Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
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
2.
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
3.
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
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.
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
6.
Am J Pathol ; 181(5): 1504-12, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22959669

RESUMO

Upon insult, such as infection or tissue injury, the innate and adaptive immune systems initiate a series of responses to defend the body. Recent studies from immune cell-specific androgen receptor (AR) knockout mice demonstrated that androgen and its receptor (androgen/AR) play significant roles in both immune regulations. In the innate immunity, androgen/AR is required for generation and proper function of neutrophils; androgen/AR also regulates wound healing processes through macrophage recruitment and proinflammatory cytokine production. In adaptive immunity, androgen/AR exerts suppressive effects on development and activation of T and B cells. Removal of such suppression causes thymic enlargement and excessive export of immature B cells. Altogether, androgen/AR plays distinct roles in individual immune cells, and targeting androgen/AR may help in treatment and management of immune-related diseases.


Assuntos
Imunidade Adaptativa/imunologia , Imunidade Inata/imunologia , Receptores Androgênicos/deficiência , Animais , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Modelos Imunológicos , Receptores Androgênicos/metabolismo , Transdução de Sinais
7.
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
8.
Proc Natl Acad Sci U S A ; 105(34): 12188-93, 2008 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-18723670

RESUMO

To differentiate roles of androgen receptor (AR) in prostate stromal and epithelial cells, we have generated inducible-(ind)ARKO-TRAMP and prostate epithelial-specific ARKO TRAMP (pes-ARKO-TRAMP) mouse models, in which the AR was knocked down in both prostate epithelium and stroma or was knocked out in the prostate epithelium, respectively. We found that loss of AR in both mouse models resulted in poorly differentiated primary tumors with expanded intermediate cell populations. Interestingly, knockdown of both epithelial and stromal AR in ind-ARKO-TRAMP mice at earlier stages resulted in smaller primary prostate tumors with lower proliferation rates, and knockout of AR in pes-ARKO-TRAMP mice resulted in larger primary prostate tumors with higher proliferation rates. The differential proliferation rates, yet with similarly expanded intermediate cell populations, indicated that the prostate stromal AR might play a more dominant role than the epithelial AR to promote primary tumor proliferation at an early stage of tumor. Tissue recombination of human prostate stromal cell lines (WPMY1-v or WPMY1-ARsi) with human prostate cancer epithelial cell lines (PC3-v or PC3-AR9) further demonstrated that the AR might function as a suppressor in epithelial cells and a proliferator in stromal cells in the primary prostate tumors. The dual roles of the AR in prostate epithelium and stroma may require us to reevaluate the target and timing of androgen-deprivation therapy for prostate cancer patients and may suggest a need to develop new drugs to selectively target stromal AR in the primary prostate tumors at earlier stages.


Assuntos
Neoplasias da Próstata/patologia , Receptores Androgênicos/deficiência , Células Estromais/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Células Epiteliais/química , Células Epiteliais/patologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Neoplasias da Próstata/tratamento farmacológico , Receptores Androgênicos/genética , Células Estromais/química , Fatores de Tempo
9.
Proc Natl Acad Sci U S A ; 105(34): 12182-7, 2008 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-18723679

RESUMO

Targeting androgens/androgen receptor (AR) functions via androgen deprivation therapy (ADT) remains the standard treatment for prostate cancer. However, most tumors eventually recur despite ADT. Here we demonstrate that the prostate AR may function as both a suppressor and a proliferator to suppress or promote prostate cancer metastasis. Results from orthotopically recombining stromal WPMY1 cells with epithelial PC3 prostate cancer cells in mice demonstrated that restoring AR in epithelial PC3 cells or knockdown of AR in stromal WPMY1 cells suppressed prostate cancer metastasis. Knockdown of the AR in epithelial CWR22rv1 prostate cancer cells also resulted in increased cell invasion in vitro and in vivo. Restoring AR in PC3 cells (PC3-AR9) results in decreased invasion in bone lesion assays and in vivo mouse models. Mice lacking the prostate epithelial AR have increased apoptosis in epithelial luminal cells and increased proliferation in epithelial basal cells. The consequences of these two contrasting results led to the expansion of CK5/CK8-positive intermediate cells, and mice developed larger and more invasive metastatic tumors in lymph nodes and died earlier than wild-type littermates. Mechanistic dissection suggested that androgens/AR might directly or indirectly modulate metastasis-related genes and suppression of TGFbeta1 signals results in the partial inhibition of AR-mediated metastasis. Collectively, our understanding of these opposing roles of prostatic AR may revolutionize the way we combat prostate cancer, and allow the development of new and better therapies by targeting only the proliferative role of AR.


Assuntos
Proliferação de Células , Neoplasias da Próstata/patologia , Receptores Androgênicos/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Apoptose , Linhagem Celular Tumoral , Células Epiteliais , Masculino , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Células Estromais , Fator de Crescimento Transformador beta1/antagonistas & inibidores
10.
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
11.
Asian J Androl ; 11(1): 39-48, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19098932

RESUMO

The androgen receptor (AR) plays an important role in the development and progression of prostate cancer (PCa). Androgen deprivation therapy is initially effective in blocking tumor growth, but it eventually leads to the hormone-refractory state. The detailed mechanisms of the conversion from androgen dependence to androgen independence remain unclear. Several PCa cell lines were established to study the role of AR in PCa, but the results were often inconsistent or contrasting in different cell lines, or in the same cell line grown under different conditions. The cellular and molecular alteration of epithelial cells and their microenvironments are complicated, and it is difficult to use a single cell line to address this important issue and also to study the pathophysiological effects of AR. In this paper, we summarize the different effects of AR on multiple cell lines and show the disadvantages of using a single human PCa cell line to study AR effects on PCa. We also discuss the advantages of widely used epithelium-stroma co-culture systems, xenograft mouse models, and genetically engineered PCa mouse models. The combination of in vitro cell line studies and in vivo mouse models might lead to more credible results and better strategies for the study of AR roles in PCa.


Assuntos
Neoplasias da Próstata/patologia , Neoplasias da Próstata/fisiopatologia , Receptores Androgênicos/fisiologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Células Epiteliais/patologia , Humanos , Masculino , Camundongos , Células Estromais/patologia
12.
Cancer Res ; 65(11): 4827-35, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15930303

RESUMO

Experimental and clinical findings support the essential role of interleukin (IL)-6 in the pathogenesis of various human cancers and provide a rationale for targeted therapeutic investigations. A novel peptide, S7, which selectively binds to IL-6 receptor (IL-6R) alpha chain (gp80) and broadly inhibits IL-6-mediated events, was identified using phage display library screening. The synthetic S7 peptide specifically bound to soluble IL-6R as well as cognate human IL-6R alpha, resulting in a dose-dependent blockade of the interaction between IL-6 and IL-6R alpha. S7 peptide prevents IL-6-mediated survival signaling and sensitizes cervical cancer cells to chemotherapeutic compounds in vitro. The in vitro analysis of antiangiogenic activity showed that S7 peptide substantially inhibits IL-6-induced vascular endothelial growth factor-A expression and angiogenesis in different cancer cell lines. Furthermore, S7 peptide was bioavailable in vivo, leading to a significant suppression of IL-6-induced vascular endothelial growth factor-mediated cervical tumor growth in severe combined immunodeficient mice. These observations show the feasibility of targeting IL-6/IL-6R interaction using the small peptide and highlight its potential in the clinical applications.


Assuntos
Inibidores da Angiogênese/metabolismo , Inibidores da Angiogênese/farmacologia , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Receptores de Interleucina-6/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Células HT29 , Células HeLa , Humanos , Interleucina-6/antagonistas & inibidores , Interleucina-6/farmacologia , Camundongos , Camundongos SCID , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Biblioteca de Peptídeos , Ligação Proteica , Receptores de Interleucina-6/antagonistas & inibidores , Especificidade por Substrato , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Oncogene ; 24(3): 390-8, 2005 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-15531921

RESUMO

Arsenic trioxide (ATO) has been implicated as a promising anticancer agent by inhibiting cell growth and inducing apoptosis in certain types of cancer cells. This study explored the antimetastasis property of arsenic, drew potential link between arsenic use and radiotherapy, and uncovered the specific mechanisms underlying these remarkable responses. Using gelatin invasion assay and intravasation assay, we report the novel finding that low-dose ATO (1 muM) reduced the intrinsic migration ability of HeLa cells and significantly inhibited radiation-promoted tumor invasive potential of CaSki cells without inducing apoptotic cell death. Using the murine Lewis lung carcinoma model, the control animals and ATO treatment animals (1 mg/kg i.p., twice weekly) displayed similar in vivo growth kinetics, whereas the radiation (30 Gy in one fraction) and concurrent treatment groups showed considerable growth inhibition. Importantly, although concurrent treatment did not enhance the effectiveness of radiation therapy to the primary tumor, further examination of the lungs revealed that all animals succumbed to radiation-accelerated lung metastases could be effectively treated by combination of ATO and radiation. Radiation-induced matrix metalloproteinase-9 (MMP-9) expression was significantly inhibited by ATO using sequential analysis of the expression of MMPs in xenografts. Supporting this observation, ATO directly downregulates radiation-induced MMP-9 mRNA expression by inhibiting nuclear factor kappaB activity in human cervical cancer cells. In sum, concurrent arsenic-radiation therapy not only achieves local tumor control but also inhibits distant metastasis. Experimental results of this study highlight a novel strategy in cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Arsenicais/farmacologia , Inibidores Enzimáticos/farmacologia , Metaloproteinase 9 da Matriz/genética , NF-kappa B/metabolismo , Invasividade Neoplásica/prevenção & controle , Óxidos/farmacologia , Animais , Trióxido de Arsênio , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Raios gama , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Células HeLa , Humanos , Metaloproteinase 9 da Matriz/efeitos da radiação , Inibidores de Metaloproteinases de Matriz , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA