Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Clin Chim Acta ; 551: 117567, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37774897

RESUMO

BACKGROUND AND AIMS: While type 2 diabetes is a well-known risk factor for pancreatic ductal adenocarcinoma (PDAC), PDAC-induced new-onset diabetes (PDAC-NOD) is a manifestation of underlying PDAC. In this study, we sought to identify potential blood-based biomarkers for distinguishing PDAC-NOD from type 2 diabetes (T2DM) without PDAC. MATERIALS AND METHODS: By ELISA analysis, a migration signature biomarker panel comprising tissue factor pathway inhibitor (TFPI), tenascin C (TNC-FNIII-C) and CA 19-9 was analyzed in plasma samples from 50 PDAC-NOD and 50 T2DM controls. RESULTS: Both TFPI (area under the curve (AUC) 0.71) and TNC-FNIII-C (AUC 0.69) outperformed CA 19-9 (AUC 0.60) in distinguishing all stages of PDAC-NOD from T2DM controls. The combined panel showed an AUC of 0.82 (95% CI = 0.73-0.90) (p = 0.002). In the PDAC-NOD early stage II samples, the three biomarkers had an AUC of 0.84 (95% CI = 0.73-0.93) vs CA 19-9, AUC = 0.60, (95% CI = 0.45-0.73), which also improved significance (p = 0.0123). CONCLUSION: The migration signature panel adds significantly to CA 19-9 to discriminate PDAC-NOD from T2DM controls and warrants further validation for high-risk group stratification.


Assuntos
Carcinoma Ductal Pancreático , Diabetes Mellitus Tipo 2 , Neoplasias Pancreáticas , Humanos , Diabetes Mellitus Tipo 2/diagnóstico , Biomarcadores Tumorais , Carcinoma Ductal Pancreático/diagnóstico , Antígeno CA-19-9
2.
Cancer Cell ; 5(6): 575-85, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15193260

RESUMO

We identified dynein light chain 1 (DLC1) as a physiologic substrate of p21-activated kinase 1 (Pak1). Pak1-DLC1 interaction plays an essential role in cell survival, which depends on Pak1's phosphorylation of DLC1 on Ser88. Pak1 associates with the complex of DLC1 and BimL, a proapoptotic BH3-only protein, and phosphorylates both proteins. Phosphorylation of BimL by Pak1 prevents it from interacting with and inactivation of Bcl-2, an antiapoptotic protein. Overexpression of DLC1 but not DLC1-Ser88Ala mutant promotes cancerous properties of breast cancer cells. DLC1 protein level is elevated in more than 90% of human breast tumors. The regulation of cell survival functions by Pak1-DLC1 interaction represents a novel mechanism by which a signaling kinase might regulate the cancerous phenotypes.


Assuntos
Apoptose , Proteínas de Transporte/farmacologia , Proteínas de Drosophila , Neoplasias/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Alanina/química , Western Blotting , Neoplasias da Mama/patologia , Ciclo Celular , Divisão Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Transformação Celular Neoplásica , Dineínas , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Glutationa Transferase/metabolismo , Humanos , Imuno-Histoquímica , Microscopia de Fluorescência , Mutação , Fenótipo , Fosforilação , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Serina/química , Transdução de Sinais , Fatores de Tempo , Técnicas do Sistema de Duplo-Híbrido , Regulação para Cima , Quinases Ativadas por p21
3.
Sci Rep ; 12(1): 19504, 2022 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-36376460

RESUMO

Triple negative breast cancer (TNBC) is a disease of poor prognosis, with the majority classified as the basal-like subtype associated with epithelial-mesenchymal transition and metastasis. Because basal breast cancers originate from proliferative luminal progenitor-like cells upon dysregulation of proper luminal differentiation, genes regulating luminal-basal transition are critical to elucidate novel therapeutic targets to improve TNBC outcomes. Herein we demonstrate that the tumor suppressor DEAR1/TRIM62 is a critical regulator of luminal cell fate. DEAR1 loss in human mammary epithelial cells results in significantly enhanced mammosphere formation that is accelerated in the presence of TGF-ß/SMAD3 signaling. Mammospheres formed following DEAR1 loss are enriched for ALDH1A1 and CK5 expression, EpCAM-/CD49f+ and CD44high/24low basal-like epithelial cells, indicating that DEAR1 regulates stem/progenitor cell properties and luminal-basal progenitor transition. We show that DEAR1 maintains luminal differentiation as a novel ubiquitin ligase for SNAI2/SLUG, a master regulator driving stemness and generation of basal-like progenitor populations. We also identify a significant inverse correlation between DEAR1 and SNAI2 expression in a 103 TNBC case cohort and show that low DEAR1 expression significantly correlates with young age of onset and shorter time to metastasis, suggesting DEAR1 could serve as a biomarker to stratify early onset TNBCs for targeted stem cell therapies.


Assuntos
Neoplasias da Mama , Neoplasias de Mama Triplo Negativas , Humanos , Feminino , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias da Mama/patologia , Mama/patologia , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal , Diferenciação Celular , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica
4.
PLoS Med ; 6(5): e1000068, 2009 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-19536326

RESUMO

BACKGROUND: Breast cancer in young women tends to have a natural history of aggressive disease for which rates of recurrence are higher than in breast cancers detected later in life. Little is known about the genetic pathways that underlie early-onset breast cancer. Here we report the discovery of DEAR1 (ductal epithelium-associated RING Chromosome 1), a novel gene encoding a member of the TRIM (tripartite motif) subfamily of RING finger proteins, and provide evidence for its role as a dominant regulator of acinar morphogenesis in the mammary gland and as an independent predictor of local recurrence-free survival in early-onset breast cancer. METHODS AND FINDINGS: Suppression subtractive hybridization identified DEAR1 as a novel gene mapping to a region of high-frequency loss of heterozygosity (LOH) in a number of histologically diverse human cancers within Chromosome 1p35.1. In the breast epithelium, DEAR1 expression is limited to the ductal and glandular epithelium and is down-regulated in transition to ductal carcinoma in situ (DCIS), an early histologic stage in breast tumorigenesis. DEAR1 missense mutations and homozygous deletion (HD) were discovered in breast cancer cell lines and tumor samples. Introduction of the DEAR1 wild type and not the missense mutant alleles to complement a mutation in a breast cancer cell line, derived from a 36-year-old female with invasive breast cancer, initiated acinar morphogenesis in three-dimensional (3D) basement membrane culture and restored tissue architecture reminiscent of normal acinar structures in the mammary gland in vivo. Stable knockdown of DEAR1 in immortalized human mammary epithelial cells (HMECs) recapitulated the growth in 3D culture of breast cancer cell lines containing mutated DEAR1, in that shDEAR1 clones demonstrated disruption of tissue architecture, loss of apical basal polarity, diffuse apoptosis, and failure of lumen formation. Furthermore, immunohistochemical staining of a tissue microarray from a cohort of 123 young female breast cancer patients with a 20-year follow-up indicated that in early-onset breast cancer, DEAR1 expression serves as an independent predictor of local recurrence-free survival and correlates significantly with strong family history of breast cancer and the triple-negative phenotype (ER(-), PR(-), HER-2(-)) of breast cancers with poor prognosis. CONCLUSIONS: Our data provide compelling evidence for the genetic alteration and loss of expression of DEAR1 in breast cancer, for the functional role of DEAR1 in the dominant regulation of acinar morphogenesis in 3D culture, and for the potential utility of an immunohistochemical assay for DEAR1 expression as an independent prognostic marker for stratification of early-onset disease.


Assuntos
Biomarcadores Tumorais/biossíntese , Neoplasias da Mama/patologia , Mama/crescimento & desenvolvimento , Recidiva Local de Neoplasia/diagnóstico , Proteínas Supressoras de Tumor/biossíntese , Biomarcadores Tumorais/genética , Mama/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Linhagem Celular Tumoral , Intervalo Livre de Doença , Regulação para Baixo/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Morfogênese/genética , Mutação/genética , Recidiva Local de Neoplasia/genética , Prognóstico , Proteínas com Motivo Tripartido , Proteínas Supressoras de Tumor/genética , Ubiquitina-Proteína Ligases
5.
Cancer Res ; 67(15): 7132-8, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17671180

RESUMO

Previously, we have shown that metastasis-associated protein 1 (MTA1) overexpression in transgenic mice was accompanied by high incidence of spontaneous B-cell lymphomas including diffuse large B-cell lymphomas (DLBCL). To understand the molecular basis of lymphoma in MTA1-transgenic (MTA1-TG) mice, we wished to identify a putative MTA1 target with a causal role in B-cell lymphogenesis. Using chromatin immunoprecipitation assays, we identified paired box gene 5 (Pax5), a molecule previously implicated in B-cell lymphogenesis, as a potential downstream effector of MTA1. Lymphomas from MTA1-TG mice also showed up-regulation of Pax5. We also found that MTA1 acetylated on Lys(626) interacted with p300 histone acetyltransferase, and that acetylated MTA1 was recruited to the Pax5 promoter to stimulate Pax5 transcription. Global gene profiling identified down-regulation of a set of genes, including those downstream of Pax5 and directly implicated in the B-cell lymphogenesis. Significance of these murine studies was established by evidence showing a widespread up-regulation of both MTA1 and Pax5 in DLBCL from humans. These observations provide in vivo genetic evidence for a role of MTA1 in lymphomagenesis.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Linfoma de Células B/genética , Linfoma Difuso de Grandes Células B/genética , Fator de Transcrição PAX5/genética , Fatores de Transcrição/fisiologia , Animais , Northern Blotting , Imunoprecipitação da Cromatina , Perfilação da Expressão Gênica , Histona Desacetilase 1 , Histona Desacetilases/genética , Humanos , Linfoma de Células B/patologia , Linfoma Difuso de Grandes Células B/patologia , Camundongos , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Plasmídeos , Regiões Promotoras Genéticas , Proteínas Repressoras , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas
6.
Cancer Res ; 67(15): 7062-7, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17671172

RESUMO

Metastasis-associated protein 1 (MTA1), a component of the nuclear remodeling complex and the founding homologue of the MTA family, has been implicated in metastasis, but definitive causative evidence in an animal model system is currently lacking. Here, we show that MTA1 overexpression in transgenic mice is accompanied by a high incidence of spontaneous B cell lymphomas including diffuse large B cell lymphomas (DLBCL). Lymphocytes and lymphoma cells from MTA1-TG mice are hyperproliferative. Lymphomas were transplantable and of clonal origin and were characterized by down-regulation of p27Kip1 as well as up-regulation of Bcl2 and cyclin D1. The significance of these murine studies was established by evidence showing a widespread up-regulation of MTA1 in DLBCL from humans. These findings reveal a previously unrecognized role for the MTA1 pathway in the development of spontaneous B cell lymphomas, and offer a potential therapeutic target in B cell lymphomas. These observations suggest that MTA1-TG mice represent a new model of spontaneous DLBCL associated with high tumor incidence and could be used for therapeutic intervention studies.


Assuntos
Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica/fisiologia , Linfoma de Células B/genética , Linfoma Difuso de Grandes Células B/genética , Fatores de Transcrição/genética , Animais , Southern Blotting , Proliferação de Células , Feminino , Histona Desacetilases/genética , Humanos , Linfonodos/patologia , Linfoma de Células B/etiologia , Linfoma de Células B/patologia , Linfoma Difuso de Grandes Células B/etiologia , Linfoma Difuso de Grandes Células B/patologia , Masculino , Camundongos , Camundongos Nus , Camundongos Transgênicos , Metástase Neoplásica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores , Células Tumorais Cultivadas
7.
Mol Cell Biol ; 25(9): 3726-36, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15831477

RESUMO

p21-activated kinase 1 (Pak1) induces cytoskeleton reorganization in part by regulating microtubule dynamics through an elusive mechanism. Using a yeast two-hybrid screen, we identified tubulin cofactor B (TCoB) (a cofactor in the assembly of the alpha/beta-tubulin heterodimers) as an interacting substrate of Pak1. Pak1 directly phosphorylated TCoB in vitro and in vivo on serines 65 and 128 and colocalized with TCoB on newly polymerized microtubules and on centrosomes. TCoB interacted with the GTPase-binding domain of Pak1 and activated Pak1 in vitro and in vivo. In contrast to wild-type TCoB, an S65A, S128A double mutant and knock-down of the endogenous TCoB or Pak1 reduced microtubule polymerization, suggesting that Pak1 phosphorylation is necessary for normal TCoB function. Overexpression of TCoB dramatically increased the number of gamma-tubulin-containing microtubule-organizing centers, a phenotype reminiscent of cells overexpressing Pak1. TCoB was overexpressed and phosphorylated in breast tumors. These findings reveal a novel role for TCoB and Pak1 in regulating microtubule dynamics.


Assuntos
Neoplasias da Mama/enzimologia , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Neoplasias da Mama/química , Feminino , Biblioteca Gênica , Humanos , Glândulas Mamárias Humanas/metabolismo , Proteínas Associadas aos Microtúbulos/análise , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/química , Chaperonas Moleculares , Mutação/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Fuso Acromático/metabolismo , Fuso Acromático/fisiologia , Ativação Transcricional , Tubulina (Proteína)/análise , Tubulina (Proteína)/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Quinases Ativadas por p21
8.
Cancer Res ; 66(3): 1694-701, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16452229

RESUMO

Here, we investigated the role of P21-activated kinase 1 (Pak1) signaling in the function of estrogen receptor-alpha (ER-alpha) as assessed by serine 305 (S305) activation and transactivation activity of ER. We found that Pak1 overexpression interfered with the antiestrogenic action of tamoxifen upon the ER transactivation function in hormone-sensitive cells. In addition, tamoxifen stimulation led to up-regulation of ER target genes in breast cancer cells with increased Pak1 expression. Tamoxifen also increased Pak1-ER interaction in tamoxifen-resistant but not in tamoxifen-sensitive cells. Results from the mutational studies discovered a role of ER-S305 phosphorylation in triggering a subsequent phosphorylation of serine 118 (S118), and these effects were further potentiated by tamoxifen treatment. We found that S305 activation-linked ER transactivation function requires a functional S118, and active Pak1 signaling is required for a sustaining S118 phosphorylation of the endogenous ER. All of these events were positively influenced by tamoxifen and thus may contribute toward the loss of antiestrogenic effect of tamoxifen. These findings suggest that Pak1 signaling-dependent activation of ER-S305 leads to an enhanced S118 phosphorylation presumably due to a conformational change, and such structural modifications may participate in the development of tamoxifen resistance.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/fisiologia , Células HeLa , Humanos , Fosforilação , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/fisiologia , Serina/metabolismo , Transdução de Sinais , Tamoxifeno/antagonistas & inibidores , Tamoxifeno/farmacologia , Ativação Transcricional , Regulação para Cima , Quinases Ativadas por p21
9.
Cancer Res ; 65(17): 7724-32, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16140940

RESUMO

It is increasingly accepted that steroidal receptor co-regulators may also function in the cytoplasmic compartment. Proline-, glutamic acid-, and leucine-rich protein-1 (PELP1) is a novel coregulator that plays a role in both the genomic and extranuclear actions of estrogen receptors (ER) in hormonally responsive tissues. In this study using breast tumor arrays, we found that PELP1 was localized only in the cytoplasm in 58% of the PELP1-positive breast tumors. To help explain the significance of the cytoplasmic localization of PELP1 in human breast tumors, we created a mutant protein that was expressed only in the cytoplasm (PELP1-cyto) and then generated a model system wherein MCF-7 breast cancer cells were engineered to specifically express this mutant. We found that PELP1-cyto cells were hypersensitive to estrogen but resistant to tamoxifen. PELP1-cyto cells, but not parental MCF-7 cells, formed xenograft tumors in nude mice. In addition, PELP1-cyto cells exhibited increased association of PELP1 with Src, enhanced mitogen-activated protein kinase (MAPK) activation, and constitutive activation of AKT. The altered localization of PELP1 was sufficient to trigger the interaction of PELP1 with the p85 subunit of phosphatidylinositol-3-kinase (PI3K), leading to PI3K activation. In addition, PELP1 interacted with epidermal growth factor receptors and participated in growth factor-mediated ER transactivation functions. Our results suggest that the altered localization of PELP1 modulates sensitivity to antiestrogens, potentiates tumorigenicity, presumably via the stimulation of extranuclear estrogen responses, such as the activation of MAPK and AKT, and also enhance cross-regulation of ER transactivation activity by growth factors.


Assuntos
Neoplasias da Mama/metabolismo , Transativadores/metabolismo , Animais , Neoplasias da Mama/enzimologia , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Proteínas Correpressoras , Citoplasma/metabolismo , Ativação Enzimática , Receptores ErbB/metabolismo , Células HeLa , Humanos , Sistema de Sinalização das MAP Quinases , Neoplasias Hormônio-Dependentes/enzimologia , Neoplasias Hormônio-Dependentes/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Frações Subcelulares/metabolismo , Fatores de Transcrição , Quinases da Família src/metabolismo
10.
J Natl Cancer Inst ; 109(8)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28376184

RESUMO

Background: Blood-based biomarkers for early detection of pancreatic ductal adenocarcinoma (PDAC) are urgently needed. Current biomarkers lack high sensitivity and specificity for population screening. The gold-standard biomarker, CA 19-9, also fails to demonstrate the predictive value necessary for early detection. Methods: To validate a functional genomics-based plasma migration signature biomarker panel, plasma tissue factor pathway inhibitor (TFPI), tenascin C (TNC-FN III-C), and CA 19-9 levels were measured by enzyme-linked immunosorbent assays in three early-stage PDAC plasma cohorts, including two independent blinded validation cohorts containing a total of 43 stage I, 163 stage II, 86 chronic pancreatitis, 31 acute biliary obstruction, and 108 controls. Logistic regression models developed classification rules combining TFPI and/or TNC-FN III-C with CA 19-9 for patient cases and control subjects, with or without adjustment for age and diabetes status. Model classification performance was evaluated and analyses repeated among subpopulations without diabetes and pancreatitis history. Two-sided P values were calculated using bootstrap method. Results: The TFPI/TNC-FN III-C/CA 19-9 panel improved CA 19-9 performance in all early-stage cohorts, including discriminating stage IA/IB/IIA, stage IIB, and all early-stage cancer from healthy controls. Statistical significance was reached for a number of subcohorts, including for all early-stage cancer vs healthy controls (cohort 1 AUC = 0.92, 95% CI = 0.86 to 0.96, P = .04; cohort 3 AUC = 0.83, 95% CI = 0.76 to 0.89, P = .045). Among subcohorts without diabetes and pancreatitis history, the panel approaches potential clinical utility for early detection to discriminate early-stage PDAC from healthy controls including an area under the curve (AUC) of 0.87 (95% CI = 0.77 to 0.95) for stage I/IIA, an AUC of 0.93 (95% CI = 0.87 to 0.98) for stage IIB, and a statistically significant AUC of 0.89 (95% CI = 0.82 to 0.95) for all early-stage cancer ( P = .03). Conclusions: TFPI/TNC-FN III-C migration signature adds statistically significantly to CA 19-9's predictive power to detect early-stage PDAC and may have clinical utility for early detection of surgically resectable PDAC, as well as for enhanced survival from this routinely lethal cancer.


Assuntos
Antígeno CA-19-9/sangue , Carcinoma Ductal Pancreático/sangue , Carcinoma Ductal Pancreático/diagnóstico , Lipoproteínas/sangue , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/diagnóstico , Tenascina/sangue , Doença Aguda , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Área Sob a Curva , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/cirurgia , Estudos de Casos e Controles , Colestase/sangue , Estudos de Coortes , Diabetes Mellitus/sangue , Detecção Precoce de Câncer/métodos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/cirurgia , Pancreatite Crônica/sangue , Curva ROC , Método Simples-Cego
11.
Hum Pathol ; 37(6): 656-61, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16733204

RESUMO

Endometrial carcinoma is one of the most common malignancies of the female genital tract. Metastasis-associated protein 1 (MTA1) is a component of the Mi-2/nucleosome remodeling and deacetylating complex and acts as a potent corepressor of estrogen receptor in breast cancer cells. MTA1 expression has been demonstrated in various cancers but has never been explored in endometrial carcinoma. We investigated the expression profile of MTA1 in different stages of benign endometrium as well as in endometrial endometrioid adenocarcinoma using immunohistochemistry and Western blotting. In the proliferative and secretory phases, MTA1 was expressed in both the glandular and the stromal compartments and was localized in nucleus and cytoplasm of these cells. MTA1 expression in secretory phase was less prominent when compared with the proliferative phase. In postmenopausal sections, MTA1 staining was observed in both glandular and stromal compartments and was localized in both nucleus and cytoplasm. Western blot analysis of 6 tumor specimens showed increased expression of MTA1 in all the tumors analyzed. Immunohistochemical staining performed on tumor microarray containing 70 endometrial endometrioid adenocarcinomas of various grades showed increased expression of MTA1 in 53 (75.7%) tumors. In grade 1 and grade 2 tumors, MTA1 was present in both nucleus and cytoplasm. Interestingly, in grade 3 tumors, MTA1 was localized in the cytoplasm only. Our results suggest a potential role of MTA1 in endometrial carcinomas.


Assuntos
Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Endométrio/metabolismo , Expressão Gênica , Histona Desacetilases/metabolismo , Proteínas Repressoras/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Endométrio/citologia , Feminino , Histona Desacetilases/genética , Humanos , Imuno-Histoquímica , Estadiamento de Neoplasias , Proteínas Repressoras/genética , Transativadores
12.
Cancer Res ; 64(18): 6416-23, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15374949

RESUMO

The estrogen receptor plays an important role in breast cancer progression. Proline-, glutamic acid-, and leucine-rich protein 1 (PELP1), also called modulator of nongenomic activity of estrogen receptor (MNAR), a novel coactivator of estrogen receptor, modulates estrogen receptor transactivation functions. The mechanisms by which PELP1 modulates estrogen receptor genomic functions is not known. Here, using biochemical and scanning confocal microscopic analysis, we have demonstrated nuclear localization and functional implications of PELP1. Subnuclear fractionation showed PELP1 association with chromatin and nuclear matrix fractions. Ligand stimulation promoted recruitment of PELP1 to 17beta-estradiol responsive promoters, its colocalization with acetylated H3, and increased PELP1-associated histone acetyltransferase enzymatic activity. Far Western analysis revealed that PELP1 interacts with histone 1 and 3, with more preference toward histone 1. Using deletion analysis, we have identified the PELP1 COOH-terminal region as the histone 1 binding site. The PELP1 mutant lacking histone 1-binding domain acts as a dominant-negative and blocks estrogen receptor alpha-mediated transcription. Chromatin immunoprecipitation analysis showed a cyclic association and dissociation of PELP1 with the promoter, with recruitment of histone 1 and PELP1 occurring in opposite phases. PELP1 overexpression increased the micrococcal nuclease sensitivity of estrogen response element-containing nucleosomes. Our results provide novel insights about the transcription regulation of PELP1 and suggest that PELP1 participates in chromatin remodeling activity via displacement of histone 1 in cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Histonas/metabolismo , Transativadores/fisiologia , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Cromatina/metabolismo , Proteínas Correpressoras , Citosol/metabolismo , Epitopos/biossíntese , Epitopos/genética , Estradiol/farmacologia , Ácido Glutâmico/metabolismo , Humanos , Matriz Nuclear/metabolismo , Osteossarcoma/genética , Osteossarcoma/metabolismo , Plasmídeos/genética , Testes de Precipitina , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Transativadores/genética , Transativadores/imunologia , Transativadores/metabolismo , Fatores de Transcrição
13.
Hum Pathol ; 36(6): 670-5, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16021574

RESUMO

Salivary duct carcinoma (SDC) is a high-grade neoplasm with marked morphological resemblance to mammary duct carcinoma. The novel estrogen receptor (ER)-interacting protein and the proline-, glutamic acid-, and leucine-rich protein 1 ( PELP1 ), also called the modulator of nongenomic activity of ER ( MNAR ), have been shown to activate steroid hormone receptors in mammary carcinomas by nongenomic and genomic mechanisms. The expression and the relationship of this gene to the ER status in SDCs are unknown. We investigated the differential expression of the PELP1 / MNAR and the ERs alpha and beta proteins in SDCs, using Western blotting and immunohistochemistry. Western blot analysis of 7 paired normal and tumor specimens showed increased expression of PELP1 / MNAR and ER beta in 3 and 4 of the SDCs, respectively. No detectable expression of ER alpha in any normal or SDC specimens was noted. Immunohistochemical staining performed on 70 SDCs revealed strong expression of PELP1 / MNAR in 51 (73%) and ER beta in 52 (74%) tumors. PELP1 / MNAR and ER beta were coexpressed in 35 (50%), individually in 17 (24.2%), and negative in 18 (25.7%) tumors. PELP1 / MNAR staining was predominantly cytoplasmic whereas ER beta staining was nuclear and occasionally cytoplasmic in tumor cells. Our results indicate that PELP1 / MNAR and ER beta are coexpressed in most SDCs and may play a role in the pathobiology of these tumors.


Assuntos
Adenocarcinoma/metabolismo , Biomarcadores Tumorais/análise , Receptor beta de Estrogênio/biossíntese , Neoplasias das Glândulas Salivares/metabolismo , Transativadores/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Western Blotting , Proteínas Correpressoras , Feminino , Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Transativadores/genética , Fatores de Transcrição
14.
Clin Cancer Res ; 9(8): 3198-203, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12912973

RESUMO

Induction of terminal differentiation of cancer cells is an evolving novel therapeutic approach, and accordingly, peroxisome proliferator-activated receptor gamma (PPARgamma), a ligand-stimulated transcription factor with differentiation-promoting activity and overexpressed in a variety of cancers, has emerged as one of the promising therapeutic targets. Because c-erbB family growth factor receptor 2 (HER2) overexpression is one of the most recognizable molecular dysfunctions in breast tumors, in the studies presented here, we explored the effect of HER2 overexpression on the status of PPARgamma expression and on the sensitivity of breast cancer cells to PPARgamma-ligand troglitazone-induced growth inhibition. We show that HER2 overexpression in MCF7 breast cancer cells enhanced the expression of PPARgamma-mRNA and -protein. Furthermore, PPARgamma expression was dramatically increased in 11 of 16 breast tumors as compared with the adjacent normal tissue. In addition, HER2 up-regulation resulted in a partial inhibition of transcriptional activity of the endogenous PPARgamma, stimulation to differentiation, and resistance to troglitazone-mediated inhibition of anchorage-independent growth of breast cancer cells. Conversely, down-regulation of HER2 by anti-HER2 monoclonal antibody Herceptin led to a decreased level of PPARgamma protein and sensitization of breast cancer cells to the inhibitory effects of troglitazone. In summary, these findings show for the first time that HER2 up-regulates PPARgamma expression and modulates the sensitivity of breast cancer cells to PPARgamma ligand therapy.


Assuntos
Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Receptor ErbB-2/fisiologia , Receptores Citoplasmáticos e Nucleares/biossíntese , Fatores de Transcrição/biossíntese , Anticorpos Monoclonais/química , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados , Compostos Azo/farmacologia , Northern Blotting , Western Blotting , Diferenciação Celular , Divisão Celular , Linhagem Celular Tumoral , Cromanos/farmacologia , Corantes/farmacologia , Citoplasma/metabolismo , Regulação para Baixo , Humanos , Imuno-Histoquímica , Ligantes , Metabolismo dos Lipídeos , Luciferases/metabolismo , RNA Mensageiro/metabolismo , Tiazolidinedionas/farmacologia , Ativação Transcricional , Trastuzumab , Troglitazona , Regulação para Cima
15.
PLoS One ; 10(10): e0139049, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26431551

RESUMO

The validation of candidate biomarkers often is hampered by the lack of a reliable means of assessing and comparing performance. We present here a reference set of serum and plasma samples to facilitate the validation of biomarkers for resectable pancreatic cancer. The reference set includes a large cohort of stage I-II pancreatic cancer patients, recruited from 5 different institutions, and relevant control groups. We characterized the performance of the current best serological biomarker for pancreatic cancer, CA 19-9, using plasma samples from the reference set to provide a benchmark for future biomarker studies and to further our knowledge of CA 19-9 in early-stage pancreatic cancer and the control groups. CA 19-9 distinguished pancreatic cancers from the healthy and chronic pancreatitis groups with an average sensitivity and specificity of 70-74%, similar to previous studies using all stages of pancreatic cancer. Chronic pancreatitis patients did not show CA 19-9 elevations, but patients with benign biliary obstruction had elevations nearly as high as the cancer patients. We gained additional information about the biomarker by comparing two distinct assays. The two CA 9-9 assays agreed well in overall performance but diverged in measurements of individual samples, potentially due to subtle differences in antibody specificity as revealed by glycan array analysis. Thus, the reference set promises be a valuable resource for biomarker validation and comparison, and the CA 19-9 data presented here will be useful for benchmarking and for exploring relationships to CA 19-9.


Assuntos
Antígeno CA-19-9/sangue , Neoplasias Pancreáticas/sangue , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Pancreatite Crônica/sangue , Sensibilidade e Especificidade
16.
J Clin Endocrinol Metab ; 89(12): 6130-8, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15579769

RESUMO

Proline-, glutamic acid-, and leucine-rich protein-1)PELP1/MNAR [modulator of nongenomic activity of estrogen receptor (ER)], a novel coregulatory protein, modulates genomic as well as nongenomic activity of ERs. We characterized the expression and localization of PELP1 in both benign and cancerous endometrium. Our results suggest that PELP1 is expressed in all stages of endometrium; however, this protein exhibits distinct localization depending on the phase. PELP1 is expressed in both the stroma and epithelial cells. Using the Ishikawa endometrial cancer model cell line and ER subtype-specific ligands, we found that PELP1 functionally interacts with both ERalpha and ERbeta and enhances their transcriptional responses. However, in endometrial cancer cells, endogenous PELP1 is also required for optimal ligand-mediated transcription and proliferation responses. PELP1 promoted a tamoxifen-mediated agonistic action in endometrial, but not in breast cancer cells. PELP1 expression and localization are widely deregulated in endometrial cancers. In addition, PELP1 and ERbeta were localized predominantly in the cytoplasm of high-grade endometrial tumors. Our results suggest that PELP1 plays an essential role in the proliferation of cancerous endometrial cells.


Assuntos
Neoplasias do Endométrio/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Transativadores/metabolismo , Antineoplásicos Hormonais/farmacologia , Divisão Celular , Linhagem Celular Tumoral , Proteínas Correpressoras , Neoplasias do Endométrio/patologia , Células Epiteliais/metabolismo , Feminino , Humanos , Células Estromais/metabolismo , Tamoxifeno/farmacologia , Fatores de Transcrição
17.
Gene ; 330: 115-22, 2004 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-15087130

RESUMO

Proline-, glutamic acid- and leucine-rich protein 1 (PELP1)/modulator of nongenomic activity of estrogen receptor (MNAR), a novel coactivator of estrogen receptors (ERs; ERalpha and ERbeta), modulates the genomic and nongenomic functions of the ERs. PELP1 expression is developmentally regulated in mammary glands and overexpressed in breast tumors. However, little is known about the regulation of PELP1. In this study, we examined whether PELP1 expression is modulated by steroid hormone 17beta-estradiol (E2)-ER pathway. We found that in MCF-7 breast cancer cells, E2 upregulated PELP1 expression threefold and that this upregulation was reduced by antiestrogen. We also found that E2 modulated PELP1 levels in an actinomycin-D-sensitive manner, suggesting transcriptional regulation. Cloning and analysis of the 2-kb PELP1 promoter region revealed two estrogen-responsive element (ERE) half sites in the PELP1 promoter region. In transient transfection assays, E2 upregulated PELP1 promoter activity in breast, endometrial and osteosarcoma model cancer cell lines in an ICI 182,780-sensitive manner. We demonstrated the recruitment of ER to the PELP1 promoter in vitro using EMSA assays and in vivo using a chromatin immunoprecipitation assay. The PELP1 promoter was similarly upregulated by both ERalpha and ERbeta and differentially regulated by selective estrogen receptor modulators in a cell line-dependent manner. Our results suggest that PELP1 expression is modulated by the E2-ER pathway and that PELP1 is an ER target gene.


Assuntos
Regiões Promotoras Genéticas/genética , Transativadores/genética , Sequência de Bases , Sítios de Ligação/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Clonagem Molecular , Proteínas Correpressoras , Códon de Iniciação/genética , DNA/química , DNA/genética , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Estradiol/farmacologia , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Luciferases/genética , Luciferases/metabolismo , Dados de Sequência Molecular , Osteossarcoma/genética , Osteossarcoma/patologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Cloridrato de Raloxifeno/farmacologia , Receptores de Estrogênio/genética , Receptores de Estrogênio/fisiologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Análise de Sequência de DNA , Tamoxifeno/farmacologia , Fatores de Transcrição
18.
FEBS Lett ; 567(2-3): 243-7, 2004 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-15178330

RESUMO

Recent studies have shown that p21-activated kinase 1 (Pak1) phosphorylates estrogen receptor-alpha (ER alpha) at Ser 305 and also promotes its transactivation function. Here, we sought to investigate whether substitution of serine 305 in ER with glutamic acid (ER alpha-S305E), which mimics the phosphorylation state, would influence the status of ER-target genes. To explore this possibility, we generated clones overexpressing ER alpha-S305E in ER-negative MDA-MB-231 cells and analyzed the status of ER-regulated genes using a gene array. Results indicated that the expression of ER alpha-S305E is sufficient to upregulate the expression of a few but not all ER-regulated genes, i.e., cyclin D1 and zinc finger protein 147 (estrogen-responsive finger protein), while there was no significant change in the expression of remaining genes on the array. In addition, we found an increased expression as well as nuclear accumulation of cyclin D1 protein in MDA-MB-231 cells expressing ER alpha-S305E as compared to the level of cyclin D1 in MDA-MB-231 cells expressing WT-ER alpha or pcDNA. Furthermore, ER alpha-S305E, but not mutation of ER alpha-S305 to alanine, enhanced the cyclin D1 promoter activity. These findings suggest that ER alpha activation at S305 is sufficient to upregulate the expression of cyclin D1, an ER-regulated gene that is implicated in the progression of breast cancer. Phosphorylation of ER alpha by Pak1 or its upstream regulators could upregulate the expression of a subset of ER-target genes in a ligand-independent manner and hence, might contribute toward the development of hormone independence in breast cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Ciclina D1/metabolismo , Receptores de Estrogênio/metabolismo , Serina/metabolismo , Substituição de Aminoácidos , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Ciclina D1/genética , Receptor alfa de Estrogênio , Estrogênios/farmacologia , Feminino , Imunofluorescência , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Microscopia Confocal , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , RNA/biossíntese , Receptores de Estrogênio/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina/genética , Transdução de Sinais , Transfecção , Regulação para Cima
19.
Cancer Res ; 74(20): 5683-9, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25261235

RESUMO

Elucidation of the regulatory controls on epithelial plasticity is pivotal not only to better understand the nature of metastasis but also for the design of targeted therapies to prevent the earliest steps in migration and invasion from the primary tumor. This review will highlight the role of the novel TRIM protein DEAR1 (annotated as TRIM62) in the regulation of apical-basal polarity and acinar morphogenesis as well as its function as a chromosome 1p35 tumor suppressor and negative regulator of TGFß-driven epithelial-mesenchymal transition (EMT). DEAR1 binds to and promotes the ubiquitination of SMAD3, the major effector of TGFß-mediated EMT, as well as downregulates SMAD3 targets SNAIL1/2, master transcriptional regulators of EMT. Cumulative results suggest a novel paradigm for DEAR1 in the regulation of the breast tumor microenvironment, polarity, and EMT. Because DEAR1 undergoes loss-of-function mutations, homozygous deletion, as well as copy-number losses in multiple epithelial cancers, including breast cancer, DEAR1 has clinical use as a predictive and prognostic biomarker as well as for stratifying breast cancers and potentially other epithelial tumor types for targeted therapies aimed at the pathways regulated by DEAR1.


Assuntos
Neoplasias da Mama/metabolismo , Polaridade Celular , Neoplasias Epiteliais e Glandulares/metabolismo , Ubiquitina-Proteína Ligases/fisiologia , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal , Epitélio/patologia , Feminino , Humanos , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/patologia , Transdução de Sinais , Fator de Crescimento Transformador beta/fisiologia , Proteínas com Motivo Tripartido , Proteínas Supressoras de Tumor/fisiologia , Ubiquitinação
20.
Nat Commun ; 4: 2545, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24089055

RESUMO

The mammalian circadian clock regulates the daily cycles of many important physiological processes, but its mechanism is not well understood. Here we provide genetic and biochemical evidence that metastasis-associated protein 1 (MTA1), a widely upregulated gene product in human cancers, is an integral component of the circadian molecular machinery. Knockout of MTA1 in mice disrupts the free-running period of circadian rhythms under constant light and normal entrainment of behaviour to 12-h-light/12-h-dark cycles. The CLOCK-BMAL1 heterodimer activates MTA1 transcription through a conserved E-box element at its promoter. MTA1, in turn, interacts with and recruits CLOCK-BMAL1 at its own and CRY1 promoters and promotes their transcription. Moreover, MTA1 deacetylates BMAL1 at lysine 538 through regulating deacetylase SIRT1 expression, thus disturbing the CRY1-mediated negative feedback loop. These findings uncover a previously unappreciated role for MTA1 in maintenance of circadian rhythmicity through acting on the positive limb of the clock machinery.


Assuntos
Ritmo Circadiano/genética , Regulação da Expressão Gênica , Histona Desacetilases/genética , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Transcrição Gênica , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Acetilação , Animais , Comportamento Animal , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Criptocromos/genética , Criptocromos/metabolismo , Retroalimentação Fisiológica , Feminino , Histona Desacetilases/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/genética , Fotoperíodo , Regiões Promotoras Genéticas , Multimerização Proteica , Proteínas Repressoras/metabolismo , Transdução de Sinais , Sirtuína 1/genética , Sirtuína 1/metabolismo , Transativadores , Fatores de Transcrição/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA