Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Matrix Biol ; 121: 179-193, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37422024

RESUMO

Integrins are cellular receptors that bind the extracellular matrix (ECM) and facilitate the transduction of biochemical and biophysical microenvironment cues into cellular responses. Upon engaging the ECM, integrin heterodimers must rapidly strengthen their binding with the ECM, resulting in the assembly of force-resistant and force-sensitive integrin associated complexes (IACs). The IACs constitute an essential apparatus for downstream signaling and fibroblast phenotypes. During wound healing, integrin signaling is essential for fibroblast motility, proliferation, ECM reorganization and, ultimately, restoration of tissue homeostasis. Semaphorin 7A (SEMA7a) has been previously implicated in post-injury inflammation and tissue fibrosis, yet little is known about SEMA7a's role in directing stromal cell, particularly fibroblast, behaviors. We demonstrate that SEMA7a regulates integrin signaling through cis-coupling with active integrin α5ß1 on the plasma membrane, enabling rapid integrin adhesion strengthening to fibronectin (Fn) and normal downstream mechanotransduction. This molecular function of SEMA7a potently regulates fibroblast adhesive, cytoskeletal, and migratory phenotype with strong evidence of downstream alterations in chromatin structure resulting in global transcriptomic reprogramming such that loss of SEMA7a expression is sufficient to impair the normal migratory and ECM assembly phenotype of fibroblasts resulting in significantly delayed tissue repair in vivo.


Assuntos
Integrina alfa5beta1 , Mecanotransdução Celular , Integrina alfa5beta1/genética , Integrina alfa5beta1/metabolismo , Integrinas/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Transdução de Sinais , Fibroblastos/metabolismo , Adesão Celular , Matriz Extracelular/metabolismo
2.
Methods Mol Biol ; 2299: 237-261, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34028748

RESUMO

Aberrant deposition of the extracellular matrix (ECM) causes fibrosis and leads to ECM stiffening. This fibrotic ECM provides biological and biophysical stimulations to alter cell activity and drive progression of fibrosis. As an emerging discipline, mechanobiology aims to access the impact of both these cues on cell behavior and relates the reciprocity of mechanical and biological interactions; it incorporates concepts from different fields, like biology and physics, to help study the mechanical and biological facets of fibrosis extensively. A useful experimental platform in mechanobiology is decellularized ECM (dECM), which mimics the native microenvironment more accurately than standard 2D culture techniques as its composition includes similar ECM protein components and stiffness. dECM, therefore, generates more reliable results that better recapitulate in vivo fibrosis.


Assuntos
Técnicas de Cultura de Células/métodos , Matriz Extracelular/patologia , Miofibroblastos/patologia , Animais , Fenômenos Biomecânicos , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fibrose , Humanos , Espectrometria de Massas , Microscopia de Força Atômica , Modelos Biológicos , Miofibroblastos/metabolismo
5.
Matrix Biol ; 82: 86-104, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31004743

RESUMO

The extracellular matrix (ECM) microenvironment is increasingly implicated in the instruction of pathologically relevant cell behaviors, from aberrant transdifferentation to invasion and beyond. Indeed, pathologic ECMs possess a panoply of alterations that provide deleterious instructions to resident cells. Here we demonstrate the precise manner in which the ECM protein fibronectin (FN) undergoes the posttranslational modification citrullination in response to peptidyl-arginine deiminase (PAD), an enzyme associated with innate immune cell activity and implicated in systemic ECM-centric diseases, like cancer, fibrosis and rheumatoid arthritis. FN can be citrullinated in at least 24 locations, 5 of which reside in FN's primary cell-binding domain. Citrullination of FN alters integrin clustering and focal adhesion stability with a concomitant enhancement in force-triggered integrin signaling along the FAK-Src and ILK-Parvin pathways within fibroblasts. In vitro migration and in vivo wound healing studies demonstrate the ability of citrullinated FN to support a more migratory/invasive phenotype that enables more rapid wound closure. These findings highlight the potential of ECM, particularly FN, to "record" inflammatory insults via post-translational modification by inflammation-associated enzymes that are subsequently "read" by resident tissue fibroblasts, establishing a direct link between inflammation and tissue homeostasis and pathogenesis through the matrix.


Assuntos
Fibronectinas/metabolismo , Adesões Focais/metabolismo , Integrinas/metabolismo , Células Estromais/citologia , Animais , Sítios de Ligação , Adesão Celular , Movimento Celular , Células Cultivadas , Citrulinação , Matriz Extracelular/metabolismo , Feminino , Fibronectinas/química , Humanos , Masculino , Camundongos , Desiminases de Arginina em Proteínas/metabolismo , Transdução de Sinais , Células Estromais/metabolismo
6.
Mol Oncol ; 12(12): 2055-2071, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30171816

RESUMO

Renal cell carcinoma (RCC) has the third highest mortality rate among urological tumors, and 20-30% of RCC patients present with metastatic RCC at the time of diagnosis. Although recent studies have indicated that estrogen receptor ß (ERß) could play promoting roles in RCC progression, the detailed mechanisms remain to be clarified. In the present study, we found that expression of ERß, but not ERα, increases with tumor stage and grade, and also observed that modification of ERß signals using estrogens/anti-estrogens, shRNA knockdown of ERß and overexpression of ERß using ectopic cDNA affects RCC cell proliferation, migration and invasion. Mechanism analysis revealed that ERß can promote RCC cell invasion via an increase in transforming growth factor ß1 (TGF-ß1)/SMAD3 signals, and interrupting TGF-ß1/SMAD3 signals with a TGFßR1 inhibitor can reverse/block ERß-increased RCC cell migration. Importantly, preclinical analyses using in vivo mouse models of RCC revealed that targeting of this newly identified ERß/TGF-ß1/SMAD3 pathway with either the FDA-approved anti-estrogen ICI182,780 (Faslodex) or a selective ERß antagonist 4-[2-phenyl-5,7 bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-3-yl]phenol can significantly reduce RCC tumor growth and invasion, which may be suitable as the basis for novel therapies to more effectively suppress metastatic RCC.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Receptor beta de Estrogênio/metabolismo , Fulvestranto/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Antagonistas do Receptor de Estrogênio/uso terapêutico , Receptor beta de Estrogênio/análise , Receptor beta de Estrogênio/antagonistas & inibidores , Feminino , Humanos , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Masculino , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/análise , Taxa de Sobrevida , Fator de Crescimento Transformador beta1/análise
7.
Oncogene ; 37(37): 5037-5053, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29789714

RESUMO

Recent studies indicated that the estrogen receptor beta (ERß) could affect the progression of prostate and bladder tumors, however, its roles in the renal cell carcinoma (RCC), remain to be elucidated. Here, we provide clinical evidence that ERß expression is correlated in a negative manner with the overall survival/disease-free survival in RCC patients. Mechanism dissection revealed that targeting ERß with ERß-shRNA and stimulating the transactivation of ERß with 17ß-estradiol or environmental endocrine disrupting chemicals, all resulted in altering the lncRNA HOTAIR expression. The ERß-modulated HOTAIR is able to function via antagonizing several microRNAs, including miR-138, miR-200c, miR-204, or miR-217 to impact various oncogenes, including ADAM9, CCND2, EZH2, VEGFA, VIM, ZEB1, and ZEB2, to promote RCC proliferation and invasion. Together, the identification of the ERß-HOTAIR axis may provide us new biomarkers and/or therapeutic targets to better suppress RCC progression in the future.


Assuntos
Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Receptor beta de Estrogênio/genética , Neoplasias Renais/genética , Neoplasias Renais/patologia , MicroRNAs/genética , RNA Longo não Codificante/genética , Animais , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Progressão da Doença , Feminino , Humanos , Camundongos , Camundongos Nus , RNA Interferente Pequeno/genética , Transdução de Sinais/genética
8.
Mol Cancer ; 15: 7, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26790618

RESUMO

BACKGROUND: Cancer associated fibroblasts (CAF) play important roles in tumor growth that involves inflammation and epithelial cell differentiation. Early studies suggested that estrogen receptor alpha (ERα) was expressed in stromal cells in normal prostates and prostate cancer (PCa), but the detailed functions of stromal ERα in the PCa remain to be further elucidated. METHODS: Migration and invasion assays demonstrated the presence of high levels of ERα in CAF cells (CAF.ERα(+)) suppressed PCa invasion via influencing the infiltration of tumor associated macrophages. ERα decreased CAF CCL5 secretion via suppressing the CCL5 promoter activity was examined by luciferase assay. ERα decreased CCL5 and IL-6 expression in conditioned media that was collected from CAF cell only or CAF cell co-cultured with macrophages as measured by ELISA assay. RESULTS: Both in vitro and in vivo studies demonstrated CAF.ERα(+) led to a reduced macrophage migration toward PCa via inhibiting CAF cells secreted chemokine CCL5. This CAF.ERα(+) suppressed macrophage infiltration affected the neighboring PCa cells invasion and the reduced invasiveness of PCa cells are at least partly due to reduced IL6 expression in the macrophages and CAF. CONCLUSION: Our data suggest that CAF ERα could be applied as a prognostic marker to predict cancer progression, and targeting CCL5 and IL6 may be applied as an alternative therapeutic approach to reduce M2 type macrophages and PCa invasion in PCa patients with low or little ERα expression in CAF cells.


Assuntos
Quimiocina CCL5/metabolismo , Receptor alfa de Estrogênio/metabolismo , Fibroblastos/metabolismo , Interleucina-6/metabolismo , Macrófagos/metabolismo , Neoplasias da Próstata/patologia , Microambiente Tumoral , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Humanos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos Nus , Modelos Biológicos , Invasividade Neoplásica , Neoplasias da Próstata/metabolismo , Microambiente Tumoral/efeitos dos fármacos
9.
Oncotarget ; 7(7): 7842-55, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26556868

RESUMO

Early clinical studies suggested that infiltrating mast cells could be associated with a poor outcome in bladder cancer (BCa) patients. The mechanisms of how mast cells influence the BCa progression, however, are unclear. Using the human clinical BCa sample survey and in vitro co-culture systems, we found BCa cells could recruit more mast cells than the surrounding non-malignant urothelial cells. The consequences of this better recruitment of mast cells toward BCa cells could then enhance BCa cell invasion. Mechanism dissection revealed that the enhanced BCa cell invasion could function via up-regulation of the estrogen receptor beta (ERß) in both mast cells and BCa cells, which resulted in the increased CCL2/CCR2/EMT/MMP9 signals. Using the pre-clinical mouse BCa model, we further validated the mast cell-promoted BCa invasion. Interruption of the newly identified ERß/CCL2/CCR2/EMT/MMP9 pathway via either ERß-siRNA, ERß antagonist PHTPP, or CCR2 antagonist can effectively reverse the mast cell-enhanced BCa cells invasion. Together, our finding could lead to the development of an alternative new therapeutic approach to better treat BCa metastasis.


Assuntos
Quimiocina CCL2/metabolismo , Receptor beta de Estrogênio/metabolismo , Mastócitos/patologia , Metaloproteinase 9 da Matriz/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Receptores CCR2/metabolismo , Microambiente Tumoral , Neoplasias da Bexiga Urinária/secundário , Animais , Apoptose , Western Blotting , Movimento Celular , Proliferação de Células , Quimiocina CCL2/genética , Receptor beta de Estrogênio/genética , Feminino , Humanos , Técnicas Imunoenzimáticas , Mastócitos/metabolismo , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Nus , Invasividade Neoplásica , Proteínas de Transporte de Cátions Orgânicos/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores CCR2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/imunologia , Neoplasias da Bexiga Urinária/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Oncotarget ; 6(42): 44346-59, 2015 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-26587829

RESUMO

Previous studies indicated the T cells, one of the most common types of immune cells existing in the microenvironment of renal cell carcinoma (RCC), may influence the progression of RCC. The potential linkage of T cells and the estrogen receptor beta (ERß), a key player to impact RCC progression, however, remains unclear. Our results demonstrate that RCC cells can recruit more T cells than non-malignant kidney cells. Using an in vitro matrigel invasion system, we found infiltrating T cells could promote RCC cells invasion via increasing ERß expression and transcriptional activity. Mechanism dissection suggested that co-culturing T cells with RCC cells released more T cell attraction factors, including IFN-γ, CCL3 and CCL5, suggesting a positive regulatory feed-back mechanism. Meanwhile, infiltrating T cells may also promote RCC cell invasion via increased ERß and decreased DAB2IP expressions, and knocking down DAB2IP can then reverse the T cells-promoted RCC cell invasion. Together, our results suggest that infiltrating T cells may promote RCC cell invasion via increasing the RCC cell ERß expression to inhibit the tumor suppressor DAB2IP signals. Further mechanism dissection showed that co-culturing T cells with RCC cells could produce more IGF-1 and FGF-7, which may enhance the ERß transcriptional activity. The newly identified relationship between infiltrating T cells/ERß/DAB2IP signals may provide a novel therapeutic target in the development of agents against RCC.


Assuntos
Carcinoma de Células Renais/metabolismo , Receptor beta de Estrogênio/metabolismo , Neoplasias Renais/metabolismo , Linfócitos do Interstício Tumoral/metabolismo , Comunicação Parácrina , Linfócitos T/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/imunologia , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Quimiocina CCL3/metabolismo , Quimiocina CCL5/metabolismo , Quimiotaxia de Leucócito , Técnicas de Cocultura , Receptor beta de Estrogênio/genética , Fator 7 de Crescimento de Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Interferon gama/metabolismo , Neoplasias Renais/genética , Neoplasias Renais/imunologia , Neoplasias Renais/patologia , Linfócitos do Interstício Tumoral/imunologia , Transdução de Sinais , Linfócitos T/imunologia , Transfecção , Microambiente Tumoral , Proteínas Ativadoras de ras GTPase/genética
11.
BMC Urol ; 15: 97, 2015 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-26391476

RESUMO

BACKGROUND: Genetically engineered mouse models are essential to the investigation of the molecular mechanisms underlying human prostate pathology and the effects of therapy on the diseased prostate. Serial in vivo volumetric imaging expands the scope and accuracy of experimental investigations of models of normal prostate physiology, benign prostatic hyperplasia and prostate cancer, which are otherwise limited by the anatomy of the mouse prostate. Moreover, accurate imaging of hyperplastic and tumorigenic prostates is now recognized as essential to rigorous pre-clinical trials of new therapies. Bioluminescent imaging has been widely used to determine prostate tumor size, but is semi-quantitative at best. Magnetic resonance imaging can determine prostate volume very accurately, but is expensive and has low throughput. We therefore sought to develop and implement a high throughput, low cost, and accurate serial imaging protocol for the mouse prostate. METHODS: We developed a high frequency ultrasound imaging technique employing 3D reconstruction that allows rapid and precise assessment of mouse prostate volume. Wild-type mouse prostates were examined (n = 4) for reproducible baseline imaging, and treatment effects on volume were compared, and blinded data analyzed for intra- and inter-operator assessments of reproducibility by correlation and for Bland-Altman analysis. Examples of benign prostatic hyperplasia mouse model prostate (n = 2) and mouse prostate implantation of orthotopic human prostate cancer tumor and its growth (n = ) are also demonstrated. RESULTS: Serial measurement volume of the mouse prostate revealed that high frequency ultrasound was very precise. Following endocrine manipulation, regression and regrowth of the prostate could be monitored with very low intra- and interobserver variability. This technique was also valuable to monitor the development of prostate growth in a model of benign prostatic hyperplasia. Additionally, we demonstrate accurate ultrasound image-guided implantation of orthotopic tumor xenografts and monitoring of subsequent tumor growth from ~10 to ~750 mm(3) volume. DISCUSSION: High frequency ultrasound imaging allows precise determination of normal, neoplastic and hyperplastic mouse prostate. Low cost and small image size allows incorporation of this imaging modality inside clean animal facilities, and thereby imaging of immunocompromised models. 3D reconstruction for volume determination is easily mastered, and both small and large relative changes in volume are accurately visualized. Ultrasound imaging does not rely on penetration of exogenous imaging agents, and so may therefore better measure poorly vascularized or necrotic diseased tissue, relative to bioluminescent imaging (IVIS). CONCLUSIONS: Our method is precise and reproducible with very low inter- and intra-observer variability. Because it is non-invasive, mouse models of prostatic disease states can be imaged serially, reducing inter-animal variability, and enhancing the power to detect small volume changes following therapeutic intervention.


Assuntos
Interpretação de Imagem Assistida por Computador/métodos , Imageamento Tridimensional/métodos , Próstata/diagnóstico por imagem , Hiperplasia Prostática/diagnóstico por imagem , Neoplasias da Próstata/diagnóstico por imagem , Ultrassonografia/métodos , Animais , Diagnóstico Diferencial , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
12.
Cancer Lett ; 368(1): 71-78, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26231735

RESUMO

Early studies found critical roles for neutrophils in renal cell carcinoma (RCC) progression. However, detailed mechanisms of how infiltrating neutrophils in the kidney tumor microenvironment impact RCC progression remain unclear. Here we found more neutrophils were infiltrated in human RCC lesions than those found in surrounding normal kidney tissues. Similarly, in vitro studies also revealed that RCC cells recruited more neutrophil HL-60N cells than normal kidney epithelial cells. Furthermore, in vitro and in vivo experiments also showed that the infiltrated neutrophils could promote RCC cell growth. Mechanism studies showed that co-culture of RCC cells with neutrophil HL-60N cells could selectively upregulate the androgen receptor (AR) signals, which might then alter the c-Myc signals. Interruption approaches using AR-siRNA to knock down AR in RCC cells blocked neutrophil-enhanced RCC cell proliferation. In vivo data using an orthotopically xenografted RCC mouse model also confirmed that infiltrated neutrophils could promote RCC proliferation via modulating the expressions of related cytokines. Together, these results conclude that infiltrated neutrophils may function through modulating the AR → c-Myc signals to promote RCC cell proliferation. Targeting this newly identified infiltrating neutrophil → AR → c-Myc signal pathway in the kidney tumor microenvironment may provide a new potential therapy to better suppress RCC progression.


Assuntos
Comunicação Celular , Proliferação de Células , Neoplasias Renais/metabolismo , Infiltração de Neutrófilos , Neutrófilos/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptores Androgênicos/metabolismo , Transdução de Sinais , Adulto , Idoso , Animais , Linhagem Celular Tumoral , Técnicas de Cocultura , Feminino , Humanos , Neoplasias Renais/imunologia , Neoplasias Renais/patologia , Masculino , Camundongos Nus , Pessoa de Meia-Idade , Neutrófilos/imunologia , Fatores de Tempo , Carga Tumoral , Microambiente Tumoral
13.
Oncotarget ; 6(22): 19290-304, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26079540

RESUMO

Neutrophils make up a significant portion of the infiltrated immune cells found in the tumor microenvironment. Here we found more infiltrated neutrophils in human renal cell carcinoma (RCC) lesions than adjacent benign areas. In vitro RCC studies showed that neutrophils (HL-60N cells) infiltrated toward RCC cells and subsequently enhanced RCC cell migration and invasion. Co-culture of RCC cells with HL-60N cells up-regulated ERß, VEGFa and HIF2α mRNA levels. ERß signals increased RCC cell migration via induction of the VEGFa/HIF2α pathway. Treatment of HIF inhibitor or rapamycin, or knockdown of ERß in RCC cells reversed HL-60N-promoted RCC migration. In vivo data using orthotopically xenografted RCC mouse model confirmed that infiltrated neutrophils promoted RCC migration via modulating the expressions of ERß, VEGFa and HIF2α signal pathways. Together, our studies revealed that neutrophils are favorably recruited to the RCC cells to promote the RCC migration and invasion. Targeting the infiltrating RCC tumor microenvironment with anti-estrogen or rapamycin may be a potential therapy to suppress RCC progression.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Carcinoma de Células Renais/patologia , Receptor beta de Estrogênio/metabolismo , Neutrófilos/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto , Idoso , Animais , Carcinoma de Células Renais/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Feminino , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Neutrófilos/metabolismo , Transdução de Sinais , Transfecção , Microambiente Tumoral
14.
Am J Cancer Res ; 5(3): 1146-57, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26045993

RESUMO

Epidemiological studies indicate that women have a higher chance of developing muscle invasive bladder cancer (BCa) than men, suggesting that estrogen and estrogen receptors (ERs) may play critical roles in BCa progression. However, the ERs roles in the bladder tumor microenvironment and impacts on BCa progression remain largely unclear. Using IHC staining in human BCa samples, we found that higher ERα expression in the stromal compartment of BCa may be correlated with unfavorable clinical outcomes. Results from cell line studies revealed that co-culturing with fibroblasts could promote BCa T24, UMUC3 and 5637 cells invasion. Importantly, addition of ERα in fibroblasts further enhanced the BCa cell invasion and knock-down of ERα in fibroblasts could then partially reduce the fibroblasts-enhanced BCa invasion. Mechanism dissection suggested that ERα could function through modulating the CCL cytokines expression in fibroblasts to increase the BCa IL-6 expression. An interruption approach using IL-6 neutralizing antibody then reversed the fibroblast ERα-enhanced BCa cell invasion. Together, these data suggest that the higher expression of ERα in fibroblasts may be the result of modulating the CCL1 expression in fibroblasts and/or IL-6 production in BCa cells to enhance BCa cells invasion. Targeting these individual molecules in this newly identified ERα-stimulated CCL1 and IL-6 signal pathways may become an alternative therapy to better suppress the BCa cell invasion.

15.
Am J Clin Exp Urol ; 2(2): 161-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25374919

RESUMO

Prostate cancer (PCa) is an androgen-sensitive disease, which can be pharmacologically controlled by androgen blockade. To date, a growing body of evidence showed that estrogen and estrogen receptors (ERs) could regulate prostate development, as well as cancer initiation and progression. This review will address the expression levels and function of ERs in different stages of PCa progression. The functions of ERs in different types of prostate cells, the ligand effect, and the potential applications of selective estrogen modulators (SERMs) will also be discussed. To further dissect ERs' roles in prostate development, cell type specific ER knockout mouse models were generated. Results collected from the prostate cell type-specific ERαKO mouse models provided new insights about the cell type specific ERα roles in prostate development prenatally and postnatally. The results of ERs' roles in mouse PCa mode and the correlation of ERs expression and biomedical outcome will also be discussed.

16.
Oncotarget ; 5(17): 7917-35, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25277204

RESUMO

Clinical reports show males have a higher bladder cancer (BCa) incidence than females. The sexual difference of BCa occurrence suggests that estrogen and its receptors may affect BCa development. Estrogen receptor alpha (ERα) is the classic receptor to convey estrogen signaling, however, the function of ERα in BCa development remains largely unknown. To understand the in vivo role of ERα in BCa development, we generated total and urothelial specific ERα knockout mice (ERαKO) and used the pre- carcinogen BBN to induce BCa. Earlier reports showed that ERα promotes breast and ovarian cancers in females. Surprisingly and of clinical importance, our results showed that ERα inhibits BCa development and loss of the ERα gene results in an earlier onset and higher incidence of BBN-induced in vivo mouse BCa. Supportively, carcinogen induced malignant transformation ability was reduced in ERα expressing urothelial cells as compared to ERα negative cells. Mechanism studies suggest that ERα could control the expression of INPP4B to reduce AKT activity and consequently reduce BCa cell growth. In addition, IHC staining of clinical sample analyses show that INPP4B expression, in correlation with reduced ERα, is significantly reduced in human BCa specimens. Together, this is the first report using the in vivo cre-loxP gene knockout mouse model to characterize ERα roles in BCa development. Our studies provide multiple in vitro cell studies and in vivo animal model data as well as human BCa tissue analyses to prove ERα plays a protective role in BCa initiation and growth at least partly via modulating the INPP4B/Akt pathway.


Assuntos
Carcinoma de Células de Transição/genética , Receptor alfa de Estrogênio/genética , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias da Bexiga Urinária/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Carcinoma de Células de Transição/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia , Análise Serial de Tecidos , Neoplasias da Bexiga Urinária/metabolismo
17.
Biomed Res Int ; 2014: 713263, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25221777

RESUMO

Estrogen and estrogen receptor (ER)-mediated signaling pathways play important roles in the etiology and progression of human breast, endometrial, and ovarian cancers. Attenuating ER activities by natural products and their derivatives is a relatively practical strategy to control and reduce breast, endometrial, and ovarian cancer risk. Here, we found 3-butoxy-1,8,9-trihydroxy-6H-benzofuro[3,2-c]benzopyran-6-one (BTB), a new derivative of wedelolactone, could effectively inhibit the 17-estradiol (E2)-induced ER transactivation and suppress the growth of breast cancer as well as endometrial and ovarian cancer cells. Our results indicate that 2.5 µM BTB effectively suppresses ER-positive, but not ER-negative, breast, endometrial, and ovarian cancer cells. Furthermore, our data indicate that BTB can modulate ER transactivation and suppress the expression of E2-mediated ER target genes (Cyclin D1, E2F1, and TERT) in the ER-positive MCF-7, Ishikawa, and SKOV-3 cells. Importantly, this BTB mediated inhibition of ER activity is selective since BTB does not suppress the activities of other nuclear receptors, including glucocorticoid receptor and progesterone receptor, suggesting that BTB functions as a selective ER signaling inhibitor with the potential to treat breast, endometrial, and ovarian cancers.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Cumarínicos/administração & dosagem , Neoplasias do Endométrio/tratamento farmacológico , Receptor alfa de Estrogênio/antagonistas & inibidores , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Estradiol/administração & dosagem , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Transdução de Sinais/efeitos dos fármacos
18.
Carcinogenesis ; 35(6): 1301-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24374826

RESUMO

The prostate cancer (PCa) microenvironment contains active stromal cells known as cancer-associated fibroblasts (CAF) that may play important roles in influencing tumor progression. Here we studied the role of CAF estrogen receptor alpha (ERα) and found that it could protect against PCa invasion. Immunohistochemistry on prostatectomy specimens showed that PCa patients with ERα-positive stroma had a significantly lower risk for biochemical recurrence. In vitro invasion assays further confirmed that the stromal ERα was able to reduce PCa cell invasion. Dissection of the molecular mechanism revealed that the CAF ERα could function through a CAF-epithelial interaction via selectively upregulating thrombospondin 2 (Thbs2) and downregulating matrix metalloproteinase 3 (MMP3) at the protein and messenger RNA levels. Chromatin immunoprecipitation assays further showed that ERα could bind to an estrogen response element on the promoter of Thbs2. Importantly, knockdown of Thbs2 led to increased MMP3 expression and interruption of the ERα mediated invasion suppression, providing further evidence of an ERα-Thbs2-MMP3 axis in CAF. In vivo studies using athymic nude mice injected with CWR22Rv1 (22Rv1) PCa epithelial cells and CAF cells ± ERα also confirmed that mice coimplanted with PCa cells and CAF ERα+ cells had less tumor foci in the pelvic lymph nodes, less metastases, and tumors showed less angiogenesis, MMP3, and MMP9 (an MMP3 downstream target) positive staining. Together, these data suggest that CAF ERα could play protective roles in suppressing PCa metastasis. Our results may lead to developing new and alternative therapeutic approaches to battle PCa via controlling ERα signaling in CAF.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Metaloproteinase 3 da Matriz/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Trombospondinas/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Imuno-Histoquímica , Masculino , Metaloproteinase 3 da Matriz/genética , Invasividade Neoplásica , Fenótipo , Neoplasias da Próstata/genética , Neoplasias da Próstata/mortalidade , Ligação Proteica , Interferência de RNA , Células Estromais/metabolismo , Células Estromais/patologia , Trombospondinas/genética
19.
J Mol Cell Biol ; 5(1): 14-26, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22831834

RESUMO

The androgen deprivation therapy (ADT) to systematically suppress/reduce androgens binding to the androgen receptor (AR) has been the standard therapy for prostate cancer (PCa); yet, most of ADT eventually fails leading to the recurrence of castration resistant PCa. Here, we found that the PCa patients who received ADT had increased PCa stem/progenitor cell population. The addition of the anti-androgen, Casodex, or AR-siRNA in various PCa cells led to increased stem/progenitor cells, whereas, in contrast, the addition of functional AR led to decreased stem/progenitor cell population but increased non-stem/progenitor cell population, suggesting that AR functions differentially in PCa stem/progenitor vs. non-stem/progenitor cells. Therefore, the current ADT might result in an undesired expansion of PCa stem/progenitor cell population, which explains why this therapy fails. Using various human PCa cell lines and three different mouse models, we concluded that targeting PCa non-stem/progenitor cells with AR degradation enhancer ASC-J9 and targeting PCa stem/progenitor cells with 5-azathioprine and γ-tocotrienol resulted in a significant suppression of the tumors at the castration resistant stage. This suggests that a combinational therapy that simultaneously targets both stem/progenitor and non-stem/progenitor cells will lead to better therapeutic efficacy and may become a new therapy to battle the PCa before and after castration resistant stages.


Assuntos
Células-Tronco Neoplásicas/metabolismo , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Transdução de Sinais , Animais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Curcumina/análogos & derivados , Curcumina/farmacologia , Decitabina , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Células-Tronco Neoplásicas/efeitos dos fármacos , Orquiectomia , Neoplasias da Próstata/genética , Neoplasias da Próstata/terapia , Receptores Androgênicos/genética , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Mol Endocrinol ; 27(1): 38-49, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23204329

RESUMO

Estrogen signaling, through estrogen receptor (ER)α, has been shown to cause hypertrophy in the prostate. Our recent report has shown that epithelial ERα knockout (KO) will not affect the normal prostate development or homeostasis. However, it remains unclear whether ERα in different types of stromal cells has distinct roles in prostate development. This study proposed to elucidate how KO of ERα in the stromal smooth muscle or fibroblast cells may interrupt cross talk between prostate stromal and epithelial cells. Smooth muscle ERαKO (smERαKO) mice showed decreased glandular infolding with the proximal area exhibiting a significant decrease. Fibroblast ERαKO mouse prostates did not exhibit this phenotype but showed a decrease in the number of ductal tips. Additionally, the amount of collagen observed in the basement membrane was reduced in smERαKO prostates. Interestingly, these phenotypes were found to be mutually exclusive among smERαKO or fibroblast ERαKO mice. Compound KO of ERα in both fibroblast and smooth muscle showed combined phenotypes from each of the single KO. Further mechanistic studies showed that IGF-I and epidermal growth factor were down-regulated in prostate smooth muscle PS-1 cells lacking ERα. Together, our results indicate the distinct functions of fibroblast vs. smERα in prostate development.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Fibroblastos/metabolismo , Miócitos de Músculo Liso/metabolismo , Próstata/crescimento & desenvolvimento , Animais , Membrana Basal/metabolismo , Linhagem Celular , Colágeno/metabolismo , Fator de Crescimento Epidérmico/fisiologia , Estradiol/fisiologia , Receptor alfa de Estrogênio/genética , Feminino , Técnicas de Inativação de Genes , Homeostase , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Organogênese , Próstata/citologia , Próstata/metabolismo , Somatomedinas/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...