Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Int J Mol Sci ; 25(16)2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39201549

RESUMO

The "AT-hook" is a peculiar DNA-binding domain that interacts with DNA in the minor groove in correspondence to AT-rich sequences. This domain has been first described in the HMGA protein family of architectural factors and later in various transcription factors and chromatin proteins, often in association with major groove DNA-binding domains. In this review, using a literature search, we identified about one hundred AT-hook-containing proteins, mainly chromatin proteins and transcription factors. After considering the prototypes of AT-hook-containing proteins, the HMGA family, we review those that have been studied in more detail and that have been involved in various pathologies with a particular focus on cancer. This review shows that the AT-hook is a domain that gives proteins not only the ability to interact with DNA but also with RNA and proteins. This domain can have enzymatic activity and can influence the activity of the major groove DNA-binding domain and chromatin docking modules when present, and its activity can be modulated by post-translational modifications. Future research on the function of AT-hook-containing proteins will allow us to better decipher their function and contribution to the different pathologies and to eventually uncover their mutual influences.


Assuntos
DNA , Ligação Proteica , Humanos , DNA/metabolismo , DNA/química , Animais , Motivos AT-Hook , Cromatina/metabolismo , Sítios de Ligação , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/química , Domínios Proteicos , Proteínas HMGA/metabolismo , Proteínas HMGA/genética , Conformação de Ácido Nucleico
2.
Int J Mol Sci ; 23(15)2022 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-35897685

RESUMO

Diet and lifestyle factors greatly affect health and susceptibility to diseases, including cancer. Stem cells' functions, including their ability to divide asymmetrically, set the rules for tissue homeostasis, contribute to health maintenance, and represent the entry point of cancer occurrence. Stem cell properties result from the complex integration of intrinsic, extrinsic, and systemic factors. In this context, diet-induced metabolic changes can have a profound impact on stem cell fate determination, lineage specification and differentiation. The purpose of this review is to provide a comprehensive description of the multiple "non-metabolic" effects of diet on stem cell functions, including little-known effects such as those on liquid-liquid phase separation and on non-random chromosome segregation (asymmetric division). A deep understanding of the specific dietetic requirements of normal and cancer stem cells may pave the way for the development of nutrition-based targeted therapeutic approaches to improve regenerative and anticancer therapies.


Assuntos
Neoplasias , Células-Tronco Neoplásicas , Diferenciação Celular/fisiologia , Segregação de Cromossomos , Dieta , Homeostase
3.
Int J Mol Sci ; 23(2)2022 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-35054987

RESUMO

The transition between epithelial and mesenchymal phenotype is emerging as a key determinant of tumor cell invasion and metastasis. It is a plastic process in which epithelial cells first acquire the ability to invade the extracellular matrix and migrate into the bloodstream via transdifferentiation into mesenchymal cells, a phenomenon known as epithelial-mesenchymal transition (EMT), and then reacquire the epithelial phenotype, the reverse process called mesenchymal-epithelial transition (MET), to colonize a new organ. During all metastatic stages, metabolic changes, which give cancer cells the ability to adapt to increased energy demand and to withstand a hostile new environment, are also important determinants of successful cancer progression. In this review, we describe the complex interaction between EMT and metabolism during tumor progression. First, we outline the main connections between the two processes, with particular emphasis on the role of cancer stem cells and LncRNAs. Then, we focus on some specific cancers, such as breast, lung, and thyroid cancer.


Assuntos
Metabolismo Energético , Transição Epitelial-Mesenquimal , Neoplasias/metabolismo , Neoplasias/patologia , Animais , Biomarcadores , Biomarcadores Tumorais , Gerenciamento Clínico , Progressão da Doença , Suscetibilidade a Doenças , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Redes e Vias Metabólicas , Terapia de Alvo Molecular , Neoplasias/etiologia , Neoplasias/terapia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Especificidade de Órgãos , Estresse Oxidativo , Interferência de RNA , RNA Longo não Codificante/genética , Transdução de Sinais , Fatores de Transcrição/metabolismo , Microambiente Tumoral
4.
Int J Mol Sci ; 21(3)2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-31979076

RESUMO

High mobility group A (HMGA) proteins are oncofoetal chromatin architectural factors that are widely involved in regulating gene expression. These proteins are unique, because they are highly expressed in embryonic and cancer cells, where they play a relevant role in cell proliferation, stemness, and the acquisition of aggressive tumour traits, i.e., motility, invasiveness, and metastatic properties. The HMGA protein expression levels and activities are controlled by a connected set of events at the transcriptional, post-transcriptional, and post-translational levels. In fact, microRNA (miRNA)-mediated RNA stability is the most-studied mechanism of HMGA protein expression modulation. In this review, we contribute to a comprehensive overview of HMGA-targeting miRNAs; we provide detailed information regarding HMGA gene structural organization and a comprehensive evaluation and description of HMGA-targeting miRNAs, while focusing on those that are widely involved in HMGA regulation; and, we aim to offer insights into HMGA-miRNA mutual cross-talk from a functional and cancer-related perspective, highlighting possible clinical implications.


Assuntos
Cromatina/genética , Proteínas HMGA/genética , MicroRNAs/genética , Animais , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias/genética
5.
J Biol Chem ; 289(25): 17425, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-29874323

RESUMO

[This retracts the article on p. 25993 in vol. 284, PMID: 19633359.].

6.
Cancer Cell ; 11(3): 275-89, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17349584

RESUMO

The FEZ1/LZTS1 (LZTS1) protein is frequently downregulated in human cancers of different histotypes. LZTS1 is expressed in normal tissues, and its introduction in cancer cells inhibits cell growth and suppresses tumorigenicity, owing to an accumulation of cells in G2/M. Here, we define its role in cell cycle regulation and tumor progression by generating Lzts1 knockout mice. In Lzts1(-/-) mouse embryo fibroblasts (MEFs), Cdc25C degradation was increased during M phase, resulting in decreased Cdk1 activity. As a consequence, Lzts1(-/-) MEFs showed accelerated mitotic progression, resistance to taxol- and nocodazole-induced M phase arrest, and improper chromosome segregation. Accordingly, Lzts1 deficiency was associated with an increased incidence of both spontaneous and carcinogen-induced cancers in mice.


Assuntos
Proteína Quinase CDC2/fisiologia , Proteínas de Ciclo Celular/fisiologia , Transformação Celular Neoplásica , Mitose , Neoplasias Gástricas/patologia , Proteínas Supressoras de Tumor/fisiologia , Fosfatases cdc25/fisiologia , Animais , Antineoplásicos/farmacologia , Carcinógenos , Divisão Celular , Células Cultivadas , Segregação de Cromossomos , Dimetilnitrosamina/análogos & derivados , Fibroblastos/metabolismo , Fibroblastos/patologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Nocodazol/farmacologia , Paclitaxel/farmacologia , Neoplasias Gástricas/induzido quimicamente , Neoplasias Gástricas/genética , Proteínas Supressoras de Tumor/genética
7.
Cancer Cell ; 9(6): 459-71, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16766265

RESUMO

HMGA2 gene amplification and overexpression in human prolactinomas and the development of pituitary adenomas in HMGA2 transgenic mice showed that HMGA2 plays a crucial role in pituitary tumorigenesis. We have explored the pRB/E2F1 pathway to investigate the mechanism by which HMGA2 acts. Here we show that HMGA2 interacts with pRB and induces E2F1 activity in mouse pituitary adenomas by displacing HDAC1 from the pRB/E2F1 complex-a process that results in E2F1 acetylation. We found that loss of E2F1 function (obtained by mating HMGA2 and E2F1(-/-) mice) suppressed pituitary tumorigenesis in HMGA2 mice. Thus, HMGA2-mediated E2F1 activation is a crucial event in the onset of these tumors in transgenic mice and probably also in human prolactinomas.


Assuntos
Fator de Transcrição E2F1/fisiologia , Proteína HMGA2/fisiologia , Neoplasias Hipofisárias/metabolismo , Acetilação , Animais , Linhagem Celular , Proliferação de Células , Transformação Celular Neoplásica , DNA/metabolismo , Fator de Transcrição E2F1/genética , Ativação Enzimática , Proteína HMGA2/biossíntese , Proteína HMGA2/genética , Histona Desacetilase 1 , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Camundongos , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Neoplasias Hipofisárias/patologia , Regiões Promotoras Genéticas , Ligação Proteica , Elementos de Resposta , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais
8.
Cells ; 13(4)2024 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-38391963

RESUMO

The classification of tumors into subtypes, characterized by phenotypes determined by specific differentiation pathways, aids diagnosis and directs therapy towards targeted approaches. However, with the advent and explosion of next-generation sequencing, cancer phenotypes are turning out to be far more heterogenous than initially thought, and the classification is continually being updated to include more subtypes. Tumors are indeed highly dynamic, and they can evolve and undergo various changes in their characteristics during disease progression. The picture becomes even more complex when the tumor responds to a therapy. In all these cases, cancer cells acquire the ability to transdifferentiate, changing subtype, and adapt to changing microenvironments. These modifications affect the tumor's growth rate, invasiveness, response to treatment, and overall clinical behavior. Studying tumor subtype transitions is crucial for understanding tumor evolution, predicting disease outcomes, and developing personalized treatment strategies. We discuss this emerging hallmark of cancer and the molecular mechanisms involved at the crossroads between tumor cells and their microenvironment, focusing on four different human cancers in which tissue plasticity causes a subtype switch: breast cancer, prostate cancer, glioblastoma, and pancreatic adenocarcinoma.


Assuntos
Adenocarcinoma , Neoplasias da Mama , Neoplasias Pancreáticas , Masculino , Humanos , Transdiferenciação Celular , Processos Neoplásicos , Neoplasias da Mama/patologia , Microambiente Tumoral/genética
9.
Antioxid Redox Signal ; 40(4-6): 236-249, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-36851901

RESUMO

Significance: Metabolic end products and intermediates can exert signaling functions as chemical sources for histone posttranslational modifications, which remodel chromatin and affect gene expression. Among them, lactic acid is responsible for histone lactylation, a recently discovered histone mark that occurs in high lactate conditions, such as those resulting from the Warburg effect in cancer cells. Recent Advances: Late-breaking studies have advanced the knowledge on the mechanisms involved in histone lactylation, requiring independent nonenzyme and enzyme-dependent reactions, which is emerging as an important hallmark of cancer cells linking metabolic changes to gene expression reprogramming. Critical Issues: In this study, we give an overview about this new epigenetic modification, focusing on its mechanism of action in tumors and tumor microenvironment. Future Directions: Further investigation on the competition mechanism between lactylation and acetylation, as well as on the mechanisms by which lactate fluctuation can control a specific gene set in a given tissue, is needed in the coming years to exploit new anticancer therapeutic approaches. Antioxid. Redox Signal. 40, 236-249.


Assuntos
Histonas , Neoplasias , Humanos , Histonas/metabolismo , Ácido Láctico , Cromatina , Epigênese Genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Microambiente Tumoral
10.
Cancers (Basel) ; 15(7)2023 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-37046851

RESUMO

Non-small cell lung cancer (NSCLC), the leading cause of cancer death worldwide, is still an unmet medical problem due to the lack of both effective therapies against advanced stages and markers to allow a diagnosis of the disease at early stages before its progression. Immunotherapy targeting the PD-1/PD-L1 checkpoint is promising for many cancers, including NSCLC, but its success depends on the tumor expression of PD-L1. PATZ1 is an emerging cancer-related transcriptional regulator and diagnostic/prognostic biomarker in different malignant tumors, but its role in lung cancer is still obscure. Here we investigated expression and role of PATZ1 in NSCLC, in correlation with NSCLC subtypes and PD-L1 expression. A cohort of 104 NSCLCs, including lung squamous cell carcinomas (LUSCs) and adenocarcinomas (LUADs), was retrospectively analyzed by immunohistochemistry for the expression of PATZ1 and PD-L1. The results were correlated with each other and with the clinical characteristics, showing on the one hand a positive correlation between the high expression of PATZ1 and the LUSC subtype and, on the other hand, a negative correlation between PATZ1 and PD-L1, validated at the mRNA level in independent NSCLC datasets. Consistently, two NSCLC cell lines transfected with a PATZ1-overexpressing plasmid showed PD-L1 downregulation, suggesting a role for PATZ1 in the negative regulation of PD-L1. We also showed that PATZ1 overexpression inhibits NSCLC cell proliferation, migration, and invasion, and that Patz1-knockout mice develop LUAD. Overall, this suggests that PATZ1 may act as a tumor suppressor in NSCLC.

11.
J Biol Chem ; 284(38): 25993-6004, 2009 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-19633359

RESUMO

It is generally accepted that the regulation of adipogenesis prevents obesity. However, the mechanisms controlling adipogenesis have not been completely defined. We have previously demonstrated that HMGA1 proteins play a critical role in adipogenesis. In fact, suppression of HMGA1 protein synthesis by antisense technology dramatically increased growth rate and impaired adipocyte differentiation in 3T3-L1 cells. Furthermore, we showed that HMGA1 strongly potentiates the capacity of the CCAAT/enhancer-binding protein beta (C/EBPbeta) transcriptional factor to transactivate the leptin promoter, an adipocytic-specific promoter. In this study we demonstrate that HMGA1 physically interacts with retinoblastoma protein (RB), which is also required in adipocyte differentiation. Moreover, we show that RB, C/EBPbeta, and HMGA1 proteins all cooperate in controlling both Id1 and leptin gene transcriptions, which are down- and up-regulated during adipocyte differentiation, respectively. We also demonstrate that HMGA1/RB interaction regulates CDC25A and CDC6 promoter activities, which are induced by E2F-1 protein during early adipocyte differentiation, by displacing HDAC1 from the RB-E2F1 complex. Furthermore, by using Hmga1(-/-) embryonic stem cells, which failed to undergo adipocyte differentiation, we show the crucial role of HMGA1 proteins in adipocyte differentiation due to its pivotal involvement in the formation of the RB-C/EBPbeta complex. Altogether these data demonstrate a key role of the interaction between HMGA1 and RB in adipocyte differentiation.


Assuntos
Adipócitos/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Proteínas HMGA/metabolismo , Proteína do Retinoblastoma/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Fator de Transcrição E2F1/biossíntese , Fator de Transcrição E2F1/genética , Células-Tronco Embrionárias/citologia , Técnicas de Silenciamento de Genes , Proteínas HMGA/genética , Histona Desacetilase 1 , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Leptina/biossíntese , Leptina/genética , Camundongos , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/fisiologia , Ligação Proteica/fisiologia , Proteína do Retinoblastoma/genética , Transcrição Gênica/fisiologia , Ativação Transcricional/fisiologia , Fosfatases cdc25/biossíntese , Fosfatases cdc25/genética
12.
Sci Rep ; 10(1): 18850, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-33139812

RESUMO

The mammalian high mobility group protein AT-hook 2 (HMGA2) is a multi-functional DNA-binding protein that plays important roles in tumorigenesis and adipogenesis. Previous results showed that HMGA2 is a potential therapeutic target of anticancer and anti-obesity drugs by inhibiting its DNA-binding activities. Here we report the development of a miniaturized, automated AlphaScreen ultra-high-throughput screening assay to identify inhibitors targeting HMGA2-DNA interactions. After screening the LOPAC1280 compound library, we identified several compounds that strongly inhibit HMGA2-DNA interactions including suramin, a century-old, negatively charged antiparasitic drug. Our results show that the inhibition is likely through suramin binding to the "AT-hook" DNA-binding motifs and therefore preventing HMGA2 from binding to the minor groove of AT-rich DNA sequences. Since HMGA1 proteins also carry multiple "AT-hook" DNA-binding motifs, suramin is expected to inhibit HMGA1-DNA interactions as well. Biochemical and biophysical studies show that charge-charge interactions and hydrogen bonding between the suramin sulfonated groups and Arg/Lys residues play critical roles in the binding of suramin to the "AT-hook" DNA-binding motifs. Furthermore, our results suggest that HMGA2 may be one of suramin's cellular targets.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , Proteína HMGA1a/antagonistas & inibidores , Proteína HMGA2/antagonistas & inibidores , Suramina/química , Adipogenia/efeitos dos fármacos , Motivos de Aminoácidos/efeitos dos fármacos , Sequência de Bases/efeitos dos fármacos , Sítios de Ligação/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteína HMGA1a/química , Proteína HMGA1a/genética , Proteína HMGA2/química , Proteína HMGA2/genética , Ensaios de Triagem em Larga Escala , Humanos , Suramina/isolamento & purificação , Suramina/farmacologia
14.
Cell Cycle ; 18(13): 1446-1457, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31116627

RESUMO

Glioblastoma (GBM) is a lethal, fast-growing brain cancer, affecting 2-3 per 100,000 adults per year. It arises from multipotent neural stem cells which have reduced their ability to divide asymmetrically and hence divide symmetrically, generating increasing number of cancer stem cells, fostering tumor growth. We have previously demonstrated that the architectural transcription factor HMGA1 is highly expressed in brain tumor stem cells (BTSCs) and that its silencing increases stem cell quiescence, reduces self-renewal and sphere-forming efficiency in serial passages, suggesting a shift from symmetric to asymmetric division. Since NUMB expression is fundamental for the fulfillment of asymmetric division in stem cells, and is lost or reduced in many tumors, including GBM, we have investigated the ability of HMGA1 to regulate NUMB expression. Here, we show that HMGA1 negatively regulates NUMB expression at transcriptional level, by binding its promoter and counteracting c/EBP-ß and at posttranscriptional level, by regulating the expression of MSI1 and of miR-146a. Finally, we report that HMGA1 knockdown-induced NUMB upregulation leads to the downregulation of the NOTCH1 pathway. Therefore, the data reported here indicate that HMGA1 negatively regulates NUMB expression in BTSCs, further supporting HMGA1 targeting as innovative and effective anti-cancer therapy.


Assuntos
Neoplasias Encefálicas/genética , Glioblastoma/genética , Proteína HMGA1a/genética , Proteínas de Membrana/genética , Células-Tronco Neoplásicas/patologia , Proteínas do Tecido Nervoso/genética , Processamento Pós-Transcricional do RNA/genética , Transcrição Gênica/genética , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Divisão Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Regulação para Baixo/genética , Glioblastoma/patologia , Células HEK293 , Humanos , MicroRNAs/genética , Células-Tronco Multipotentes/patologia , Regiões Promotoras Genéticas/genética
15.
Cancer Res ; 66(5): 2536-43, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16510570

RESUMO

The HMGA1 protein is a major factor in chromatin architecture and gene control. It plays a critical role in neoplastic transformation. In fact, blockage of HMGA1 synthesis prevents rat thyroid cell transformation by murine transforming retroviruses, and an adenovirus carrying the HMGA1 gene in the antisense orientation induces apoptotic cell death in anaplastic human thyroid carcinoma cell lines, but not in normal thyroid cells. Moreover, both in vitro and in vivo studies have established the oncogenic role of the HMGA1 gene. In this study, to define HMGA1 function in vivo, we examined the consequences of disrupting the Hmga1 gene in mice. Both heterozygous and homozygous mice for the Hmga1-null allele show cardiac hypertrophy due to the direct role of HMGA1 on cardiomyocytic cell growth regulation. These mice also developed hematologic malignancies, including B cell lymphoma and myeloid granuloerythroblastic leukemia. The B cell expansion and the increased expression of the RAG1/2 endonuclease, observed in HMGA1-knockout spleen tissues, might be responsible for the high rate of abnormal IgH rearrangements observed in these neoplasias. Therefore, the data reported here indicate the critical role of HMGA1 in heart development and growth, and reveal an unsuspected antioncogenic potential for this gene in hematologic malignancies.


Assuntos
Cardiomegalia/genética , Proteína HMGA1a/genética , Transtornos Linfoproliferativos/genética , Animais , Linfócitos B/imunologia , Linfócitos B/patologia , Cardiomegalia/imunologia , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica , Proteína HMGA1a/deficiência , Proteína HMGA1a/imunologia , Haploidia , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Interleucinas/biossíntese , Interleucinas/genética , Linfoma de Células B/genética , Linfoma de Células B/imunologia , Transtornos Linfoproliferativos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Ratos Wistar
17.
Mol Cell Biol ; 23(11): 3774-87, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12748281

RESUMO

Since the majority of high-grade breast cancers express reduced levels of BRCA1 mRNA, we investigated the factors regulating BRCA1 transcription. Factors with specific affinity for the previously identified positive regulatory region (PRR) in the BRCA1 promoter were purified from whole-cell extracts. Identified proteins included replication protein A and a series of related factors with affinity for the sense strand of PRR. A subset of the identified factors activated the BRCA1 promoter. Identification of these families of proteins regulating the BRCA1 promoter represents an important step in the comprehension of the mechanisms responsible for breast cancer development.


Assuntos
DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Genes BRCA1 , Transcrição Gênica , Sequência de Bases , Neoplasias da Mama/genética , Proteínas de Ligação a DNA/isolamento & purificação , Feminino , Humanos , Dados de Sequência Molecular , Mutação Puntual , Regiões Promotoras Genéticas , Células Tumorais Cultivadas
18.
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
19.
J Biomed Mater Res A ; 80(2): 297-305, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16960833

RESUMO

The fate of any tissue engineering implant relies upon an adequate oxygen and nutrients supply throughout the cellular construct and, hence, by the ability of the scaffold to induce and guide vascular ingrowth. However, implant vascularization is usually an uncontrolled process that takes several weeks. In this work, we assessed the feasibility of controlling vascular sprout rate and direction within three-dimensional collagen-hyaluronic acid semi-interpenetrated networks by modulating the spatial distribution of the matricellular cues. Results indicated that increasing amount of hyaluronic acid (HA) within the matrix led to a progressive inhibition of sprouting. In HA-rich matrices, the sprout number and the propagation rate showed a 2.7- and 4-fold reduction, respectively, compared to collagen matrices. Furthermore, by creating HA gradients within the collagen network, we were able to direct and enhance the sprouting rate. This study provides an experimental platform for controlling vascularization of engineered tissues.


Assuntos
Neovascularização Fisiológica , Engenharia Tecidual/métodos , Animais , Bovinos , Colágeno , Meios de Cultura , Ácido Hialurônico/farmacologia , Cinética , Neovascularização Fisiológica/efeitos dos fármacos
20.
J Mol Med (Berl) ; 95(4): 353-360, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28293697

RESUMO

The high-mobility group A (HMGA) proteins are frequently overexpressed in human malignancies and correlate with the presence of metastases and reduced patient survival. Here, we highlight the main studies evidencing a critical role of HMGA in chemoresistance, mainly by activating Akt signaling, impairing p53 activity, and regulating the expression of microRNAs that target genes involved in the susceptibility of cancer cells to antineoplastic agents. Therefore, these studies account for the association of HMGA overexpression with patient poor outcome, indicating the impairment of HMGA as a fascinating perspective for effectively improving cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Proteínas HMGA/genética , MicroRNAs/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas HMGA/metabolismo , Humanos , MicroRNAs/metabolismo , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA