724-35, 2016 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-26895108

RESUMO

Pseudogenes are DNA sequences with high homology to the corresponding functional gene, but, because of the accumulation of various mutations, they have lost their initial functions to code for proteins. Consequently, pseudogenes have been considered until few years ago dysfunctional relatives of the corresponding ancestral genes, and then useless in the course of genome evolution. However, several studies have recently established that pseudogenes are owners of key biological functions. Indeed, some pseudogenes control the expression of functional genes by competitively binding to the miRNAs, some of them generate small interference RNAs to negatively modulate the expression of functional genes, and some of them even encode functional mutated proteins. Here, we concentrate our attention on the pseudogenes of the HMGA1 gene, that codes for the HMGA1a and HMGA1b proteins having a critical role in development and cancer progression. In this review, we analyze the family of HMGA1 pseudogenes through three aspects: classification, characterization, and their possible function and involvement in cancer.


Assuntos
Proteína HMGA1a/genética , Proteína HMGA1b/genética , Neoplasias/genética , Pseudogenes/genética , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Proteína HMGA1a/metabolismo , Proteína HMGA1b/metabolismo , Humanos , Modelos Genéticos , Mutação , Neoplasias/metabolismo , Neoplasias/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
3.
Acta Biochim Biophys Sin (Shanghai) ; 47(11): 880-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26423116

RESUMO

A large number of calmodulin-like (CML) proteins are present in plants, but there is little detailed information on the functions of these proteins in rice (Oryza sativa L.). Here, the CML3 protein from rice (OsCML3) and its truncated form lacking the C-terminal extension (OsCML3m) were found to exhibit a Ca2+-binding property and subsequent conformational change, but the ability to bind the CaM kinase II peptide was only observed for OsCML3m. Changes in their secondary structure upon Ca2+-binding measured by circular dichroism revealed that OsCML3m had a higher helical content than OsCML3. Moreover, OsCML3 was mainly localized in the plasma membrane, whereas OsCML3m was found in the nucleus. The rice high mobility group B1 (OsHMGB1) protein was identified as one of the putative OsCML3 target proteins. Bimolecular fluorescence complementation analysis revealed that OsHMGB1 bound OsCML3, OsCML3m or OsCML3s (cysteine to serine mutation at the prenylation site) in the nucleus presumably through the methionine and phenylalanine-rich hydrophobic patches, confirming that OsHMGB1 is a target protein in planta. The effect of OsCML3 or OsCML3m on the DNA-binding ability of OsHMGB1 was measured using an electrophoretic mobility shift assay. OsCML3m decreased the level of OsHMGB1 binding to pUC19 double-stranded DNA whereas OsCML3 did not. Taken together, OsCML3 probably provides a mechanism for manipulating the DNA-binding ability of OsHMGB1 in the nucleus and its C-terminal extension provides an intracellular Ca2+ regulatory switch.


Assuntos
Sinalização do Cálcio , Calmodulina/metabolismo , Proteína HMGA1b/metabolismo , Oryza/metabolismo , Proteínas de Plantas/metabolismo , Calmodulina/química , Calmodulina/genética , Proteínas de Ligação a DNA/metabolismo , Proteína HMGA1b/genética , Proteínas de Plantas/química , Proteínas de Plantas/genética
4.
Oncotarget ; 6(5): 2680-92, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25595895

RESUMO

Several recent studies have reported the Polycomb Repressive Complex 1 member CBX7 as a tumor-suppressor gene whose expression progressively decreases in different human carcinomas in relation with tumor grade, malignant stage and poor prognosis. We have previously demonstrated that CBX7 is able to inhibit the expression of the SPP1 gene, encoding the chemokine osteopontin that is over-expressed in cancer and has a critical role in cancer progression. Here, we have analyzed the mechanism by which CBX7 regulates the SPP1 gene expression. We show that the SPP1 transcriptional regulation mechanism involves the CBX7-interacting protein HMGA1b, that acts as a positive regulator of the SPP1 gene. In fact, we demonstrate that, in contrast with the transcriptional activity of CBX7, HMGA1b is able to increase the SPP1 expression by inducing the activity of its promoter. Moreover, we show that CBX7 interferes with HMGA1b on the SPP1 promoter and counteracts the positive transcriptional activity of HMGA1b on the SPP1 expression. Furthermore, since we found that also the NF-κB complex resulted involved in the modulation of the SPP1 expression in thyroid cells, we suppose that CBX7/HMGA1b/NF-κB could take part in the same transcriptional mechanism that finally leads to the regulation of the SPP1 gene expression. Taken together, our data show the important role played by CBX7 in the negative regulation of the SPP1 gene expression, thus contributing to prevent the acquisition of a malignant phenotype.


Assuntos
Carcinoma/metabolismo , Proteína HMGA1b/metabolismo , Osteopontina/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Sítios de Ligação , Carcinoma/genética , Carcinoma/patologia , Carcinoma Papilar , Linhagem Celular Tumoral , Movimento Celular , Regulação da Expressão Gênica , Células HEK293 , Proteína HMGA1b/genética , Humanos , NF-kappa B/genética , NF-kappa B/metabolismo , Osteopontina/genética , Complexo Repressor Polycomb 1/genética , Regiões Promotoras Genéticas , Transdução de Sinais , Câncer Papilífero da Tireoide , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia , Transcrição Gênica , Transfecção
5.
BMC Cancer ; 14: 694, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25245141

RESUMO

BACKGROUND: Humans and dogs are affected by squamous cell carcinomas of the oral cavity (OSCC) in a considerably high frequency. The high mobility group A2 (HMGA2) protein was found to be highly expressed in human OSCC and its expression was suggested to act as a useful predictive and prognostic tool in clinical management of oral carcinomas. Herein the expression of HMGA2 and its sister gene HMGA1 were analysed within human and canine OSCC samples. Additionally, the HMGA negatively regulating miRNAs of the let-7 family as well as the let-7 regulating gene Lin28 were also comparatively analysed. Deregulations of either one of these members could affect the progression of human and canine OSCC. METHODS: Expression levels of HMGA1, HMGA2, Lin28, let-7a and mir-98 were analysed via relative qPCR in primary human and canine OSCC, thereof derived cell lines and non-neoplastic samples. Additionally, comparative HMGA2 protein expression was analysed by immunohistochemistry. RESULTS: In both species, a significant up-regulation of the HMGA2 gene was found within the neoplastic samples while HMGA1 expression did not show significant deregulations. Comparative analyses showed down-regulation of mir-98 in human samples and up-regulation of let-7a and mir-98 in canine neoplastic samples. HMGA2 immunostainings showed higher intensities within the invasive front of the tumours than in the centre of the tumour in both species. CONCLUSIONS: HMGA2 could potentially serve as tumour marker in both species while HMGA1 might play a minor role in OSCC progression. Comparative studies indicate an inverse correlation of HMGA2 and mir-98 expression in human samples whereas in dogs no such characteristic could be found.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/veterinária , Doenças do Cão/metabolismo , Proteína HMGA2/metabolismo , MicroRNAs/metabolismo , Neoplasias Bucais/veterinária , Proteínas de Ligação a RNA/metabolismo , Adulto , Idoso , Animais , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Doenças do Cão/genética , Cães , Feminino , Expressão Gênica , Proteína HMGA1a/genética , Proteína HMGA1a/metabolismo , Proteína HMGA1b/genética , Proteína HMGA1b/metabolismo , Proteína HMGA2/genética , Humanos , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Neoplasias Bucais/genética , Neoplasias Bucais/metabolismo , Proteínas de Ligação a RNA/genética
6.
Expert Opin Ther Targets ; 18(5): 541-53, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24684280

RESUMO

INTRODUCTION: Emerging evidence demonstrates that the high mobility group A1 (HMGA1) chromatin remodeling protein is a key molecular switch required by cancer cells for tumor progression and a poorly differentiated, stem-like state. Because the HMGA1 gene and proteins are expressed at high levels in all aggressive tumors studied to date, research is needed to determine how to 'turn off' this master regulatory switch in cancer. AREAS COVERED: In this review, we describe prior studies that underscore the central role of HMGA1 in refractory cancers and we discuss approaches to target HMGA1 in cancer therapy. EXPERT OPINION: Given the widespread overexpression of HMGA1 in diverse, aggressive tumors, further research to develop technology to target HMGA1 holds immense promise as potent anticancer therapy. Previous work in preclinical models indicates that delivery of short hairpin RNA or interfering RNA molecules to 'switch off' HMGA1 expression dramatically impairs cancer cell growth and tumor progression. The advent of nanoparticle technology to systemically deliver DNA or RNA molecules to tumors brings this approach even closer to clinical applications, although further efforts are needed to translate these advances into therapies for cancer patients.


Assuntos
Antineoplásicos/farmacologia , Proteína HMGA1a/antagonistas & inibidores , Proteína HMGA1b/antagonistas & inibidores , Terapia de Alvo Molecular/métodos , Proteínas de Neoplasias/antagonistas & inibidores , Animais , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/uso terapêutico , Antineoplásicos/uso terapêutico , Diferenciação Celular , Cromatina/ultraestrutura , Ensaios Clínicos como Assunto , Inibidores de Ciclo-Oxigenase 2/farmacologia , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Progressão da Doença , Ensaios de Seleção de Medicamentos Antitumorais , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Terapia Genética , Proteína HMGA1a/genética , Proteína HMGA1a/fisiologia , Proteína HMGA1b/genética , Proteína HMGA1b/fisiologia , Humanos , Camundongos , Nanopartículas/administração & dosagem , Invasividade Neoplásica/fisiopatologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Neoplasias/tratamento farmacológico , Oxazinas/farmacologia , Oxazinas/uso terapêutico , Piperidinas/farmacologia , Piperidinas/uso terapêutico , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/farmacologia , RNA Interferente Pequeno/uso terapêutico , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
7.
Cell Biochem Funct ; 31(3): 228-36, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22961697

RESUMO

High-mobility group A1 (HMGA1) encodes proteins that act as mediators in viral integration, modification of chromatin structure, neoplastic transformation and metastatic progression. Because HMGA1 is overexpressed in most cancers and has transcriptional relationships with several Wnt-responsive genes, we explored the involvement of HMGA1 in Wnt/ß-catenin/TCF-4 signalling. In adenomatous polyposis coli (APC(Min/+)) mice, we observed significant up-regulation of HMGA1 mRNA and protein in intestinal tumours when compared with normal intestinal mucosa. Conversely, restoration of Wnt signalling by the zinc induction of wild-type APC resulted in HMGA1 down-regulation in HT-29 cells. Because APC mutations are associated with mobilization of the ß-catenin/TCF-4 transcriptional complex and subsequent activation of downstream oncogenic targets, we analyzed the 5'-flanking sequence of HMGA1 for putative TCF-4 binding elements. We identified two regions that specifically bind the ß-catenin/TCF-4 complex in vitro and in vivo, identifying HMGA1 as an immediate target of the ß-catenin/TCF-4 signalling pathway in colon cancer. Collectively, these findings strongly implicate Wnt/ß-catenin/TCF-4 signalling in regulating HMGA1 to further expand the extensive regulatory network affected by Wnt/ß-catenin/TCF-4 signalling.


Assuntos
Proteína HMGA1a/metabolismo , Proteína HMGA1b/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Regiões 5' não Traduzidas , Proteína da Polipose Adenomatosa do Colo/genética , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Regulação para Baixo/efeitos dos fármacos , Células HCT116 , Proteína HMGA1a/genética , Proteína HMGA1b/genética , Células HT29 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas , Ligação Proteica , Transdução de Sinais , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Ativação Transcricional/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Zinco/farmacologia , beta Catenina/genética
8.
Mol Biosyst ; 7(4): 1336-46, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21336378

RESUMO

The high mobility group A1 (HMGA1) are non-histone chromosomal proteins consisting of HMGA1a and HMGA1b which act as architectural transcription factors. Elevated levels of HMGA1 are reported in a number of human cancers and suggested as tumor markers. Due to their role in neoplastic transformation and tumor progression, we considered HMGA1 as a potential target for downregulation at the transcriptional level. The present paper deals with the binding of a widely used chemotherapeutic agent, adriamycin (ADM), to hmg a1 gene promoter (-304 to -284), 21RY, and its effect on the expression of hmga1 at the mRNA and protein levels and further its cytotoxic efficacy in A431 cells. A strong complex (21RY-ADM) formation caused hypochromic and bathochromic changes in UV-absorption, considerable spectral changes in circular dichroism of adriamycin and DNA and significant quenching of fluorescence emission of ADM. Thermodynamics of 21RY-ADM interaction was studied by isothermal titration and differential scanning calorimetric techniques that revealed the binding to be exothermic and favored by both negative enthalpy and positive entropy changes. Further, even low concentrations (10.3 nM) of ADM showed cytotoxicity on human squamous carcinoma cells (A431) and caused downregulation (by ∼ 70%) of hmga1 at mRNA and protein levels. Present findings clearly support the inhibitory effect of ADM on hmg a1 which quite probably is the consequence of its binding to the targeted region of hmg a1. Therefore, it appears that hmg a1 is a novel potential chemotherapeutic target in treating carcinomas of epithelial origin like prostate, breast, thyroid etc.


Assuntos
Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteína HMGA1a , Regiões Promotoras Genéticas , Antineoplásicos/química , Antineoplásicos/toxicidade , Sequência de Bases , Carcinoma de Células Escamosas , Linhagem Celular Tumoral , Doxorrubicina/química , Doxorrubicina/toxicidade , Ordem dos Genes , Proteína HMGA1a/genética , Proteína HMGA1a/metabolismo , Proteína HMGA1b/genética , Proteína HMGA1b/metabolismo , Humanos , Termodinâmica
9.
Eur J Cancer ; 47(3): 470-8, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21044834

RESUMO

HMGA1 gene rearrangements have been frequently described in human lipomas. In vitro studies suggest that HMGA1 proteins have a negative role in the control of adipocyte cell growth, and that HMGA1 gene truncation acts in a dominant-negative fashion. Therefore, to define better the role of the HMGA1 alterations in the generation of human lipomas, we generated mice carrying an Hmga1b truncated (Hmga1b/T) gene. These mice develop a giant phenotype together with a drastic expansion of the retroperitoneal and subcutaneous white adipose tissue. We show that the activation of the E2F pathway likely accounts, at least in part, for this phenotype. Interestingly, the Hmga1b/T mice also develop B-cell lymphomas similar to that occurring in Hmga1-knockout mice, supporting a dominant-negative role of the Hmga1b/T mutant also in vivo.


Assuntos
Gigantismo/genética , Proteína HMGA1b/genética , Lipomatose/genética , Linfoma de Células B/genética , Adipócitos/patologia , Animais , Proliferação de Células , Fator de Transcrição E2F1/metabolismo , Feminino , Proteína HMGA1b/metabolismo , Linfoma de Células B/metabolismo , Linfoma de Células B/patologia , Masculino , Camundongos , Camundongos Transgênicos , Mutação/genética , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Neurochem Int ; 56(6-7): 736-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20347905

RESUMO

The high-mobility group A protein 1a (HMGA1a) is a well-documented DNA-binding protein acting as an architectural transcription regulator. Recently, HMGA1a protein has been identified as a hypoxia-inducible RNA-binding trans-acting factor for aberrant splicing of presenilin-2 (PS2) pre-mRNA observed in the brains of sporadic Alzheimer's disease. Interestingly, this aberrant splicing of PS2 was also observed in the brains of bipolar disorder and schizophrenia. Many downstream genes under the control of HMGA1a could be associated with schizophrenia. On the other hand, many gene transcripts are aberrantly spliced in schizophrenia. Therefore, we examined the expression at the mRNA and protein levels of this DNA- and RNA-binding factor HMGA1a in the lymphoblastoid cell lines obtained from 16 schizophrenia patients with age-matched controls. We observed markedly higher HMGA1a mRNA and the increased HMGA1a protein in the nuclear fractions of schizophrenia patients. In contrast, there were no significant differences in the expression levels of HMGA1b, which is an alternatively spliced isoform of HMGA1a. The present study is the first to report a significant upregulation of HMGA1a in schizophrenia, suggesting its potential roles in both transcription and splicing of target genes linked with schizophrenia.


Assuntos
Proteína HMGA1a/genética , Linfócitos/química , Esquizofrenia/genética , Processamento Alternativo/genética , Linhagem Celular Transformada , Núcleo Celular/química , Expressão Gênica , Proteína HMGA1a/análise , Proteína HMGA1b/genética , Humanos , Immunoblotting , Linfócitos/ultraestrutura , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
11.
Cancer Invest ; 28(4): 340-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19916743

RESUMO

It is well known that HMGA1 group of non-histone chromosomal proteins are up-regulated in several human cancers. We studied the HMGA1 expression in squamous cell carcinoma of skin in mice followed by the treatment with Cisplatin, which is often used in combination therapies of cancers. A short course of Cisplatin treatment led to apoptotic cell death and downregulation (by 40%) of HMGA1. However, extended treatment of Cisplatin caused necrotic cell death; concomitantly HMGA1 expression decreased by 90%. Present results indicate a strong association of HMGA1 with Cisplatin-linked tumor regression. Therefore, HMGA1 could be a potential target in designing therapeutic strategies against cancer.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/tratamento farmacológico , Cisplatino/farmacologia , Proteína HMGA1a/fisiologia , Proteína HMGA1b/fisiologia , Neoplasias Cutâneas/tratamento farmacológico , Animais , Carcinoma de Células Escamosas/química , Carcinoma de Células Escamosas/patologia , Feminino , Proteína HMGA1a/análise , Proteína HMGA1a/genética , Proteína HMGA1b/análise , Proteína HMGA1b/genética , Camundongos , Neoplasias Cutâneas/química , Neoplasias Cutâneas/patologia
12.
Biol Cell ; 101(9): 511-24, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19250063

RESUMO

BACKGROUND INFORMATION: miRNAs (microRNAs) are a class of non-coding RNAs that inhibit gene expression by binding to recognition elements, mainly in the 3' UTR (untranslated region) of mRNA. A single miRNA can target several hundred mRNAs, leading to a complex metabolic network. miR-16 (miRNA-16), located on chromosome 13q14, is involved in cell proliferation and apoptosis regulation; it may interfere with either oncogenic or tumour suppressor pathways, and is implicated in leukaemogenesis. These data prompted us to search for and validate novel targets of miR-16. RESULTS: In the present study, by using a combined bioinformatics and molecular approach, we identified two novel putative targets of miR-16, caprin-1 (cytoplasmic activation/proliferation-associated protein-1) and HMGA1 (high-mobility group A1), and we also studied cyclin E which had been previously recognized as an miR-16 target by bioinformatics database. Using luciferase activity assays, we demonstrated that miR-16 interacts with the 3' UTR of the three target mRNAs. We showed that miR-16, in MCF-7 and HeLa cell lines, down-regulates the expression of caprin-1, HMGA1a, HMGA1b and cyclin E at the protein level, and of cyclin E, HMGA1a and HMGA1b at the mRNA levels. CONCLUSIONS: Taken together, our data demonstrated that miR-16 can negatively regulate two new targets, HMGA1 and caprin-1, which are involved in cell proliferation. In addition, we also showed that the inhibition of cyclin E expression was due, at least in part, to a decrease in its mRNA stability.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Proteína HMGA1a/metabolismo , MicroRNAs/metabolismo , Sequência de Aminoácidos , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Proteína HMGA1a/química , Proteína HMGA1a/genética , Proteína HMGA1b/química , Proteína HMGA1b/genética , Proteína HMGA1b/metabolismo , Humanos , MicroRNAs/química , MicroRNAs/genética , Dados de Sequência Molecular , Ligação Proteica , Alinhamento de Sequência
13.
J Med Chem ; 52(5): 1247-50, 2009 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-19215087

RESUMO

A three-dimensional pharmacophore model was generated utilizing a set of known inhibitors of c-Myc-Max heterodimer formation. The model successfully identified a set of structurally diverse compounds with potential inhibitory activity against c-Myc. Nine compounds were tested in vitro, and four displayed affinities in the micromolar range and growth inhibitory activity against c-Myc-overexpressing cells. These studies demonstrate the applicability of pharmacophore modeling to the identification of novel and potentially more puissant inhibitors of the c-Myc oncoprotein.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/química , Modelos Moleculares , Proteínas Proto-Oncogênicas c-myc/química , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Benzofuranos/química , Benzofuranos/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Dicroísmo Circular , Ensaio de Desvio de Mobilidade Eletroforética , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Técnicas de Inativação de Genes , Células HL-60 , Proteína HMGA1b/biossíntese , Proteína HMGA1b/genética , Humanos , Ligação Proteica , Multimerização Proteica , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Piridinas/química , Piridinas/farmacologia , Pirrolidinas/química , Pirrolidinas/farmacologia , Ratos , Tiazóis/química , Tiazóis/farmacologia
14.
Oncogene ; 28(11): 1432-42, 2009 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-19169275

RESUMO

The High Mobility Group proteins HMGA1 are nuclear architectural factors that play a critical role in a wide range of biological processes. Since recent studies have identified the microRNAs (miRNAs) as important regulators of gene expression, modulating critical cellular functions such as proliferation, apoptosis and differentiation, the aim of our work was to identify the miRNAs that are physiologically regulated by HMGA1 proteins. To this purpose, we have analysed the miRNA expression profile of mouse embryonic fibroblasts (MEFs) carrying two, one or no Hmga1 functional alleles using a microarray (miRNA microarray). By this approach, we found a miRNA expression profile that differentiates Hmga1-null MEFs from the wild-type ones. In particular, a significant decrease in miR-196a-2, miR-101b, miR-331 and miR-29a was detected in homozygous Hmga1-knockout MEFs in comparison with wild-type cells. Consistently, these miRNAs are downregulated in most of the analysed tissues of Hmga1-null mice in comparison with the wild-type mice. ChIP assay shows that HMGA1 is able to bind regions upstream of these miRNAs. Moreover, we identified the HMGA2 gene product as a putative target of miR-196a-2, suggesting that HMGA1 proteins are able to downregulate the expression of the other member of the HMGA family through the regulation of the miR-196a-2 expression. Finally, ATXN1 and STC1 gene products have been identified as targets of miR-101b. Therefore, it is reasonable to hypothesize that HMGA1 proteins are involved in several functions by regulating miRNA expression.


Assuntos
Regulação da Expressão Gênica , Proteína HMGA1a/fisiologia , Proteína HMGA1b/fisiologia , MicroRNAs/genética , Animais , Ataxina-1 , Ataxinas , Perfilação da Expressão Gênica , Glicoproteínas/genética , Glicoproteínas/fisiologia , Proteína HMGA1a/genética , Proteína HMGA1b/genética , Camundongos , MicroRNAs/fisiologia , Células NIH 3T3 , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia
15.
J Biol Chem ; 282(18): 13363-71, 2007 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-17350957

RESUMO

Members of the HMGA1 (high mobility group A1) family of architectural transcription factors, HMGA1a and HMGA1b, play important roles in many normal cellular processes and in tumorigenesis. We performed a yeast two-hybrid screen for HMGA1-interacting proteins and identified the SUMO E2 conjugase Ubc9 as one such partner. The Ubc9-interacting domain of HMGA1 is bipartite, consisting of a proline-rich region near the N terminus and an acidic domain at the extreme C terminus, whereas the HMGA1-interacting domain of Ubc9 comprises a single region previously shown to associate with and SUMOylate other transcription factors. Consistent with these findings, endogenous HMGA1 proteins and Ubc9 could be co-immunoprecipitated from several human cell lines. Studies with HMGA1b proteins containing mutations of either or both Ubc9-interacting domains and with Ubc9-depleted cell lines indicated that the proline-rich domain of HMGA1b positively influences transformation and growth, whereas the acidic domain negatively influences these properties. None of the changes in HMGA1 protein functions mediated by Ubc9 appears to require SUMOylation. These findings are consistent with the idea that Ubc9 can act as both a positive and negative regulator of proliferation and transformation via its non-SUMO-dependent interaction with HMGA1 proteins.


Assuntos
Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Proteína HMGA1a/metabolismo , Processamento de Proteína Pós-Traducional , Proteína SUMO-1/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Animais , Linhagem Celular , Transformação Celular Neoplásica/genética , Proteína HMGA1a/genética , Proteína HMGA1b/genética , Proteína HMGA1b/metabolismo , Humanos , Mutação , Processamento de Proteína Pós-Traducional/genética , Estrutura Terciária de Proteína/genética , Proteína SUMO-1/genética , Técnicas do Sistema de Duplo-Híbrido , Enzimas de Conjugação de Ubiquitina/genética
16.
Gene ; 330: 93-9, 2004 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-15087128

RESUMO

Due to the emerging advantages of numerous canine diseases as a genetic model for their human orthologs, the dog could join the mouse as the species of choice to unravel genetic mechanisms, e.g. of cancer predisposition, development and progression. However, precondition for such studies is the characterisation of the corresponding canine genes. Human and murine HMGA1 non-histone proteins participate in a wide variety of cellular processes including regulation of inducible gene transcription, integration of retroviruses into chromosomes, and the induction of neoplastic transformation and promotion of metastatic progression of cancer cells. Chromosomal aberrations affecting the human HMGA1 gene at 6p21 were described in several tumours like pulmonary chondroid hamartomas, uterine leiomyomas, follicular thyroid adenomas and others. Over-expression of the proteins of HMGA1 is characteristic for various malignant tumours suggesting a relation between high titer of the protein and the neoplastic phenotype. In this study, we characterised the molecular structure of the canine HMGA1 cDNA, its splice variants and predicted proteins HMGA1a and HMGA1b. Furthermore, we compared the coding sequence(s) (CDS) of both splice variants for 12 different breeds, screened them for single nucleotide polymorphisms (SNPs) and characterised a basic expression pattern.


Assuntos
Cães/genética , Proteína HMGA1a/genética , Proteína HMGA1b/genética , Processamento Alternativo , Sequência de Aminoácidos , Animais , DNA Complementar/química , DNA Complementar/genética , Éxons , Expressão Gênica , Genes/genética , Íntrons , Masculino , Dados de Sequência Molecular , Alinhamento de Sequência , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
17.
Mol Med ; 9(5-8): 154-65, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14571323

RESUMO

To assess whether retinoblastoma formation is associated with the expression of high mobility group (HMG) A2 protein, a transcription factor that is highly expressed during embryogenesis and completely repressed in normal adult tissues, we performed Northern and Western blots and RT-PCR analyses, and immunohistochemistry to test for HMGA2 expression. We used established retinoblastoma cell lines in tumors grown in nude mice and clinical retinoblastoma specimens, and contrasted these tumors with normal embryonic and adult retina. Adenoviral-mediated antisense experiments were conducted on the retinoblastoma cell lines to suppress HMGA2 expression and determine if cell proliferation is HMGA2-dependent. We also transfected a retinoblastoma cell line to identify cis-regulatory elements and transcription initiation sites on the HMGA2 gene promoter. HMGA2 gene expression was silenced in terminally differentiated retina of 6-wk-old mice, but it was detected in retina of a 13.5-d postcoitum embryo. Reactivation of HMGA2 gene expression was observed in the retinoblastoma cell lines Y79, WERI-Rb1, and TOTL-1, in tumors derived from some of these cells propagated in nude mice, and in a high frequency of retinoblastomas excised from human patients. This suggests that expression of HMGA2 gene in retinoblastoma cells involves a derepression process. By using an antisense approach to block HMGA2 expression, we observed a decrease in the number of proliferating retinoblastoma cells. As a 1st step toward understanding HMGA2 gene reactivation in retinoblastoma, we mapped the 2 transcription initiation sites and associated positive regulatory elements within the WERI-Rb1 cells. Our discovery of derepression of HMGA2 gene expression in retinoblastoma provides the 1st evidence that this protein might contribute to neoplastic transformation of retina cells.


Assuntos
Proteína HMGA2/genética , Retinoblastoma/metabolismo , Animais , Sequência de Bases , Divisão Celular/fisiologia , Proteína HMGA1a/biossíntese , Proteína HMGA1a/genética , Proteína HMGA1b/biossíntese , Proteína HMGA1b/genética , Proteína HMGA2/biossíntese , Humanos , Técnicas In Vitro , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Regiões Promotoras Genéticas , RNA Antissenso/metabolismo , Células Tumorais Cultivadas
18.
Nucleic Acids Res ; 31(17): 5025-32, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12930952

RESUMO

The high mobility group A (HMGA) family of non-histone chromosomal proteins is encoded by two related genes, HMGA1 and HMGA2. HMGA proteins are architectural transcription factors that have been found to regulate the transcription of a large number of genes. They are also some of the most commonly dysregulated genes in human neoplasias, highlighting a role in growth control. HMGA1 and HMGA2 have also been found to stimulate retroviral integration in vitro. In this study, we have cloned chicken HMGA1, and used the chicken DT40 B-cell lymphoma line to generate cells lacking HMGA1, HMGA2 and both in combination. We tested these lines for effects on cellular growth, gene control and retroviral integration. Surprisingly, we found that the HMGA gene family is dispensable for growth in DT40 cells, and that there is no apparent defect in retroviral integration in the absence of HMGA1 or HMGA2. We also analyzed the activity of approximately 4000 chicken genes, but found no significant changes. We conclude that HMGA proteins are not strictly required for growth control or retroviral integration in DT40 cells and may well be redundant with other factors.


Assuntos
Proteínas HMGA/genética , Família Multigênica/genética , Sequência de Aminoácidos , Animais , Divisão Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Embrião de Galinha , Galinhas , Clonagem Molecular , DNA Complementar/química , DNA Complementar/genética , Expressão Gênica , Perfilação da Expressão Gênica , Vetores Genéticos/genética , Genótipo , Proteína HMGA1b/genética , Proteína HMGA2/genética , Humanos , Lentivirus/genética , Luciferases/genética , Luciferases/metabolismo , Dados de Sequência Molecular , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Retroviridae/genética , Alinhamento de Sequência , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos
19.
Mol Cell Biol ; 23(7): 2225-38, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12640109

RESUMO

A drastic reduction in BRCA1 gene expression is a characteristic feature of aggressive sporadic breast carcinoma. However, the mechanisms underlying BRCA1 downregulation in breast cancer are not well understood. Here we report that both in vitro and in vivo HMGA1b protein binds to and inhibits the activity of both human and mouse BRCA1 promoters. Consistently, murine embryonic stem (ES) cells with the Hmga1 gene deleted display higher Brca1 mRNA and protein levels than do wild-type ES cells. Stable transfection of MCF-7 cells with the HMGA1b cDNA results in a decrease of BRCA1 gene expression and in a lack of BRCA1 induction after estrogen treatment. Finally, we found an inverse correlation between HMGA1 and BRCA1 mRNA and protein expression in human mammary carcinoma cell lines and tissues. These data indicate that HMGA1 proteins are involved in transcriptional regulation of the BRCA1 gene, and their overexpression may have a role in BRCA1 downregulation observed in aggressive mammary carcinomas.


Assuntos
Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Neoplasias da Mama/metabolismo , Carcinoma/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteína HMGA1b/metabolismo , Regiões Promotoras Genéticas/fisiologia , Animais , Proteína BRCA1/deficiência , Neoplasias da Mama/genética , Carcinoma/genética , Linhagem Celular , Estrogênios/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Marcação de Genes , Genes Reporter , Proteína HMGA1b/genética , Proteína HMGA1b/farmacologia , Humanos , Camundongos , Regiões Promotoras Genéticas/efeitos dos fármacos , Ligação Proteica/genética , RNA Mensageiro/metabolismo , Fase S , Células-Tronco/citologia , Células-Tronco/metabolismo , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...