Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
EMBO Mol Med ; 5(9): 1383-401, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23982944

ABSTRACT

Increased CCL2 expression in prostate cancer (PCa) cells enhanced metastasis via macrophage recruitment. However, its linkage to androgen receptor (AR)-mediated PCa progression remains unclear. Here, we identified a previously unrecognized regulation: targeting AR with siRNA in PCa cells increased macrophage recruitment via CCL2 up-regulation, which might then result in enhancing PCa invasiveness. Molecular mechanism dissection revealed that targeting PCa AR with siRNA promoted PCa cell migration/invasion via CCL2-dependent STAT3 activation and epithelial-mesenchymal transition (EMT) pathways. Importantly, pharmacologic interruption of the CCL2/CCR2-STAT3 axis suppressed EMT and PCa cell migration, providing a new mechanism linking CCL2 and EMT. Simultaneously targeting PCa AR with siRNA and the CCL2/CCR2-STAT3 axis resulted in better suppression of PCa growth and metastasis in a xenograft PCa mouse model. Human PCa tissue microarray analysis suggests that increased CCL2 expression may be potentially associated with poor prognosis of PCa patients. Together, these results may provide a novel therapeutic approach to better battle PCa progression and metastasis at the castration resistant stage via the combination of targeting AR with siRNA and anti-CCL2/CCR2-STAT3 signalling.


Subject(s)
Chemokine CCL2/metabolism , Macrophages/physiology , Neoplasm Metastasis/pathology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Receptors, CCR2/metabolism , STAT3 Transcription Factor/metabolism , Animals , Heterografts , Humans , Male , Mice , Microarray Analysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Androgen/genetics
2.
Mol Endocrinol ; 27(10): 1617-31, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23893956

ABSTRACT

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.


Subject(s)
Fibroblasts/metabolism , Prostatic Hyperplasia/metabolism , Receptors, Androgen/genetics , Stromal Cells/metabolism , Androgen-Binding Protein/genetics , Animals , Cell Proliferation , Cells, Cultured , Curcumin/analogs & derivatives , Curcumin/pharmacology , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Organ Size/drug effects , Prolactin/physiology , Prostate/metabolism , Prostate/pathology , Prostatic Hyperplasia/drug therapy , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/physiopathology , Proteolysis/drug effects , Receptors, Androgen/metabolism , STAT3 Transcription Factor/metabolism
3.
Cancer Res ; 73(18): 5633-46, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23878190

ABSTRACT

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.


Subject(s)
Chemokine CCL4/metabolism , Macrophages/pathology , Prostate/pathology , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , STAT3 Transcription Factor/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Cell Transformation, Neoplastic , Cells, Cultured , Curcumin/analogs & derivatives , Curcumin/pharmacology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Epithelial-Mesenchymal Transition , Humans , Immunoenzyme Techniques , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Knockout , PTEN Phosphohydrolase/physiology , Prostate/immunology , Prostate/metabolism , Prostatic Intraepithelial Neoplasia/immunology , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Signal Transduction
4.
Diabetes ; 62(2): 411-23, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23139353

ABSTRACT

Clinical investigations highlight the increased incidence of metabolic syndrome in prostate cancer (PCa) patients receiving androgen deprivation therapy (ADT). Studies using global androgen receptor (AR) knockout mice demonstrate that AR deficiency results in the development of insulin resistance in males. However, mechanisms by which AR in individual organs coordinately regulates insulin sensitivity remain unexplored. Here we tested the hypothesis that functional AR in the brain contributes to whole-body insulin sensitivity regulation and to the metabolic abnormalities developed in AR-deficient male mice. The mouse model selectively lacking AR in the central nervous system and AR-expressing GT1-7 neuronal cells were established and used to delineate molecular mechanisms in insulin signaling modulated by AR. Neuronal AR deficiency leads to reduced insulin sensitivity in middle-aged mice. Neuronal AR regulates hypothalamic insulin signaling by repressing nuclear factor-κB (NF-κB)-mediated induction of protein-tyrosine phosphatase 1B (PTP1B). Hypothalamic insulin resistance leads to hepatic insulin resistance, lipid accumulation, and visceral obesity. The functional deficiency of AR in the hypothalamus leads to male mice being more susceptible to the effects of high-fat diet consumption on PTP1B expression and NF-κB activation. These findings suggest that in men with PCa undergoing ADT, reduction of AR function in the brain may contribute to insulin resistance and visceral obesity. Pharmacotherapies targeting neuronal AR and NF-κB may be developed to combat the metabolic syndrome in men receiving ADT and in elderly men with age-associated hypogonadism.


Subject(s)
Gene Expression Regulation/physiology , Hypothalamus/metabolism , Insulin Resistance/physiology , NF-kappa B/metabolism , Neurons/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/biosynthesis , Receptors, Androgen/metabolism , Animals , Brain/metabolism , Cell Line , Diet, High-Fat , Insulin/blood , Insulin/metabolism , Insulin Resistance/genetics , Leptin/blood , Leptin/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity, Abdominal/genetics , Obesity, Abdominal/metabolism , Receptors, Androgen/genetics , Signal Transduction/genetics , Signal Transduction/physiology
5.
Mol Endocrinol ; 26(10): 1707-15, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22915828

ABSTRACT

Early studies suggested macrophages might play roles in inflammation-associated benign prostatic hyperplasia (BPH) development, yet the underlying mechanisms remain unclear. Here we first showed that CD68(+) macrophages were identified in both epithelium and the stromal area of human BPH tissues. We then established an in vitro co-culture model with prostate epithelial and macrophage cell lines to study the potential impacts of infiltrating macrophages in the BPH development and found that co-culturing prostate epithelial cells with macrophages promoted migration of macrophages. In a three-dimensional culture system, the sphere diameter of BPH-1 prostate cells was significantly increased during coculture with THP-1 macrophage cells. Mechanism dissection suggested that expression levels of epithelial-mesenchymal transition (EMT) markers, such as N-cadherin, Snail, and TGF-ß2, were increased, and administration of anti-TGF-ß2 neutralizing antibody during co-culture suppressed the EMT and THP-1-mediated growth of BPH-1 cells, suggesting THP-1 might go through EMT to influence the BPH development and progression. Importantly, we found that modulation of androgen receptor (AR) in BPH-1 and mPrE cells significantly increased THP-1 and RAW264.7 cell migration, respectively, and enhanced expression levels of EMT markers, suggesting that AR in prostate epithelial cells might play a role in promoting macrophage-mediated EMT in prostate epithelial cells. Silencing AR function via an AR degradation enhancer, ASC-J9, decreased the macrophage migration to BPH-1 cells and suppressed EMT marker expression. Together, these results provide the first evidence to demonstrate that prostate epithelial AR function is important for macrophage-mediated EMT and proliferation of prostate epithelial cells, which represents a previously unrecognized role of AR in the cross-talk between macrophages and prostate epithelial cells. These results may provide new insights for a new therapeutic approach to battle BPH via targeting AR and AR-mediated inflammatory signaling pathways.


Subject(s)
Epithelial-Mesenchymal Transition , Macrophages/physiology , Prostatic Hyperplasia/metabolism , Receptors, Androgen/physiology , Androgen Receptor Antagonists/pharmacology , Animals , Cell Line , Cell Movement , Coculture Techniques , Curcumin/analogs & derivatives , Curcumin/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/physiology , Gene Expression , Humans , Macrophages/pathology , Male , Mice , Molecular Targeted Therapy , Prostate/pathology , Prostatic Hyperplasia/pathology , Proteolysis , Receptors, Androgen/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transforming Growth Factor beta2
6.
J Biol Chem ; 287(22): 18376-85, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22474290

ABSTRACT

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.


Subject(s)
Macrophages/pathology , Prostate/pathology , Prostatic Hyperplasia/pathology , Receptors, Androgen/physiology , Stromal Cells/pathology , Animals , Base Sequence , Blotting, Western , DNA Primers , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Male , Mice , Prostatic Hyperplasia/physiopathology , Reverse Transcriptase Polymerase Chain Reaction
7.
J Oral Pathol Med ; 39(4): 342-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20040021

ABSTRACT

BACKGROUND: Oral squamous cell carcinoma (OSCC) is one of the most malignant neoplasms worldwide, and the molecular mechanism of oral tumorigenesis is still unclear. Fatty-acid-binding protein 5 (FABP5) was found in our previous study to be upregulated in oral squamous cell carcinomas by proteomic analysis. The implications of FABP5 overexpression in oral cancer progression have not yet been elucidated. MATERIALS AND METHODS: In this study, the recombinant adeno-associated virus vectors were used to deliver and increase the expression of FABP5 in human OSCC cell lines. U6 promoter-driven short-hairpin RNA (shRNA)-triggered RNA interference was used to block FABP5 gene expression. Transwell Matrigel invasion assay, MTS cell proliferation assay, reverse transcription-polymerase chain reaction, Western blot, and gelatin zymography analysis were used to investigate the effects of FABP5 on cell invasion, growth, and matrix metalloproteinase (MMP) production. RESULTS: Overexpression of FABP5 in oral cancer cells increased cell proliferation and invasiveness by increasing the expression of MMP-9. Silencing FABP5 with shRNA significantly suppressed cell proliferation, MMP-9 activities, and invasiveness. CONCLUSION: Our study provides the first evidence that FABP5 expression modulated MMP-9 production and the invasive behavior of oral cancer cells and suggests that FABP5 may provide novel targets for therapeutic intervention.


Subject(s)
Carcinoma, Squamous Cell/pathology , Fatty Acid-Binding Proteins/physiology , Mouth Neoplasms/pathology , Adenoviridae/genetics , Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/secondary , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Epithelium/pathology , Fatty Acid-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Genetic Vectors/genetics , Humans , Inverted Repeat Sequences/genetics , Keratinocytes/pathology , Lymphatic Metastasis/pathology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mouth Mucosa/pathology , Mouth Neoplasms/genetics , Neoplasm Invasiveness/genetics , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Nuclear/genetics
8.
Proc Natl Acad Sci U S A ; 106(32): 13353-8, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19666541

ABSTRACT

Testicular orphan nuclear receptor 4 (TR4) is an orphan member of the nuclear receptor superfamily with diverse physiological functions. Using TR4 knockout (TR4(-/-)) mice to study its function in cardiovascular diseases, we found reduced cluster of differentiation (CD)36 expression with reduced foam cell formation in TR4(-/-) mice. Mechanistic dissection suggests that TR4 induces CD36 protein and mRNA expression via a transcriptional regulation. Interestingly, we found this TR4-mediated CD36 transactivation can be further enhanced by polyunsaturated fatty acids (PUFAs), such as omega-3 and -6 fatty acids, and their metabolites such as 15-hydroxyeico-satetraonic acid (15-HETE) and 13-hydroxy octa-deca dieonic acid (13-HODE) and thiazolidinedione (TZD)-rosiglitazone. Both electrophoretic mobility shift assays (EMSA) and chromatin immunoprecipitation (ChIP) assays demonstrate that TR4 binds to the TR4 response element located on the CD36 5'-promoter region for the induction of CD36 expression. Stably transfected TR4-siRNA or functional TR4 cDNA in the RAW264.7 macrophage cells resulted in either decreased or increased CD36 expression with decreased or increased foam cell formation. Restoring functional CD36 cDNA in the TR4 knockdown macrophage cells reversed the decreased foam cell formation. Together, these results reveal an important signaling pathway controlling CD36-mediated foam cell formation/cardiovascular diseases, and findings that TR4 transactivation can be activated via its ligands/activators, such as PUFA metabolites and TZD, may provide a platform to screen new drug(s) to battle the metabolism syndrome, diabetes, and cardiovascular diseases.


Subject(s)
CD36 Antigens/metabolism , Cell Nucleus/metabolism , Fatty Acids, Unsaturated/metabolism , Foam Cells/cytology , Foam Cells/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Animals , Base Sequence , CD36 Antigens/genetics , Cell Nucleus/drug effects , Foam Cells/drug effects , Ligands , Mice , Molecular Sequence Data , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/genetics , Rosiglitazone , Thiazolidinediones/pharmacology , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...