Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(14): eadc9446, 2023 04 05.
Article in English | MEDLINE | ID: mdl-37018402

ABSTRACT

The mechanisms underlying ETS-driven prostate cancer initiation and progression remain poorly understood due to a lack of model systems that recapitulate this phenotype. We generated a genetically engineered mouse with prostate-specific expression of the ETS factor, ETV4, at lower and higher protein dosage through mutation of its degron. Lower-level expression of ETV4 caused mild luminal cell expansion without histologic abnormalities, and higher-level expression of stabilized ETV4 caused prostatic intraepithelial neoplasia (mPIN) with 100% penetrance within 1 week. Tumor progression was limited by p53-mediated senescence and Trp53 deletion cooperated with stabilized ETV4. The neoplastic cells expressed differentiation markers such as Nkx3.1 recapitulating luminal gene expression features of untreated human prostate cancer. Single-cell and bulk RNA sequencing showed that stabilized ETV4 induced a previously unidentified luminal-derived expression cluster with signatures of cell cycle, senescence, and epithelial-to-mesenchymal transition. These data suggest that ETS overexpression alone, at sufficient dosage, can initiate prostate neoplasia.


Subject(s)
Prostatic Intraepithelial Neoplasia , Prostatic Neoplasms , Male , Mice , Animals , Humans , Prostate/metabolism , Prostate/pathology , Tumor Suppressor Protein p53/metabolism , Prostatic Neoplasms/genetics , Transcription Factors/metabolism , Prostatic Intraepithelial Neoplasia/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-ets/genetics
2.
Clin Cancer Res ; 28(21): 4600-4602, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35984355

ABSTRACT

Through analysis of the cancer dependency map of CRISPR and short hairpin RNA datasets, the antiapoptotic BCL-XL was found to be a selective dependency in kidney cancer. Among kidney cancers, BCL-XL inhibition is most active in those with a mesenchymal gene signature, which portends a poor prognosis and response to current therapies. See related article by Grubb et al., p. 4689.


Subject(s)
Apoptosis , Kidney Neoplasms , Humans , bcl-X Protein/genetics , bcl-X Protein/metabolism , Apoptosis/genetics , RNA, Small Interfering/genetics , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics
3.
Cancer Discov ; 12(9): 2120-2139, 2022 09 02.
Article in English | MEDLINE | ID: mdl-35789380

ABSTRACT

Polycomb repressive complex 2 (PRC2) has oncogenic and tumor-suppressive roles in cancer. There is clinical success of targeting this complex in PRC2-dependent cancers, but an unmet therapeutic need exists in PRC2-loss cancer. PRC2-inactivating mutations are a hallmark feature of high-grade malignant peripheral nerve sheath tumor (MPNST), an aggressive sarcoma with poor prognosis and no effective targeted therapy. Through RNAi screening in MPNST, we found that PRC2 inactivation increases sensitivity to genetic or small-molecule inhibition of DNA methyltransferase 1 (DNMT1), which results in enhanced cytotoxicity and antitumor response. Mechanistically, PRC2 inactivation amplifies DNMT inhibitor-mediated expression of retrotransposons, subsequent viral mimicry response, and robust cell death in part through a protein kinase R (PKR)-dependent double-stranded RNA sensor. Collectively, our observations posit DNA methylation as a safeguard against antitumorigenic cell-fate decisions in PRC2-loss cancer to promote cancer pathogenesis, which can be therapeutically exploited by DNMT1-targeted therapy. SIGNIFICANCE: PRC2 inactivation drives oncogenesis in various cancers, but therapeutically targeting PRC2 loss has remained challenging. Here we show that PRC2-inactivating mutations set up a tumor context-specific liability for therapeutic intervention via DNMT1 inhibitors, which leads to innate immune signaling mediated by sensing of derepressed retrotransposons and accompanied by enhanced cytotoxicity. See related commentary by Guil and Esteller, p. 2020. This article is highlighted in the In This Issue feature, p. 2007.


Subject(s)
Antineoplastic Agents , Neoplasms , Neurofibrosarcoma , Carcinogenesis/genetics , Humans , Mutation , Neoplasms/genetics , Neurofibrosarcoma/diagnosis , Neurofibrosarcoma/genetics , Neurofibrosarcoma/pathology , Polycomb Repressive Complex 2/genetics , Retroelements
4.
Clin Cancer Res ; 27(5): 1476-1490, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33229459

ABSTRACT

PURPOSE: All uveal melanoma and a fraction of other melanoma subtypes are driven by activation of the G-protein alpha-q (Gαq) pathway. Targeting these melanomas has proven difficult despite advances in the molecular understanding of key driver signaling pathways in the disease pathogenesis. Inhibitors of Gαq have shown promising preclinical results, but their therapeutic activity in distinct Gαq mutational contexts and in vivo have remained elusive. EXPERIMENTAL DESIGN: We used an isogenic melanocytic cellular system to systematically examine hotspot mutations in GNAQ (e.g., G48V, R183Q, Q209L) and CYSLTR2 (L129Q) found in human uveal melanoma. This cellular system and human uveal melanoma cell lines were used in vitro and in in vivo xenograft studies to assess the efficacy of Gαq inhibition as a single agent and in combination with MEK inhibition. RESULTS: We demonstrate that the Gαq inhibitor YM-254890 inhibited downstream signaling and in vitro growth in all mutants. In vivo, YM-254890 slowed tumor growth but did not cause regression in human uveal melanoma xenografts. Through comprehensive transcriptome analysis, we observed that YM-254890 caused inhibition of the MAPK signaling with evidence of rebound by 24 hours and combination treatment of YM-254890 and a MEK inhibitor led to sustained MAPK inhibition. We further demonstrated that the combination caused synergistic growth inhibition in vitro and tumor shrinkage in vivo. CONCLUSIONS: These data suggest that the combination of Gαq and MEK inhibition provides a promising therapeutic strategy and improved therapeutic window of broadly targeting Gαq in uveal melanoma.See related commentary by Neelature Sriramareddy and Smalley, p. 1217.


Subject(s)
Melanoma , Uveal Neoplasms , Cell Line, Tumor , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Humans , Melanoma/drug therapy , Melanoma/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Uveal Neoplasms/drug therapy , Uveal Neoplasms/genetics
5.
Biochem Biophys Res Commun ; 527(1): 242-249, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32446375

ABSTRACT

Intratumoral heterogeneity remains as a major challenge in the treatment resistance of prostate cancer. Understanding the mechanism of prostate cancer heterogeneity is essential for developing effective therapies. In this study, we reported the heterogeneous activation of Wnt/ß-catenin signaling in prostate cancer. We developed a Wnt/ß-catenin signaling reporting system to directly characterize the differences between Wnt/ß-catenin signaling active (GFP+) and inactive (GFP-) cells. Compared to GFP- cells, GFP+ cells demonstrated cancer stem cell properties with higher colony formation efficiency, slower cell cycle, higher resistance to docetaxel and higher expression of cancer stem cell markers. In addition, we found that Wnt/ß-catenin signaling is negatively correlated with H3K27me3 levels. Further studies demonstrated that Wnt/ß-catenin signaling affected H3K27me3 levels by regulating the expression of KDM6A, one of the H3K27me3 demethylases. H3K27me3 suppressed Wnt/ß-catenin signaling by directly silencing LEF1 promoter. Together, our studies suggest that Wnt/ß-catenin signaling makes a major contribution to prostate cancer heterogeneity and targeting both Wnt/ß-catenin signaling active and inactive populations is essential for developing more effective therapies.


Subject(s)
Prostatic Neoplasms/metabolism , Wnt Signaling Pathway , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Docetaxel/pharmacology , Dose-Response Relationship, Drug , Humans , Male , Methylation/drug effects , PC-3 Cells , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Structure-Activity Relationship , Wnt Signaling Pathway/drug effects
6.
Stem Cell Reports ; 9(1): 231-246, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28669599

ABSTRACT

Although STAT3 signaling is demonstrated to regulate sensory cell differentiation and regeneration in the zebrafish, its exact role is still unclear in mammalian cochleae. Here, we report that STAT3 and its activated form are specifically expressed in hair cells during mouse cochlear development. Importantly, conditional cochlear deletion of Stat3 leads to an inhibition on hair cell differentiation in mice in vivo and in vitro. By cell fate analysis, inactivation of STAT3 signaling shifts the cell division modes from asymmetric to symmetric divisions from supporting cells. Moreover, inhibition of Notch signaling stimulates STAT3 phosphorylation, and inactivation of STAT3 signaling attenuates production of supernumerary hair cells induced by a Notch pathway inhibitor. Our findings highlight an important role of the STAT3 signaling during mouse cochlear hair cell differentiation and may have clinical implications for the recovery of hair cell loss-induced hearing impairment.


Subject(s)
Cell Differentiation , Cochlea/growth & development , Hair Cells, Auditory/cytology , STAT3 Transcription Factor/metabolism , Signal Transduction , Animals , Cell Division , Cells, Cultured , Cochlea/cytology , Cochlea/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Mice , Receptors, Notch/metabolism , STAT3 Transcription Factor/genetics
7.
Stem Cell Reports ; 6(5): 668-678, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27167157

ABSTRACT

Regulation of prostate epithelial progenitor cells is important in prostate development and prostate diseases. Our previous study demonstrated a function of autocrine cholinergic signaling (ACS) in promoting prostate cancer growth and castration resistance. However, whether or not such ACS also plays a role in prostate development is unknown. Here, we report that ACS promoted the proliferation and inhibited the differentiation of prostate epithelial progenitor cells in organotypic cultures. These results were confirmed by ex vivo lineage tracing assays and in vivo renal capsule recombination assays. Moreover, we found that M3 cholinergic receptor (CHRM3) was upregulated in a large subset of benign prostatic hyperplasia (BPH) tissues compared with normal tissues. Activation of CHRM3 also promoted the proliferation of BPH cells. Together, our findings identify a role of ACS in maintaining prostate epithelial progenitor cells in the proliferating state, and blockade of ACS may have clinical implications for the management of BPH.


Subject(s)
Epithelial Cells/metabolism , Prostatic Hyperplasia/genetics , Receptor, Muscarinic M3/genetics , Stem Cells/metabolism , Animals , Autocrine Communication/genetics , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation/genetics , Epithelial Cells/pathology , Humans , Male , Mice , Prostate/metabolism , Prostate/pathology , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Stem Cells/pathology
8.
Oncotarget ; 6(28): 26494-507, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26317998

ABSTRACT

SIRT3 is involved in aging-related diseases including cancer, but its role in prostate cancer and detailed regulatory function are not known. We found that SIRT3 was moderately down-regulated in prostate carcinomas. Overexpression of SIRT3 by lentiviral transfection inhibited prostate cancer growth both in vitro and in vivo, whereas knockdown of SIRT3 increased prostate tumor growth. Mechanistically, the tumor suppression effect of SIRT3 was achieved via its inhibition of the PI3K/Akt pathway. Notably, upregulation of SIRT3 suppressed the phosphorylation of Akt, leading to the ubiquitination and degradation of oncoprotein c-MYC; this could be attenuated by constitutive activation of PI3K/Akt signaling. Collectively, our results unveiled SIRT3's tumor suppressive function and the underlying mechanism in prostate cancer, which might provide therapeutic implications for the disease.


Subject(s)
Phosphatidylinositol 3-Kinase/metabolism , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Sirtuin 3/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Male , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Protein Stability , Proteolysis , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , Signal Transduction , Sirtuin 3/genetics , Time Factors , Transfection , Ubiquitination
9.
Clin Cancer Res ; 21(20): 4676-85, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26071486

ABSTRACT

PURPOSE: Although a previous study reported nerve ending-derived acetylcholine promoted prostate cancer invasion and metastasis by regulating the microenvironment of cancer cells, the present study aims to determine whether there is autocrine cholinergic signaling in prostate epithelial cells that promotes prostate cancer growth and castration resistance. EXPERIMENTAL DESIGN: In this study, IHC was performed to detect protein expression in mouse prostate tissue sections and human prostate cancer tissue sections. Subcutaneously and orthotopically xenografted tumor models were established to evaluate the functions of autocrine cholinergic signaling in regulating prostate cancer growth and castration resistance. Western blotting analysis was performed to assess the autocrine cholinergic signaling-induced signaling pathway. RESULTS: We found the expression of choline acetyltransferase (ChAT), the secretion of acetylcholine and the expression of CHRM3 in prostate epithelial cells, supporting the presence of autocrine cholinergic signaling in the prostate epithelium. In addition, we found that CHRM3 was upregulated in clinical prostate cancer tissues compared with adjacent non-cancer tissues. Overexpression of CHRM3 or activation of CHRM3 by carbachol promoted cell proliferation, migration, and castration resistance. On the contrary, blockading CHRM3 by shRNA or treatment with darifenacin inhibited prostate cancer growth and castration resistance both in vitro and in vivo. Furthermore, we found that autocrine cholinergic signaling caused calmodulin/calmodulin-dependent protein kinase kinase (CaM/CaMKK)-mediated phosphorylation of Akt. CONCLUSIONS: These findings suggest that blockade of CHRM3 may represent a novel adjuvant therapy for castration-resistant prostate cancer.


Subject(s)
Autocrine Communication/physiology , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Phosphorylation/physiology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Muscarinic/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostate/metabolism , Prostate/pathology , Receptor, Muscarinic M3 , Signal Transduction/physiology , Up-Regulation/genetics
10.
PLoS One ; 10(4): e0116197, 2015.
Article in English | MEDLINE | ID: mdl-25860954

ABSTRACT

Prostate cancer (PCa) is the most frequently diagnosed cancer for men in the developed world. Androgen receptor signaling pathway plays an important role in prostate cancer progression. Recent studies show that microRNA miR-124 exerts a tumor suppressive function in prostate cancer. However, the relationship between AR and miR-124 is unclear. In the present study, we found a negative feedback loop between AR and miR-124 expression. On one hand, miR-124 was a positively regulated target gene of the AR, on the other hand, overexpression of miR-124 inhibited the expression of AR. In addition, we found that miR-124-2 and miR-124-3 promoters were hypermethylated in AR-negative PCa cells. Furthermore, overexpression of miR-124 inhibited proliferation rates and invasiveness capacity of PCa cells in vitro, and suppressed xenograft tumor growth in vivo. Taken together, our results support a negative feedback loop between AR and miR-124 expression. Methylation of miR-124-2 and miR-124-3 may serve as a biomarker for AR-negative PCa cells, and overexpression of miR-124 might be of potential therapeutic value for the treatment of PCa.


Subject(s)
MicroRNAs/genetics , MicroRNAs/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , CpG Islands , DNA Methylation , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/therapy , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
11.
Exp Biol Med (Maywood) ; 239(7): 823-828, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24764242

ABSTRACT

Prostate cancer is the most common type of cancer for men in the developed world. Androgen receptor (AR) is very important in prostate cancer progression. TMPRSS2 is an AR signaling downstream gene and closely related to prostate carcinogenesis. DNA methylation is a key mechanism to influence gene expression. Though previous reports have shown that AR signaling plays a critical role in the regulation of TMPRSS2 in prostate cancer, hardly any studies have examined whether the DNA methylation has been involved in the regulation of TMPRSS2. In the present study, we demonstrated that AR-negative prostate cancer (PCa) cells showed low expression levels and hypermethylation of TMPRSS2. In contrast, AR-positive PCa cells displayed high levels and hypomethylation of TMPRSS2. Treatment with the DNA methylation inhibitor 5-Aza-2'-deoxycytidine reversed the low expression levels of TMPRSS2 in the AR-negative PCa cells. Additionally, we found that the level of DNA methyltransferases 1 (DNMT1) was high in AR-negative PCa cells, in which hypermethylation of TMPRSS2 and low expression level of TMPRSS2 were observed. Collectively, these data suggest that the high level of DNMT1 might be the mechanism for the hypermethylation-mediated transcriptional repression of TMPRSS2 in AR-negative PCa cells.

SELECTION OF CITATIONS
SEARCH DETAIL