Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
3.
Hypertension ; 66(4): 881-91, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26324502

RESUMO

Sex difference is a risk factor for abdominal aortic aneurysm (AAA) formation yet the reason for male predominance remains unclear. Androgen and the androgen receptor (AR) influence the male sex difference, indicating that AR signaling may affect AAA development. Using angiotensin II­induced AAA in apolipoprotein E null mouse models (82.4% AAA incidence), we found that mice lacking AR failed to develop AAA and aorta had dramatically reduced macrophages infiltration and intact elastic fibers. These findings suggested that AR expression in endothelial cells, macrophages, or smooth muscle cells might play a role in AAA development. Selective knockout of AR in each of these cell types further demonstrated that mice lacking AR in macrophages (20% AAA incidence) or smooth muscle cells (12.5% AAA incidence) but not in endothelial cells (71.4% AAA incidence) had suppressed AAA development. Mechanism dissection showed that AR functioned through modulation of interleukin-1α (IL-1α) and transforming growth factor-ß1 signals and by targeting AR with the AR degradation enhancer ASC-J9 led to significant suppression of AAA development. These results demonstrate the underlying mechanism by which AR influences AAA development is through IL-1α and transforming growth factor-ß1, and provides a potential new therapy to suppress/prevent AAA by targeting AR with ASC-J9.


Assuntos
Aneurisma da Aorta Abdominal/genética , Regulação da Expressão Gênica , Inflamação/genética , Interleucina-1alfa/genética , RNA/genética , Receptores Androgênicos/genética , Fator de Crescimento Transformador beta1/genética , Animais , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Inflamação/metabolismo , Inflamação/patologia , Interleucina-1alfa/biossíntese , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Receptores Androgênicos/biossíntese , Transdução de Sinais , Fator de Crescimento Transformador beta1/biossíntese
4.
Tumour Biol ; 35(3): 1881-90, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24122203

RESUMO

Recent data suggested that tissue human kallikrein 2 (KLK2) might be involved in the carcinogenesis and tumor metastasis of prostate cancer (PCa). However, the detailed pathophysiological roles of KLK2 in PCa remain unclear. We report here that KLK2 may be treated as a potential therapeutic target in castration-resistant PCa (CRPC). Histologic analyses show that the increased KLK2 expression is correlated with higher cell proliferation rate and lower cell apoptosis index in CRPC specimens. Adding functional KLK2 cDNA into high passage LNCaP cells led to increased cell growth, and knockdown of KLK2 expression with KLK2-siRNA in LNCaP cells resulted in increased cell apoptosis with cell growth arrest at the G1 phase. Results from in vitro colony formation assay and in vivo xenografted PCa tissues also demonstrated that targeting KLK2 led to suppressed growth of PCa in the castration-resistant stage. Further mechanism dissection shows that KLK2 may cooperate with the AR coregulator, ARA70, to enhance AR transactivation that may result in alteration of PCa formation. Together, these results suggested KLK2 might become a new therapeutic target to battle the CRPC and KLK2-siRNA may be developed as an alternative approach to suppress PCa growth.


Assuntos
Proliferação de Células , Coativadores de Receptor Nuclear/metabolismo , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Calicreínas Teciduais/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/fisiologia , Xenoenxertos , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos Endogâmicos BALB C , Neoplasias da Próstata/patologia , Reação em Cadeia da Polimerase em Tempo Real , Ativação Transcricional , Transfecção
5.
Cancer Res ; 73(18): 5633-46, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23878190

RESUMO

Infiltrating macrophages are a key component of inflammation during tumorigenesis, but the direct evidence of such linkage remains unclear. We report here that persistent coculturing of immortalized prostate epithelial cells with macrophages, without adding any carcinogens, induces prostate tumorigenesis and that induction involves the alteration of signaling of macrophage androgen receptor (AR)-inflammatory chemokine CCL4-STAT3 activation as well as epithelial-to-mesenchymal transition and downregulation of p53/PTEN tumor suppressors. In vivo studies further showed that PTEN(+/-) mice lacking macrophage AR developed far fewer prostatic intraepithelial neoplasia (PIN) lesions, supporting an in vivo role for macrophage AR during prostate tumorigenesis. CCL4-neutralizing antibody effectively blocked macrophage-induced prostate tumorigenic signaling and targeting AR via an AR-degradation enhancer, ASC-J9, reduced CCL4 expression, and xenografted tumor growth in vivo. Importantly, CCL4 upregulation was associated with increased Snail expression and downregulation of p53/PTEN in high-grade PIN and prostate cancer. Together, our results identify the AR-CCL4-STAT3 axis as key regulators during prostate tumor initiation and highlight the important roles of infiltrating macrophages and inflammatory cytokines for the prostate tumorigenesis.


Assuntos
Quimiocina CCL4/metabolismo , Macrófagos/patologia , Próstata/patologia , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/patologia , Receptores Androgênicos/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Transformação Celular Neoplásica , Células Cultivadas , Curcumina/análogos & derivados , Curcumina/farmacologia , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Transição Epitelial-Mesenquimal , Humanos , Técnicas Imunoenzimáticas , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , PTEN Fosfo-Hidrolase/fisiologia , Próstata/imunologia , Próstata/metabolismo , Neoplasia Prostática Intraepitelial/imunologia , Neoplasia Prostática Intraepitelial/metabolismo , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/metabolismo , Transdução de Sinais
6.
Mol Endocrinol ; 27(10): 1617-31, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23893956

RESUMO

Stromal-epithelial interaction plays a pivotal role to mediate the normal prostate growth, the pathogenesis of benign prostatic hyperplasia (BPH), and prostate cancer development. Until now, the stromal androgen receptor (AR) functions in the BPH development, and the underlying mechanisms remain largely unknown. Here we used a genetic knockout approach to ablate stromal fibromuscular (fibroblasts and smooth muscle cells) AR in a probasin promoter-driven prolactin transgenic mouse model (Pb-PRL tg mice) that could spontaneously develop prostate hyperplasia to partially mimic human BPH development. We found Pb-PRL tg mice lacking stromal fibromuscular AR developed smaller prostates, with more marked changes in the dorsolateral prostate lobes with less proliferation index. Mechanistically, prolactin mediated hyperplastic prostate growth involved epithelial-stromal interaction through epithelial prolactin/prolactin receptor signals to regulate granulocyte macrophage-colony stimulating factor expression to facilitate stromal cell growth via sustaining signal transducer and activator of transcription-3 activity. Importantly, the stromal fibromuscular AR could modulate such epithelial-stromal interacting signals. Targeting stromal fibromuscular AR with the AR degradation enhancer, ASC-J9(®), led to the reduction of prostate size, which could be used in future therapy.


Assuntos
Fibroblastos/metabolismo , Hiperplasia Prostática/metabolismo , Receptores Androgênicos/genética , Células Estromais/metabolismo , Proteína de Ligação a Androgênios/genética , Animais , Proliferação de Células , Células Cultivadas , Curcumina/análogos & derivados , Curcumina/farmacologia , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal , Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Prolactina/fisiologia , Próstata/metabolismo , Próstata/patologia , Hiperplasia Prostática/tratamento farmacológico , Hiperplasia Prostática/patologia , Hiperplasia Prostática/fisiopatologia , Proteólise/efeitos dos fármacos , Receptores Androgênicos/metabolismo , Fator de Transcrição STAT3/metabolismo
7.
Stem Cell Res ; 11(2): 938-50, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23859805

RESUMO

Gender differences have been described in osteoporosis with females having a higher risk of osteoporosis than males. The differentiation of bone marrow stromal cells (BMSCs) into bone or fat is a critical step for osteoporosis. Here we demonstrated that loss of the androgen receptor (AR) in BMSCs suppressed osteogenesis but promoted adipogenesis. The mechanism dissection studies revealed that AR deficiency suppressed osteogenesis-related genes to inhibit osteoblast differentiation from BMSCs. Knockout of AR promoted adipogenesis of BMSCs via Akt activation through IGFBP3-mediated IGF signaling, and the 5' promoter assay and chromatin immunoprecipitation assays further proved that AR could modulate IGFBP3 expression at the transcriptional level. Finally, addition of IGF inhibitors successfully masked the AR deficiency-induced Akt activation, and inhibitions of Akt, IGF1, and IGF2 pathways reversed the AR depletion effects on the adipogenesis process. These results suggested that AR-mediated changes in IGFBP3 might modulate the IGF-Akt axis to regulate adipogenesis in BMSCs.


Assuntos
Adipogenia/fisiologia , Células da Medula Óssea/citologia , Osteogênese/efeitos dos fármacos , Receptores Androgênicos/deficiência , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Animais , Células da Medula Óssea/metabolismo , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Feminino , Técnicas de Inativação de Genes , Humanos , Masculino , Camundongos , Osteogênese/genética , Osteogênese/fisiologia , Osteoporose/genética , Osteoporose/metabolismo , Receptores Androgênicos/metabolismo , Transdução de Sinais , Células Estromais/citologia , Células Estromais/fisiologia
8.
Am J Pathol ; 182(6): 1942-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23570837

RESUMO

Benign prostate hyperplasia (BPH) is a major cause of lower urinary tract symptoms, with an increased volume of transitional zone and associated with increased stromal cells. It is known that androgen/androgen receptor (AR) signaling plays a key role in development of BPH, and that blockade of this signaling decreases BPH volume and can relieve lower urinary tract symptoms, but the mechanisms of androgen/AR signaling in BPH development remain unclear, and the effectiveness of current drugs for treating BPH is still limited. The detailed mechanisms of androgen/AR signaling need to be clarified, and new therapies are needed for better treatment of BPH patients. This review focuses on roles of AR in epithelial and stromal cells in BPH development. In epithelial cells, AR may contribute to BPH development via epithelial cell-stromal cell interaction with alterations of epithelial-mesenchymal transition, leading to proliferation of stromal cells. Data from several mouse models with selective knockout of AR in stromal smooth-muscle cells and/or fibroblasts indicate that the AR in stromal cells can also promote BPH development. In prostatic inflammation, AR roles in infiltrating macrophages and epithelial and stromal cells have been linked to BPH development, which has led to discovery of new therapeutic targets. For example, targeting AR with the novel AR degradation enhancer, ASC-J9 offers a potential therapeutic approach against BPH development.


Assuntos
Hiperplasia Prostática/patologia , Receptores Androgênicos/fisiologia , Antagonistas de Receptores de Andrógenos/uso terapêutico , Animais , Comunicação Celular/fisiologia , Proliferação de Células , Células Epiteliais/patologia , Células Epiteliais/fisiologia , Humanos , Masculino , Camundongos , Hiperplasia Prostática/tratamento farmacológico , Hiperplasia Prostática/fisiopatologia , Transdução de Sinais/fisiologia , Células Estromais/patologia , Células Estromais/fisiologia
9.
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
10.
Am J Pathol ; 182(2): 460-73, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23219429

RESUMO

Using androgen receptor (AR) knockout mice to determine AR functions in selective prostate cancer (PCa) cells, we determined that AR might play differential roles in various cell types, either to promote or suppress PCa development/progression. These observations partially explain the failure of current androgen deprivation therapy (ADT) to reduce/prevent androgen binding to AR in every cell. Herein, we identified the AR degradation enhancer ASC-J9, which selectively degrades AR protein via interruption of the AR-AR selective coregulator interaction. Such selective interruption could, therefore, suppress AR-mediated PCa growth in the androgen-sensitive stage before ADT and in the castration-resistant stage after ADT. Mechanistic dissection suggested that ASC-J9 could activate the proteasome-dependent pathway to promote AR degradation through the enhanced association of AR-Mdm2 complex. The consequences of ASC-J9-promoted AR degradation included reduced androgen binding to AR, AR N-C terminal interaction, and AR nuclear translocation. Such inhibitory regulation could then result in suppression of AR transactivation and AR-mediated cell growth in eight different mouse models, including intact or castrated nude mice xenografted with androgen-sensitive LNCaP cells or androgen-insensitive C81 cells and castrated nude mice xenografted with castration-resistant C4-2 and CWR22Rv1 cells, and TRAMP and Pten(+/-) mice. These results demonstrate that ASC-J9 could serve as an AR degradation enhancer that effectively suppresses PCa development/progression in the androgen-sensitive and castration-resistant stages.


Assuntos
Castração , Curcumina/análogos & derivados , Próstata/metabolismo , Próstata/patologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Receptores Androgênicos/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Quimioprevenção , Curcumina/efeitos adversos , Curcumina/uso terapêutico , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas com Domínio LIM/metabolismo , Masculino , Camundongos , Camundongos Nus , Coativadores de Receptor Nuclear/metabolismo , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/metabolismo , Próstata/efeitos dos fármacos , Próstata/cirurgia , Neoplasias da Próstata/cirurgia , Proteólise/efeitos dos fármacos , Receptores Androgênicos/genética , Transcrição Gênica/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Hepatology ; 57(4): 1550-63, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23150236

RESUMO

UNLABELLED: Transplantation of bone marrow mesenchymal stem cells (BM-MSCs) has been considered as an alternative therapy, replacing liver transplantation in clinical trials, to treat liver cirrhosis, an irreversible disease that may eventually lead to liver cancer development. However, low survival rate of the BM-MSCs leading to unsatisfactory efficacy remains a major concern. Gender differences have been suggested in BM-MSCs therapeutic application, but the effect of the androgen receptor (AR), a key factor in male sexual phenotype, in this application is not clear. Using two liver cirrhosis mouse models induced by CCl4 or thioacetamide, we showed that targeting AR in the BM-MSCs improved their self-renewal and migration potentials and increased paracrine effects to exert anti-inflammatory and anti-fibrotic actions to enhance liver repair. Mechanism dissection studies suggested that knocking out AR in BM-MSCs led to improved self-renewal and migration by alteration of the signaling of epidermal growth factor receptor and matrix metalloproteinase 9 and resulted in suppression of infiltrating macrophages and hepatic stellate cell activation through modulation of interleukin (IL)1R/IL1Ra signaling. Therapeutic approaches using either AR/small interfering RNA or the AR degradation enhancer, ASC-J9, to target AR in BM-MSCs all led to increased efficacy for liver repair. CONCLUSION: Targeting AR, a key factor in male sexual phenotype, in BM-MSCs improves transplantation therapeutic efficacy for treating liver fibrosis.


Assuntos
Tetracloreto de Carbono/efeitos adversos , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/cirurgia , Transplante de Células-Tronco Mesenquimais , Receptores Androgênicos/genética , Tioacetamida/efeitos adversos , Animais , Modelos Animais de Doenças , Receptores ErbB/metabolismo , Feminino , Cirrose Hepática/metabolismo , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Fenótipo , RNA Interferente Pequeno/farmacologia , Receptores Androgênicos/efeitos dos fármacos , Receptores de Interleucina-1/metabolismo , Transdução de Sinais/fisiologia , Resultado do Tratamento
12.
Mol Endocrinol ; 27(1): 25-37, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23250486

RESUMO

Although thymic involution has been linked to the increased testosterone in males after puberty, its detailed mechanism and clinical application related to T-cell reconstitution in bone marrow transplantation (BMT) remain unclear. By performing studies with reciprocal BMT and cell-specific androgen receptor (AR) knockout mice, we found that AR in thymic epithelial cells, but not thymocytes or fibroblasts, played a more critical role to determine thymic cellularity. Further dissecting the mechanism using cell-specific thymic epithelial cell-AR knockout mice bearing T-cell receptor transgene revealed that elevating thymocyte survival was due to the enhancement of positive selection resulting in increased positively selected T-cells in both male and female mice. Targeting AR, instead of androgens, either via genetic knockout of thymic epithelial AR or using an AR-degradation enhancer (ASC-J9®), led to increased BMT grafting efficacy, which may provide a new therapeutic approach to boost T-cell reconstitution in the future.


Assuntos
Transplante de Medula Óssea/métodos , Células Epiteliais/metabolismo , Receptores Androgênicos/metabolismo , Linfócitos T/fisiologia , Timo/citologia , Animais , Células da Medula Óssea/metabolismo , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Curcumina/análogos & derivados , Curcumina/farmacologia , Feminino , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão , Proteólise/efeitos dos fármacos , Receptores Androgênicos/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Timócitos/metabolismo , Timócitos/fisiologia , Timo/anatomia & histologia
13.
EMBO Mol Med ; 4(8): 791-807, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22745041

RESUMO

Stromal-epithelial interaction is crucial to mediate normal prostate and prostate cancer (PCa) development. The indispensable roles of mesenchymal/stromal androgen receptor (AR) for the prostate organogenesis have been demonstrated by using tissue recombination from wild-type and testicular feminized mice. However, the stromal AR functions in the tumour microenvironment and the underlying mechanisms governing the interactions between the epithelium and stroma are not completely understood. Here, we have established the first animal model with AR deletion in stromal fibromuscular cells (dARKO, AR knockout in fibroblasts and smooth muscle cells) in the Pten(+/-) mouse model that can spontaneously develop prostatic intraepithelial neoplasia (PIN). We found that loss of stromal fibromuscular AR led to suppression of PIN lesion development with alleviation of epithelium proliferation and tumour-promoting microenvironments, including extracellular matrix (ECM) remodelling, immune cell infiltration and neovasculature formation due, in part, to the modulation of pro-inflammatory cytokines/chemokines. Finally, targeting stromal fibromuscular AR with the AR degradation enhancer, ASC-J9®, resulted in the reduction of PIN development/progression, which might provide a new approach to suppress PIN development.


Assuntos
Citocinas/biossíntese , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/patologia , Receptores Androgênicos/deficiência , Receptores Androgênicos/metabolismo , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Knockout , PTEN Fosfo-Hidrolase/genética
14.
J Biol Chem ; 287(22): 18376-85, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22474290

RESUMO

Infiltrated macrophages may play important roles in the development and progression of benign prostatic hyperplasia (BPH), but the underlying mechanisms remain largely unknown. We found increased macrophages infiltration in human and mouse BPH tissues. By establishing a co-culture transwell system, we found increased migration of macrophages and proliferation of prostate stromal cells during co-culture. Importantly, stromal androgen receptor (AR) could enhance the migration of macrophages and macrophage-mediated stromal cell proliferation. We identified CCL3 as an AR downstream player, and found CCL3 levels were notably increased in human and mouse BPH prostates. Ablation of prostate stromal AR in a mouse BPH model significantly reduced CCL3 expression levels in prostates. Consistently, targeting AR via an AR degradation enhancer, ASC-J9®, or neutralization of CCL3 with an antibody, resulted in suppression of macrophage migration and prostate stromal cell growth. Our study provides mechanistic insights on the regulation of prostate stromal cells by macrophages via stromal AR/CCL3 signaling pathways, which could potentially allow the development of therapeutic approaches for battling BPH with persistent inflammation.


Assuntos
Macrófagos/patologia , Próstata/patologia , Hiperplasia Prostática/patologia , Receptores Androgênicos/fisiologia , Células Estromais/patologia , Animais , Sequência de Bases , Western Blotting , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Hiperplasia Prostática/fisiopatologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
J Endocrinol ; 213(2): 173-82, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22393245

RESUMO

Early studies have reported the differential roles of androgen receptor (AR) in different types (luminal, basal intermediate, and stromal) of prostate cancer cells. In vivo mouse model tumor studies using the total prostate epithelial knockout mice (pes-ARKO) also revealed that AR played a suppressive role in proliferation of the CK5(+)/CK8(+) progenitor/intermediate cells but a positive role in the CK5(-)/CK8(+) luminal epithelial cells. Using three different resources (one human basal epithelial cell line, one mouse basal epithelial originated progenitor cell line, and a basal epithelium-specific ARKO mouse model), we here demonstrated that the AR in basal epithelial cells of normal prostate plays a suppressive role in their proliferation but a positive role in differentiation into luminal epithelial cells. These results led us to conclude that ARs may play a negative role to suppress CK5(+) basal epithelial and progenitor cell proliferation, yet play an essential role to drive basal epithelial cells into more differentiated states. These results may explain why differential AR expression in different cell types within normal prostate is needed and suggest that ARs in prostate basal epithelial cells, although expressed at a very low level, are necessary to maintain the balance between progenitor cells and differentiated luminal epithelial cells.


Assuntos
Próstata/citologia , Receptores Androgênicos/metabolismo , Células-Tronco/citologia , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Proliferação de Células , Células Epiteliais/citologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Receptores Androgênicos/biossíntese , Receptores Androgênicos/genética
16.
Neoplasia ; 14(1): 74-83, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22355276

RESUMO

Early studies suggested androgen receptor (AR) splice variants might contribute to the progression of prostate cancer (PCa) into castration resistance. However, the therapeutic strategy to target these AR splice variants still remains unresolved. Through tissue survey of tumors from the same patients before and after castration resistance, we found that the expression of AR3, a major AR splice variant that lacks the AR ligand-binding domain, was substantially increased after castration resistance development. The currently used antiandrogen, Casodex, showed little growth suppression in CWR22Rv1 cells. Importantly, we found that AR degradation enhancer ASC-J9 could degrade both full-length (fAR) and AR3 in CWR22Rv1 cells as well as in C4-2 and C81 cells with addition of AR3. The consequences of such degradation of both fAR and AR3 might then result in the inhibition of AR transcriptional activity and cell growth in vitro. More importantly, suppression of AR3 specifically by short-hairpin AR3 or degradation of AR3 by ASC-J9 resulted in suppression of AR transcriptional activity and cell growth in CWR22Rv1-fARKD (fAR knockdown) cells in which DHT failed to induce, suggesting the importance of targeting AR3. Finally, we demonstrated the in vivo therapeutic effects of ASC-J9 by showing the inhibition of PCa growth using the xenografted model of CWR22Rv1 cells orthotopically implanted into castrated nude mice with undetectable serum testosterone. These results suggested that targeting both fAR- and AR3-mediated PCa growth by ASC-J9 may represent the novel therapeutic approach to suppress castration-resistant PCa. Successful clinical trials targeting both fAR and AR3 may help us to battle castration-resistant PCa in the future.


Assuntos
Antineoplásicos/farmacologia , Curcumina/análogos & derivados , Neoplasias da Próstata/genética , Receptores Androgênicos/genética , Antagonistas de Androgênios/farmacologia , Anilidas/farmacologia , Animais , Western Blotting , Castração , Linhagem Celular Tumoral , Curcumina/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Nus , Nitrilas/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores Androgênicos/metabolismo , Compostos de Tosil/farmacologia
17.
Mol Endocrinol ; 26(1): 52-66, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22135068

RESUMO

Using the cre-loxP system, we generated a new mouse model [double stromal androgen receptor knockout (dARKO)] with selectively deleted androgen receptor (AR) in both stromal fibroblasts and smooth muscle cells, and found the size of the anterior prostate (AP) lobes was significantly reduced as compared with those from wild-type littermate controls. The reduction in prostate size of the dARKO mouse was accompanied by impaired branching morphogenesis and partial loss of the infolding glandular structure. Further dissection found decreased proliferation and increased apoptosis of the prostate epithelium in the dARKO mouse AP. These phenotype changes were further confirmed with newly established immortalized prostate stromal cells (PrSC) from wild-type and dARKO mice. Mechanistically, IGF-1, placental growth factor, and secreted phosphoprotein-1 controlled by stromal AR were differentially expressed in PrSC-wt and PrSC-ARKO. Moreover, the conditioned media (CM) from PrSC-wt promoted prostate epithelium growth significantly as compared with CM from PrSC-dARKO. Finally, adding IGF-1/placental growth factor recombinant proteins into PrSC-dARKO CM was able to partially rescue epithelium growth. Together, our data concluded that stromal fibromuscular AR could modulate epithelium growth and maintain cellular homeostasis through identified growth factors.


Assuntos
Células Epiteliais/metabolismo , Epitélio/crescimento & desenvolvimento , Morfogênese , Próstata/crescimento & desenvolvimento , Receptores Androgênicos/genética , Receptores Androgênicos/fisiologia , Células Estromais/metabolismo , Animais , Apoptose , Proliferação de Células , Células Cultivadas , Meios de Cultivo Condicionados , Células Epiteliais/citologia , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Tamanho do Órgão , Comunicação Parácrina , Fosfoproteínas/metabolismo , Fator de Crescimento Placentário , Proteínas da Gravidez/metabolismo , Próstata/anatomia & histologia , Próstata/citologia , Próstata/metabolismo , Receptores Androgênicos/deficiência , Proteínas Recombinantes/metabolismo , Células Estromais/citologia
18.
Prostate ; 71(5): 517-24, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20945497

RESUMO

BACKGROUND: Androgens and the androgen receptor (AR) play critical roles in the prostate development via mesenchymal-epithelial interactions. Smooth muscle cells (SMC), differentiated from mesenchyme, are one of the basic components of the prostate stroma. However, the roles of smooth muscle AR in prostate development are still obscure. METHODS: We established the smooth muscle selective AR knockout (SM-ARKO) mouse model using the Cre-loxP system, and confirmed the ARKO efficiency at RNA, DNA and protein levels. Then, we observed the prostate morphology changes, and determined the epithelial proliferation, apoptosis, and differentiation. We also knocked down the AR in a prostate smooth muscle cell line (PS-1) to confirm the in vivo findings and to probe the mechanism. RESULTS: The AR was selectively and efficiently knocked out in the anterior prostates of SM-ARKO mouse. The SM-ARKO prostates have defects with loss of infolding structures, and decrease of epithelial proliferation, but with little change of apoptosis and differentiation. The mechanism studies showed that IGF-1 expression level decreased in the SM-ARKO prostates and AR-knockdown PS-1 cells. The decreased IGF-1 expression might contribute to the defective development of SM-ARKO prostates. CONCLUSIONS: The AR in SMCs plays important roles in the prostate development via the regulation of IGF-1 signal.


Assuntos
Fator de Crescimento Insulin-Like I/metabolismo , Músculo Liso/fisiologia , Próstata/fisiologia , Receptores Androgênicos/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Células Epiteliais/metabolismo , Epitélio/fisiologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Fator de Crescimento Insulin-Like I/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Músculo Liso/citologia , Músculo Liso/metabolismo , Próstata/citologia , Próstata/metabolismo , RNA/química , RNA/genética , Receptores Androgênicos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
19.
J Clin Invest ; 119(12): 3739-51, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19907077

RESUMO

Cutaneous wounds heal more slowly in elderly males than in elderly females, suggesting a role for sex hormones in the healing process. Indeed, androgen/androgen receptor (AR) signaling has been shown to inhibit cutaneous wound healing. AR is expressed in several cell types in healing skin, including keratinocytes, dermal fibroblasts, and infiltrating macrophages, but the exact role of androgen/AR signaling in these different cell types remains unclear. To address this question, we generated and studied cutaneous wound healing in cell-specific AR knockout (ARKO) mice. General and myeloid-specific ARKO mice exhibited accelerated wound healing compared with WT mice, whereas keratinocyte- and fibroblast-specific ARKO mice did not. Importantly, the rate of wound healing in the general ARKO mice was dependent on AR and not serum androgen levels. Interestingly, although dispensable for wound closure, keratinocyte AR promoted re-epithelialization, while fibroblast AR suppressed it. Further analysis indicated that AR suppressed wound healing by enhancing the inflammatory response through a localized increase in TNF-alpha expression. Furthermore, AR enhanced local TNF-alpha expression via multiple mechanisms, including increasing the inflammatory monocyte population, enhancing monocyte chemotaxis by upregulating CCR2 expression, and enhancing TNF-alpha expression in macrophages. Finally, targeting AR by topical application of a compound (ASC-J9) that degrades AR protein resulted in accelerated healing, suggesting a potential new therapeutic approach that may lead to better treatment of wound healing.


Assuntos
Macrófagos/imunologia , Macrófagos/fisiologia , Receptores Androgênicos/fisiologia , Pele/lesões , Fator de Necrose Tumoral alfa/biossíntese , Cicatrização/imunologia , Cicatrização/fisiologia , Administração Tópica , Idoso , Antagonistas de Receptores de Andrógenos , Animais , Curcumina/administração & dosagem , Curcumina/análogos & derivados , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Receptores Androgênicos/deficiência , Receptores Androgênicos/genética , Caracteres Sexuais , Pele/efeitos dos fármacos , Pele/imunologia , Pele/fisiopatologia , Testosterona/sangue , Cicatrização/efeitos dos fármacos , Cicatrização/genética
20.
Mol Endocrinol ; 23(4): 444-53, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19164450

RESUMO

Estrogens have been linked to a higher female incidence of autoimmune diseases. The role of androgen and the androgen receptor (AR) in autoimmune diseases, however, remains unclear. Here we report that the lack of AR in B cells in different strains of mice, namely general AR knockout, B cell-specific AR knockout, and naturally occurring testicular feminization mutation AR-mutant mice, as well as castrated wild-type mice, results in increased B cells in blood and bone marrow. Analysis of the targeted mice, together with bone marrow transplantation using Rag1(-/-) recipients, overexpression of retrovirally encoded AR-cDNA, and small interfering RNA-mediated AR mRNA knockdown approaches also show that the B cell expansion results from resistance to apoptosis and increased proliferation of bone marrow precursor B cells, accompanied by changes in several key modulators related to apoptosis, such as Fas/FasL signals, caspases-3/-8, nuclear factor-kappaB, and Bcl-2. We also show that the effects of AR loss are, in part, B cell intrinsic. Mice bearing AR-deficient B cells show increased levels of serum IgG2a and IgG3 as well as basal double-stranded DNA-IgG antibodies and are more vulnerable to development of collagen-induced arthritis. Together, these data indicate that androgen/AR play a crucial role in B cell homeostasis and tolerance. Therapies targeting AR might provide an alternative strategy with which to battle autoimmune diseases.


Assuntos
Apoptose/fisiologia , Autoimunidade/fisiologia , Linfócitos B/imunologia , Receptores Androgênicos/metabolismo , Transferência Adotiva , Animais , Artrite Experimental/imunologia , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/fisiologia , Linfócitos B/fisiologia , Feminino , Homeostase , Imunoglobulinas/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Fenótipo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Androgênicos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA