Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 15(19)2023 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-37835417

RESUMO

MicroRNA (miRNA) are small noncoding RNAs that play vital roles in post-transcriptional gene regulation by inhibiting mRNA translation or promoting mRNA degradation. The dysregulation of miRNA has been implicated in numerous human diseases, including cancers. miR-34 family members (miR-34s), including miR-34a, miR-34b, and miR-34c, have emerged as the most extensively studied tumor-suppressive miRNAs. In this comprehensive review, we aim to provide an overview of the major signaling pathways and gene networks regulated by miR-34s in various cancers and highlight the critical tumor suppressor role of miR-34s. Furthermore, we will discuss the potential of using miR-34 mimics as a novel therapeutic approach against cancer, while also addressing the challenges associated with their development and delivery. It is anticipated that gaining a deeper understanding of the functions and mechanisms of miR-34s in cancer will greatly contribute to the development of effective miR-34-based cancer therapeutics.

2.
Cell Rep ; 37(8): 110036, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34818550

RESUMO

Balance between the hematopoietic stem cell (HSC) duality to either possess self-renewal capacity or differentiate into multipotency progenitors (MPPs) is crucial for maintaining homeostasis of the hematopoietic stem/progenitor cell (HSPC) compartment. To retain the HSC self-renewal activity, KIT, a receptor tyrosine kinase, in HSCs is activated by its cognate ligand KITLG originating from niche cells. Here, we show that AT-rich interaction domain 4B (ARID4B) interferes with KITLG/KIT signaling, consequently allowing HSC differentiation. Conditional Arid4b knockout in mouse hematopoietic cells blocks fetal HSC differentiation, preventing hematopoiesis. Mechanistically, ARID4B-deficient HSCs self-express KITLG and overexpress KIT. As to downstream pathways of KITLG/KIT signaling, inhibition of Src family kinases rescues the HSC differentiation defect elicited by ARID4B loss. In summary, the intrinsic ARID4B-KITLG/KIT-Src axis is an HSPC regulatory program that enables the differentiation state, while KIT stimulation by KITLG from niche cells preserves the HSPC undifferentiated pool.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Animais , Comunicação Autócrina , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Autorrenovação Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Feminino , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-kit/genética , Transdução de Sinais/fisiologia , Fator de Células-Tronco/metabolismo , Nicho de Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo , Quinases da Família src/metabolismo
3.
Cancer Res ; 80(9): 1819-1832, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32127355

RESUMO

RING-finger E3 ligases are instrumental in the regulation of inflammatory cascades, apoptosis, and cancer. However, their roles are relatively unknown in TGFß/SMAD signaling. SMAD3 and its adaptors, such as ß2SP, are important mediators of TGFß signaling and regulate gene expression to suppress stem cell-like phenotypes in diverse cancers, including hepatocellular carcinoma (HCC). Here, PJA1, an E3 ligase, promoted ubiquitination and degradation of phosphorylated SMAD3 and impaired a SMAD3/ß2SP-dependent tumor-suppressing pathway in multiple HCC cell lines. In mice deficient for SMAD3 (Smad3 +/-), PJA1 overexpression promoted the transformation of liver stem cells. Analysis of genes regulated by PJA1 knockdown and TGFß1 signaling revealed 1,584 co-upregulated genes and 1,280 co-downregulated genes, including many implicated in cancer. The E3 ligase inhibitor RTA405 enhanced SMAD3-regulated gene expression and reduced growth of HCC cells in culture and xenografts of HCC tumors, suggesting that inhibition of PJA1 may be beneficial in treating HCC or preventing HCC development in at-risk patients.Significance: These findings provide a novel mechanism regulating the tumor suppressor function of TGFß in liver carcinogenesis.


Assuntos
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Regulação para Baixo , Deleção de Genes , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Inativação Gênica , Xenoenxertos , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/farmacologia , Fosforilação , RNA Interferente Pequeno , Proteínas Smad/metabolismo , Proteína Smad2/metabolismo , Proteína Smad3/deficiência , Proteína Smad3/genética , Espectrina/genética , Espectrina/metabolismo , Células-Tronco/patologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta1/metabolismo , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/genética , Ubiquitinação , Regulação para Cima , Sequenciamento do Exoma
4.
Nat Commun ; 10(1): 4332, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31551414

RESUMO

PTEN is frequently mutated in prostate cancer. The tumor suppressor function of PTEN is attributed to its lipid phosphatase activity that counters PI3K action. Here, we report a PTEN-ARID4B-PI3K axis in which PTEN inhibits expression of ARID4B, while ARID4B is a transcriptional activator of the PI3K subunit genes PIK3CA and PIK3R2 that are crucial for activation of the PI3K/AKT pathway. Reciprocal binding of ARID4B and histone H1 to the PIK3CA and PIK3R2 promoters modulates chromatin condensation, suggesting a mechanism by which ARID4B activates these promoters. Functional analyses reveals that ARID4B is required for prostate tumorigenesis when PTEN is deficient. The biological significance is further substantiated by the existence of a PTEN/ARID4B/PIK3CA three-gene signature that improves the predictive power for prostate cancer recurrence in patients. In summary, we identify ARID4B as a master regulator in the PTEN-PI3K pathway, thus providing a potential therapeutic target for prostate cancer carrying PTEN mutations.


Assuntos
Antígenos de Neoplasias/metabolismo , Proteínas de Neoplasias/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias da Próstata/genética , Animais , Antígenos de Neoplasias/genética , Histonas/metabolismo , Humanos , Masculino , Camundongos Knockout , Proteínas de Neoplasias/genética , PTEN Fosfo-Hidrolase/genética , Neoplasias da Próstata/metabolismo , Transdução de Sinais
5.
J Cancer ; 9(20): 3765-3775, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30405848

RESUMO

MicroRNA (miRNA)-34 family (miR-34s), including miR-34a/b/c, is the most well studied non-coding RNAs that regulate gene expression post-transcriptionally. The miR-34s mediates the tumor suppressor function of p53 in the pathogenesis of breast cancer by targeting different oncogenes. This review focuses on the anti-oncogenic regulation of the miR-34s, emphasizing the major signaling pathways that are involved in the modulation of miR-34s in breast cancer. Moreover, it highlights how epigenetic modification by the p53/miR-34s axis regulates the proliferation, invasiveness, chemoresistance, and sternness of breast cancer. A better understanding of the molecular mechanisms of miR-34s will open new opportunities for the development of novel therapeutic strategies and define a new approach in identifying potential biomarkers for early diagnosis of breast cancer.

6.
Gastroenterology ; 154(1): 195-210, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28918914

RESUMO

BACKGROUND & AIMS: Development of hepatocellular carcinoma (HCC) is associated with alterations in the transforming growth factor-beta (TGF-ß) signaling pathway, which regulates liver inflammation and can have tumor suppressor or promoter activities. Little is known about the roles of specific members of this pathway at specific of HCC development. We took an integrated approach to identify and validate the effects of changes in this pathway in HCC and identify therapeutic targets. METHODS: We performed transcriptome analyses for a total of 488 HCCs that include data from The Cancer Genome Atlas. We also screened 301 HCCs reported in the Catalogue of Somatic Mutations in Cancer and 202 from Cancer Genome Atlas for mutations in genome sequences. We expressed mutant forms of spectrin beta, non-erythrocytic 1 (SPTBN1) in HepG2, SNU398, and SNU475 cells and measured phosphorylation, nuclear translocation, and transcriptional activity of SMAD family member 3 (SMAD3). RESULTS: We found somatic mutations in at least 1 gene whose product is a member of TGF-ß signaling pathway in 38% of HCC samples. SPTBN1 was mutated in the largest proportion of samples (12 of 202, 6%). Unsupervised clustering of transcriptome data identified a group of HCCs with activation of the TGF-ß signaling pathway (increased transcription of genes in the pathway) and a group of HCCs with inactivation of TGF-ß signaling (reduced expression of genes in this pathway). Patients with tumors with inactivation of TGF-ß signaling had shorter survival times than patients with tumors with activation of TGF-ß signaling (P = .0129). Patterns of TGF-ß signaling correlated with activation of the DNA damage response and sirtuin signaling pathways. HepG2, SNU398, and SNU475 cells that expressed the D1089Y mutant or with knockdown of SPTBN1 had increased sensitivity to DNA crosslinking agents and reduced survival compared with cells that expressed normal SPTBN1 (controls). CONCLUSIONS: In genome and transcriptome analyses of HCC samples, we found mutations in genes in the TGF-ß signaling pathway in almost 40% of samples. These correlated with changes in expression of genes in the pathways; up-regulation of genes in this pathway would contribute to inflammation and fibrosis, whereas down-regulation would indicate loss of TGF-ß tumor suppressor activity. Our findings indicate that therapeutic agents for HCCs can be effective, based on genetic features of the TGF-ß pathway; agents that block TGF-ß should be used only in patients with specific types of HCCs.


Assuntos
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Mutação/genética , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Idoso , Carcinoma Hepatocelular/mortalidade , Estudos de Casos e Controles , Análise por Conglomerados , Feminino , Humanos , Neoplasias Hepáticas/mortalidade , Masculino , Pessoa de Meia-Idade
7.
Stem Cells ; 35(6): 1554-1565, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28207192

RESUMO

During neonatal testis development, centrally located gonocytes migrate to basement membrane of the seminiferous cords, where physical contact with a niche established by Sertoli cells is essential for transition of gonocytes into spermatogonial stem cells (SSCs). To provide structural support and signaling stimuli for the gonocyte-to-SSC transition that occurs at a specific location during a finite phase, temporal-spatial establishment of the niche is critical. To date, the factors that guide Sertoli cells to establish the initial stem cell niche remain largely unknown. Using the Sertoli cell-specific Arid4b knockout (Arid4bSCKO) mice, we demonstrated that ablation of AT-rich interaction domain 4B (ARID4B) resulted in abnormal detachment of Sertoli cells from the basement membrane of seminiferous cords during the gonocyte-to-SSC transition phase, suggesting failure to establish a niche for the SSC formation. Without support by a niche environment, gonocytes showed disarranged cell distribution in the Arid4bSCKO testes and underwent apoptosis. The commitment of gonocytes to differentiate into the spermatogonial lineage was broken and the capability of SSCs to self-renew and differentiate was also impaired. Gene expression profiling revealed the molecular mechanisms responsible for the phenotypic changes in the Arid4bSCKO testes, by identifying genes important for stem cell niche function as downstream effectors of ARID4B, including genes that encode gap junction protein alpha-1, KIT ligand, anti-Müllerian hormone, Glial cell-line derived neurotrophic factor, inhibin alpha, inhibin beta, and cytochrome P450 family 26 subfamily b polypeptide 1. Our results identified ARID4B as a master regulator of a signaling network that governs the establishment of a niche during the critical gonocyte-to-SSC transition phase to control the fate of gonocytes and SSCs. Stem Cells 2017;35:1554-1565.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Redes Reguladoras de Genes , Espermatogônias/citologia , Nicho de Células-Tronco , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Diferenciação Celular , Linhagem da Célula , Autorrenovação Celular , Proteínas de Ligação a DNA/genética , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade de Órgãos , Testículo/embriologia , Testículo/metabolismo , Fatores de Tempo
8.
Hepatol Commun ; 1(6): 477-493, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-29404474

RESUMO

Cancer stem cells have established mechanisms that contribute to tumor heterogeneity as well as resistance to therapy. Over 40% of hepatocellular carcinomas (HCCs) are considered to be clonal and arise from a stem-like/cancer stem cell. Moreover, HCC is the second leading cause of cancer death worldwide, and an improved understanding of cancer stem cells and targeting these in this cancer are urgently needed. Multiple studies have revealed etiological patterns and multiple genes/pathways signifying initiation and progression of HCC; however, unlike the transforming growth factor ß (TGF-ß) pathway, loss of p53 and/or activation of ß-catenin do not spontaneously drive HCC in animal models. Despite many advances in cancer genetics that include identifying the dominant role of TGF-ß signaling in gastrointestinal cancers, we have not reached an integrated view of genetic mutations, copy number changes, driver pathways, and animal models that support effective targeted therapies for these common and lethal cancers. Moreover, pathways involved in stem cell transformation into gastrointestinal cancers remain largely undefined. Identifying the key mechanisms and developing models that reflect the human disease can lead to effective new treatment strategies. In this review, we dissect the evidence obtained from mouse and human liver regeneration, and mouse genetics, to provide insight into the role of TGF-ß in regulating the cancer stem cell niche. (Hepatology Communications 2017;1:477-493).

9.
Methods Mol Biol ; 1436: 3-13, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27246204

RESUMO

Histones acetylation and deacetylation constitute part of the so-called "histone code" and work in concert with other posttranslational modifications to determine the activity of genes. Deacetylation of histone is carried out by a class of enzymes, known as histone deacetylases (HDACs). The action of HDAC is countered by histone acetyltransferases. Although histone is the best characterized substrate of HDACs, increasing evidence also indicates that non-histone proteins are equally important subtract of HDACs. Since HDACs play an important role in normal physiological and pathophysiological conditions, a sensitive and flexible deacetylation assay that can reliably detect HDAC activity and identify potential novel targets of HDACs is critical.


Assuntos
Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Immunoblotting/métodos , Acetilação , Células HEK293 , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos
10.
Mol Endocrinol ; 29(9): 1334-46, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26258622

RESUMO

Defects in spermatogenesis, a process that produces spermatozoa inside seminiferous tubules of the testis, result in male infertility. Spermatogenic progression is highly dependent on a microenvironment provided by Sertoli cells, the only somatic cells and epithelium of seminiferous tubules. However, genes that regulate such an important activity of Sertoli cells are poorly understood. Here, we found that AT-rich interactive domain 4B (ARID4B), is essential for the function of Sertoli cells to regulate spermatogenesis. Specifically, we generated Sertoli cell-specific Arid4b knockout (Arid4bSCKO) mice, and showed that the Arid4bSCKO male mice were completely infertile with impaired testis development and significantly reduced testis size. Importantly, severe structural defects accompanied by loss of germ cells and Sertoli cell-only phenotype were found in many seminiferous tubules of the Arid4bSCKO testes. In addition, maturation of Sertoli cells was significantly delayed in the Arid4bSCKO mice, associated with delayed onset of spermatogenesis. Spermatogenic progression was also defective, showing an arrest at the round spermatid stage in the Arid4bSCKO testes. Interestingly, we showed that ARID4B functions as a "coactivator" of androgen receptor and is required for optimal transcriptional activation of reproductive homeobox 5, an androgen receptor target gene specifically expressed in Sertoli cells and critical for spermatogenesis. Together, our study identified ARID4B to be a key regulator of Sertoli cell function important for male germ cell development.


Assuntos
Proteínas de Ligação a DNA/genética , Túbulos Seminíferos/embriologia , Células de Sertoli/metabolismo , Espermatogênese/fisiologia , Espermatozoides/crescimento & desenvolvimento , Animais , Apoptose/genética , Linhagem Celular , Claudina-3/biossíntese , Regulação para Baixo/genética , Epididimo/crescimento & desenvolvimento , Proteínas de Homeodomínio/biossíntese , Infertilidade Masculina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Androgênicos/metabolismo , Túbulos Seminíferos/citologia , Túbulos Seminíferos/fisiopatologia , Células de Sertoli/citologia , Espermatogênese/genética , Espermatozoides/citologia , Fatores de Transcrição/biossíntese
11.
Cancer Lett ; 354(2): 311-9, 2014 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-25173798

RESUMO

Therapy resistance increases mortality of cancer patients and remains a major obstacle for cancer treatment. The molecular mechanism underlying the therapy resistance in cancer remains not fully understood, and there is an urgent need to identify the cause of therapy resistance. MiR-34a is an important tumor suppressor whose expression is suppressed in cancer stem cells (CSCs), and re-expression of miR-34a is able to inhibit the tumorigenic activity of CSCs. Because of its tumor suppressor function, elucidating the mechanism by which miR-34a regulates therapy resistance is clearly important but remains a challenge. Our current study addresses this challenge. We identified HDAC1 and HDAC7 as novel targets of miR-34a in breast cancer, and further uncovered that deacetylation of HSP70 K246 by HDAC1 and HDAC7 promotes cancer cell survival and therapy resistance by inhibiting autophagic cell death. Our study is significant as it not only identifies the miR-34a-HDAC1/HDAC7-HSP70 K246 axis as a novel molecular signature predictive of therapy resistance, but also a viable target for potential new anti-cancer therapies to reduce such resistance in breast cancer.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Histona Desacetilase 1/metabolismo , Histona Desacetilases/metabolismo , MicroRNAs/metabolismo , Acetilação , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Proteínas de Choque Térmico HSP70/metabolismo , Células HeLa , Histona Desacetilase 1/antagonistas & inibidores , Histona Desacetilase 1/genética , Inibidores de Histona Desacetilases/metabolismo , Histona Desacetilases/genética , Humanos , Células MCF-7 , MicroRNAs/genética , Terapia de Alvo Molecular
12.
J Cell Physiol ; 229(10): 1529-37, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24585635

RESUMO

Despite a regain of interest recently in ERK3 kinase signaling, the molecular regulations of both ERK3 gene expression and protein kinase activity are still largely unknown. While it is shown that disruption of ERK3 gene causes neonatal lethality, cell type-specific functions of ERK3 signaling remain to be explored. In this study, we report that ERK3 gene expression is upregulated by cytokines through c-Jun in endothelial cells; c-Jun binds to the ERK3 gene and regulates its transcription. We further reveal a new role for ERK3 in regulating endothelial cell migration, proliferation and tube formation by upregulating SRC-3/SP-1-mediated VEGFR2 expression. The underlying molecular mechanism involves ERK3-stimulated formation of a transcriptional complex involving coactivator SRC-3, transcription factor SP-1 and the secondary coactivator CBP. Taken together, our study identified a molecular regulatory mechanism of ERK3 gene expression and revealed a previously unknown role of ERK3 in regulating endothelial cell functions.


Assuntos
Células Endoteliais da Veia Umbilical Humana/enzimologia , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Neovascularização Fisiológica , Coativador 3 de Receptor Nuclear/metabolismo , Fator de Transcrição Sp1/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Sítios de Ligação , Proteína de Ligação a CREB/metabolismo , Movimento Celular , Proliferação de Células , Regulação Enzimológica da Expressão Gênica , Células HeLa , Humanos , Proteína Quinase 6 Ativada por Mitógeno/genética , Coativador 3 de Receptor Nuclear/genética , Regiões Promotoras Genéticas , Interferência de RNA , Transdução de Sinais , Fator de Transcrição Sp1/genética , Fatores de Tempo , Transcrição Gênica , Transfecção , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
13.
Proc Natl Acad Sci U S A ; 110(12): 4616-21, 2013 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-23487765

RESUMO

ARID4A and ARID4B are homologous members of the ARID (AT-rich interaction domain) gene family. ARID4A and ARID4B physically interact with each other. ARID4A is a retinoblastoma (RB)-binding protein. Biological function of these interactions is still unknown. Here, we report that mice with complete deficiency of Arid4a, combined with haploinsufficiency of Arid4b (Arid4a(-/-)Arid4b(+/-)), showed progressive loss of male fertility, accompanied by hypogonadism and seminal vesicle agenesis/hypodysplasia. Arid4a and Arid4b are expressed mainly in Sertoli cells of testes, which implies that their roles in Sertoli cell function are to support spermatogenesis and create the impermeable blood-testis barrier. In fact, evaluation of germ cell development in the Arid4a(-/-)Arid4b(+/-) mice showed spermatogenic arrest at the stages of meiotic spermatocytes and postmeiotic haploid spermatids. Analysis of the integrity of the blood-testis barrier showed increased permeability of seminiferous tubules in the Arid4a(-/-)Arid4b(+/-) testes. Interestingly, phenotypic Sertoli cell dysfunction in the Arid4a(-/-)Arid4b(+/-) mice, including spermatogenic failures and the impaired blood-testis barrier, recapitulated the defects found in the Sertoli cell-specific androgen receptor (AR) knockout mice and the Sertoli cell-specific RB knockout mice. Investigation of the molecular mechanism identified several AR- and RB-responsive genes as downstream targets of ARID4A and ARID4B. Our results thus indicate that ARID4A and ARID4B function as transcriptional coactivators for AR and RB and play an integral part in the AR and RB regulatory pathways involved in the regulation of Sertoli cell function and male fertility.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fertilidade/fisiologia , Receptores Androgênicos/metabolismo , Proteína do Retinoblastoma/metabolismo , Proteína 1 de Ligação ao Retinoblastoma/metabolismo , Células de Sertoli/metabolismo , Espermatogênese/fisiologia , Animais , Barreira Hematotesticular/fisiologia , Proteínas de Ligação a DNA/genética , Feminino , Hipogonadismo/genética , Hipogonadismo/metabolismo , Hipogonadismo/patologia , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Masculino , Meiose/fisiologia , Camundongos , Camundongos Knockout , Receptores Androgênicos/genética , Proteína do Retinoblastoma/genética , Proteína 1 de Ligação ao Retinoblastoma/genética , Espermátides/metabolismo
14.
Genes Dev ; 27(3): 274-87, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23388826

RESUMO

Steroid receptor coactivator 3 (SRC-3) is an oncogenic nuclear receptor coactivator that plays a significant role in drug resistance. Using a lentiviral cDNA library rescue screening approach, we identified a SRC-3 downstream gene-TRAF4 (tumor necrosis factor [TNF] receptor associated-factor 4)-that functions in cell resistance to cytotoxic stress. TRAF4 expression is positively correlated with SRC-3 expression in human breast cancers. Similar to that observed for SRC-3 overexpression, breast cancer cells overexpressing TRAF4 are more resistant to stress-induced death. Here, we further dissected the underlying molecular mechanism for SRC-3 and TRAF4-mediated resistance to cytotoxic agents. We observed that SRC-3 expression is inversely correlated with the expression of p53-regulated proapoptotic genes in breast cancers and further found that SRC-3 and TRAF4 overexpression diminished cytotoxic stress-induced up-regulation of the tumor suppressor p53 protein. To determine the mechanism, we showed that the TRAF domain of TRAF4 bound to the N-terminal TRAF-like region of the deubiquitinase HAUSP (herpesvirus-associated ubiquitin-specific protease; also named USP7) and blocked the access of p53 to the same region of HAUSP. This TRAF4-mediated inhibition of HAUSP then led to the loss of p53 deubiquitination and its stabilization in response to cellular stress. Consistent with this cellular function, we also found that TRAF4 overexpression in breast cancer patients was associated significantly with poor prognosis. Because of SRC-3's ability to abrogate p53 function, our results suggest that SRC-3 overexpression may be especially important in tumors in which p53 is not mutated.


Assuntos
Coativador 3 de Receptor Nuclear/metabolismo , Estresse Fisiológico/fisiologia , Fator 4 Associado a Receptor de TNF/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Antineoplásicos/farmacologia , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Endopeptidases/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Camundongos , Coativador 3 de Receptor Nuclear/genética , Ligação Proteica , Estabilidade Proteica , Peptidase 7 Específica de Ubiquitina , Proteases Específicas de Ubiquitina , Ubiquitinação
15.
Hepatology ; 56(3): 1015-24, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22473901

RESUMO

UNLABELLED: Chronic infection of hepatitis B virus (HBV) is closely associated with the development of human hepatocellular carcinoma (HCC). HBV X protein (HBx) plays a key role in the progression of HCC. We recently found that amplified in breast cancer 1 (AIB1) protein is overexpressed in 68% of human HCC specimens and promotes HCC progression by enhancing cell proliferation and invasiveness. Given that both HBx and AIB1 play important oncogenic roles in HCC, we aimed to determine whether they could cooperatively promote human HCC development. Herein, we show that HBx-positive HCC tissues had a higher level of AIB1 protein, compared to HBx-negative HCC tissues. A positive correlation between HBx protein level and AIB1 protein level was established in HCC specimens. Without affecting its messenger RNA level, HBx induced a significant increase of the protein level of AIB1, which correlated with a significant extension of the half-life of AIB1 protein. Mechanistically, HBx could interact with AIB1 to prevent the interaction between envelope protein 3 ubiquitin ligase F-box and WD repeat domain containing 7 (Fbw7)α and AIB1, then inhibited the Fbw7α-mediated ubiquitination and degradation of AIB1. In addition, reporter assays and chromatin immunoprecipitation assays revealed that both HBx and AIB1 were recruited to matrix metalloproteinase-9 (MMP-9) promoter to enhance MMP-9 promoter activity cooperatively. Consistently, HBx and AIB1 cooperatively enhanced MMP-9 expression in HepG2 cells, which, in turn, increased cell-invasive ability. CONCLUSION: Our study demonstrates that HBx can stabilize AIB1 protein and cooperate with it to promote human HCC cell invasiveness, highlighting the essential role of the cross-talk between HBx and AIB1 in HBV-related HCC progression.


Assuntos
Proteína BRCA1/fisiologia , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Transativadores/fisiologia , Humanos , Invasividade Neoplásica , Proteínas Virais Reguladoras e Acessórias
16.
PLoS One ; 7(4): e34348, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22496793

RESUMO

Genomic imprinting is a phenomenon that some genes are expressed differentially according to the parent of origin. Prader-Willi syndrome (PWS) and Angelman syndrome (AS) are neurobehavioral disorders caused by deficiency of imprinted gene expression from paternal and maternal chromosome 15q11-q13, respectively. Imprinted genes at the PWS/AS domain are regulated through a bipartite imprinting center, the PWS-IC and AS-IC. The PWS-IC activates paternal-specific gene expression and is responsible for the paternal imprint, whereas the AS-IC functions in the maternal imprint by allele-specific repression of the PWS-IC to prevent the paternal imprinting program. Although mouse chromosome 7C has a conserved PWS/AS imprinted domain, the mouse equivalent of the human AS-IC element has not yet been identified. Here, we suggest another dimension that the PWS-IC also functions in maternal imprinting by negatively regulating the paternally expressed imprinted genes in mice, in contrast to its known function as a positive regulator for paternal-specific gene expression. Using a mouse model carrying a 4.8-kb deletion at the PWS-IC, we demonstrated that maternal transmission of the PWS-IC deletion resulted in a maternal imprinting defect with activation of the paternally expressed imprinted genes and decreased expression of the maternally expressed imprinted gene on the maternal chromosome, accompanied by alteration of the maternal epigenotype toward a paternal state spread over the PWS/AS domain. The functional significance of this acquired paternal pattern of gene expression was demonstrated by the ability to complement PWS phenotypes by maternal inheritance of the PWS-IC deletion, which is in stark contrast to paternal inheritance of the PWS-IC deletion that resulted in the PWS phenotypes. Importantly, low levels of expression of the paternally expressed imprinted genes are sufficient to rescue postnatal lethality and growth retardation in two PWS mouse models. These findings open the opportunity for a novel approach to the treatment of PWS.


Assuntos
Modelos Animais de Doenças , Impressão Genômica/genética , Síndrome de Prader-Willi/genética , Deleção de Sequência , Ubiquitina-Proteína Ligases/genética , Proteínas Centrais de snRNP/genética , Animais , Western Blotting , Imunoprecipitação da Cromatina , Mapeamento Cromossômico , Cromossomos de Mamíferos/genética , Feminino , Transtornos do Crescimento/etiologia , Humanos , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Fenótipo , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real
17.
Cell Res ; 22(6): 1003-21, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22430150

RESUMO

SRC-3/AIB1 (steroid receptor coactivator 3/amplified in breast cancer 1) is an authentic oncogene that contributes to the development of drug resistance and poor disease-free survival in cancer patients. Autophagy is also an important cell death mechanism that has tumor suppressor function. In this study, we identified macrophage migration inhibitory factor (MIF) as a novel target gene of SRC-3 and demonstrated its importance in cell survival. Specifically, we showed that MIF is a strong suppressor of autophagic cell death. We further showed that suppression of MIF, in turn, induced autophagic cell death, enhanced chemosensitivity and inhibited tumorigenesis in a xenograft mouse tumorigenesis model. Our study demonstrated that regulation of MIF expression and suppression of autophagic cell death is a potent mechanism by which SRC-3 contributes to increased chemoresistance and tumorigenicity.


Assuntos
Autofagia , Fatores Inibidores da Migração de Macrófagos/metabolismo , Coativador 3 de Receptor Nuclear/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Hipóxia Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Células HEK293 , Células HeLa , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Quinase I-kappa B/metabolismo , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Fatores Inibidores da Migração de Macrófagos/genética , Camundongos , Coativador 3 de Receptor Nuclear/antagonistas & inibidores , Coativador 3 de Receptor Nuclear/genética , Fragmentos de Peptídeos/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/uso terapêutico , Sialoglicoproteínas/metabolismo , Transplante Heterólogo
18.
PLoS One ; 6(2): e17116, 2011 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-21359227

RESUMO

Autism, a pervasive neurodevelopmental disorder manifested by deficits in social behavior and interpersonal communication, and by stereotyped, repetitive behaviors, is inexplicably biased towards males by a ratio of ∼4∶1, with no clear understanding of whether or how the sex hormones may play a role in autism susceptibility. Here, we show that male and female hormones differentially regulate the expression of a novel autism candidate gene, retinoic acid-related orphan receptor-alpha (RORA) in a neuronal cell line, SH-SY5Y. In addition, we demonstrate that RORA transcriptionally regulates aromatase, an enzyme that converts testosterone to estrogen. We further show that aromatase protein is significantly reduced in the frontal cortex of autistic subjects relative to sex- and age-matched controls, and is strongly correlated with RORA protein levels in the brain. These results indicate that RORA has the potential to be under both negative and positive feedback regulation by male and female hormones, respectively, through one of its transcriptional targets, aromatase, and further suggest a mechanism for introducing sex bias in autism.


Assuntos
Androgênios/farmacologia , Transtorno Autístico/etiologia , Estrogênios/farmacologia , Hormônios Esteroides Gonadais/fisiologia , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Aromatase/genética , Aromatase/metabolismo , Transtorno Autístico/genética , Estudos de Casos e Controles , Criança , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Masculino , Modelos Biológicos , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Caracteres Sexuais , Células Tumorais Cultivadas
19.
Proc Natl Acad Sci U S A ; 107(24): 11122-7, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20534466

RESUMO

Here we demonstrate that reprogramming steroid receptor coactivator-3 (SRC-3) function by changing its posttranslational modification (PTM) code drastically influences systems biology. These findings support the physiological importance of PTMs in directing in vivo functions of a master coregulator. We previously reported that the transactivation potential of SRC-3 is controlled in part by PTMs, although this data emanated from in vitro studies. To test the physiological implications of PTMs on SRC-3, we developed a knock-in mouse model containing mutations at four conserved phosphorylation sites. These mice displayed a systems biology phenotype with increased body weight and adiposity, coupled with reduced peripheral insulin sensitivity. Collectively, these phenotypes result from increased IGF1 signaling, due to elevated IGFBP3 levels. We provide convincing evidence that these mutations in SRC-3 promoted enhanced transcription of the IGFBP3 gene and globally influenced growth and metabolism. Consequently, these mice displayed increased liver tumorigenesis, which likely results from elevated IGF1 signaling.


Assuntos
Coativador 3 de Receptor Nuclear/genética , Coativador 3 de Receptor Nuclear/metabolismo , Adiposidade/genética , Adiposidade/fisiologia , Substituição de Aminoácidos , Animais , Sítios de Ligação/genética , Composição Corporal , Peso Corporal , Técnicas de Introdução de Genes , Humanos , Resistência à Insulina , Fator de Crescimento Insulin-Like I/metabolismo , Neoplasias Hepáticas Experimentais/etiologia , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/metabolismo , Masculino , Mamíferos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Fenótipo , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Biologia de Sistemas
20.
Nat Rev Cancer ; 9(9): 615-30, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19701241

RESUMO

The three homologous members of the p160 SRC family (SRC1, SRC2 and SRC3) mediate the transcriptional functions of nuclear receptors and other transcription factors, and are the most studied of all the transcriptional co-activators. Recent work has indicated that the SRCgenes are subject to amplification and overexpression in various human cancers. Some of the molecular mechanisms responsible for SRC overexpression, along with the mechanisms by which SRCs promote breast and prostate cancer cell proliferation and survival, have been identified, as have the specific contributions of individual SRC family members to spontaneous breast and prostate carcinogenesis in genetically manipulated mouse models. These studies have identified new challenges for cancer research and therapy.


Assuntos
Histona Acetiltransferases/fisiologia , Neoplasias/metabolismo , Coativador 2 de Receptor Nuclear/fisiologia , Transativadores/fisiologia , Fatores de Transcrição/fisiologia , Animais , Humanos , Coativador 1 de Receptor Nuclear , Coativador 3 de Receptor Nuclear , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...