Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Mol Oncol ; 16(22): 3994-4010, 2022 12.
Article in English | MEDLINE | ID: mdl-36087093

ABSTRACT

Loss of the histone demethylase KDM5D (lysine-specific demethylase 5D) leads to in vitro resistance of prostate cancer cells to androgen deprivation therapy (ADT) with and without docetaxel. We aimed to define downstream drivers of the KDM5D effect. Using chromatin immunoprecipitation sequencing (ChIP-seq) of the LNCaP cell line (androgen-sensitive human prostate adenocarcinoma) with and without silenced KDM5D, MYBL2-binding sites were analyzed. Associations between MYBL2 mRNA expression and clinical outcomes were assessed in cohorts of men with localized and metastatic hormone-sensitive prostate cancer. In vitro assays with silencing and overexpression of MYBL2 and KDM5D in androgen receptor (AR)-positive hormone-sensitive prostate cancer cell lines, LNCaP and LAPC4, were used to assess their influence on cellular proliferation, apoptosis, and cell cycle distribution, as well as sensitivity to androgen deprivation, docetaxel, and cabazitaxel. We found that silencing KDM5D increased histone H3 lysine K4 (H3K4) trimethylation and increased MYBL2 expression. KDM5D and MYBL2 were negatively correlated with some but not all clinical samples. Higher MYBL2 expression was associated with a higher rate of relapse in localized disease and poorer overall survival in men with metastatic disease in the CHAARTED trial. Lower MYBL2 levels enhanced LNCaP and LAPC4 sensitivity to androgen deprivation and taxanes. In vitro, modifications of KDM5D and MYBL2 altered cell cycle distribution and apoptosis in a cell line-specific manner. These results show that the transcription factor MYBL2 impacts in vitro hormone-sensitive prostate cancer sensitivity to androgen deprivation and taxanes, and lower levels are associated with better clinical outcomes in men with hormone-sensitive prostate cancer.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Docetaxel/pharmacology , Androgen Antagonists/therapeutic use , Prostatic Neoplasms, Castration-Resistant/metabolism , Androgens , Lysine , Taxoids/therapeutic use , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/therapeutic use , Histone Demethylases , Trans-Activators , Cell Cycle Proteins
2.
Clin Cancer Res ; 28(16): 3603-3617, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35670774

ABSTRACT

PURPOSE: Oncogenic alterations of the PI3K/AKT pathway occur in >40% of patients with metastatic castration-resistant prostate cancer, predominantly via PTEN loss. The significance of other PI3K pathway components in prostate cancer is largely unknown. EXPERIMENTAL DESIGN: Patients in this study underwent tumor sequencing using the MSK-IMPACT clinical assay to capture single-nucleotide variants, insertions, and deletions; copy-number alterations; and structural rearrangements, or were profiled through The Cancer Genome Atlas. The association between PIK3R1 alteration/expression and survival was evaluated using univariable and multivariable Cox proportional-hazards regression models. We used the siRNA-based knockdown of PIK3R1 for functional studies. FDG-PET/CT examinations were performed with a hybrid positron emission tomography (PET)/CT scanner for some prostate cancer patients in the MSK-IMPACT cohort. RESULTS: Analyzing 1,417 human prostate cancers, we found a significant enrichment of PIK3R1 alterations in metastatic cancers compared with primary cancers. PIK3R1 alterations or reduced mRNA expression tended to be associated with worse clinical outcomes in prostate cancer, particularly in primary disease, as well as in breast, gastric, and several other cancers. In prostate cancer cell lines, PIK3R1 knockdown resulted in increased cell proliferation and AKT activity, including insulin-stimulated AKT activity. In cell lines and organoids, PIK3R1 loss/mutation was associated with increased sensitivity to AKT inhibitors. PIK3R1-altered patient prostate tumors had increased uptake of the glucose analogue 18F-fluorodeoxyglucose in PET imaging, suggesting increased glycolysis. CONCLUSIONS: Our findings describe a novel genomic feature in metastatic prostate cancer and suggest that PIK3R1 alteration may be a key event for insulin-PI3K-glycolytic pathway regulation in prostate cancer.


Subject(s)
Phosphatidylinositol 3-Kinases , Prostatic Neoplasms , Class Ia Phosphatidylinositol 3-Kinase/genetics , Glycolysis , Humans , Insulin/genetics , Insulin/metabolism , Male , Mutation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
3.
Mol Oncol ; 16(13): 2451-2469, 2022 07.
Article in English | MEDLINE | ID: mdl-34792282

ABSTRACT

Prostate cancer is a highly heterogeneous disease, understanding the crosstalk between complex genomic and epigenomic alterations will aid in developing targeted therapeutics. We demonstrate that, even though snail family transcriptional repressor 2 (SNAI2) is frequently amplified in prostate cancer, it is epigenetically silenced in this disease, with dynamic changes in SNAI2 levels showing distinct clinical relevance. Integrative clinical data from 18 prostate cancer cohorts and experimental evidence showed that gene fusion between transmembrane serine protease 2 (TMPRSS2) and ETS transcription factor ERG (ERG) (TMPRSS2-ERG fusion) is involved in the silencing of SNAI2. We created a silencer score to evaluate epigenetic repression of SNAI2, which can be reversed by treatment with DNA methyltransferase inhibitors and histone deacetylase inhibitors. Silencing of SNAI2 facilitated tumor cell proliferation and luminal differentiation. Furthermore, SNAI2 has a major influence on the tumor microenvironment by reactivating tumor stroma and creating an immunosuppressive microenvironment in prostate cancer. Importantly, SNAI2 expression levels in part determine sensitivity to the cancer drugs dasatinib and panobinostat. For the first time, we defined the distinct clinical relevance of SNAI2 expression at different disease stages. We elucidated how epigenetic silencing of SNAI2 controls the dynamic changes of SNAI2 expression that are essential for tumor initiation and progression and discovered that restoring SNAI2 expression by treatment with panobinostat enhances dasatinib sensitivity, indicating a new therapeutic strategy for prostate cancer.


Subject(s)
Oncogene Proteins, Fusion , Prostatic Neoplasms , Snail Family Transcription Factors , Cell Line, Tumor , Dasatinib/therapeutic use , Humans , Male , Oncogene Proteins, Fusion/genetics , Panobinostat/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Snail Family Transcription Factors/genetics , Tumor Microenvironment
4.
Oncogene ; 41(5): 671-682, 2022 01.
Article in English | MEDLINE | ID: mdl-34802033

ABSTRACT

Chromosome 8q gain is associated with poor clinical outcomes in prostate cancer, but the underlying biological mechanisms remain to be clarified. CSN5, a putative androgen receptor (AR) partner that is located on chromosome 8q, is the key subunit of the COP9 signalosome, which deactivates ubiquitin ligases. Deregulation of CSN5 could affect diverse cellular functions that contribute to tumor development, but there has been no comprehensive study of its function in prostate cancer. The clinical significance of CSN5 amplification/overexpression was evaluated in 16 prostate cancer clinical cohorts. Its oncogenic activity was assessed by genetic and pharmacologic perturbations of CSN5 activity in prostate cancer cell lines. The molecular mechanisms of CSN5 function were assessed, as was the efficacy of the CSN5 inhibitor CSN5i-3 in vitro and in vivo. Finally, the transcription cofactor activity of CSN5 in prostate cancer cells was determined. The prognostic significance of CSN5 amplification and overexpression in prostate cancer was independent of MYC amplification. Inhibition of CSN5 inhibited its oncogenic function by targeting AR signaling, DNA repair, multiple oncogenic pathways, and spliceosome regulation. Furthermore, inhibition of CSN5 repressed metabolic pathways, including oxidative phosphorylation and glycolysis in AR-negative prostate cancer cells. Targeting CSN5 with CSN5i-3 showed potent antitumor activity in vitro and in vivo. Importantly, CSN5i-3 synergizes with PARP inhibitors to inhibit prostate cancer cell growth. CSN5 functions as a transcription cofactor to cooperate with multiple transcription factors in prostate cancer. Inhibiting CSN5 strongly attenuates prostate cancer progression and could enhance PARP inhibition efficacy in the treatment of prostate cancer.


Subject(s)
COP9 Signalosome Complex
5.
Clin Cancer Res ; 27(6): 1792-1806, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33334906

ABSTRACT

PURPOSE: Alterations in DNA damage repair (DDR) pathway genes occur in 20%-25% of men with metastatic castration-resistant prostate cancer (mCRPC). Although PARP inhibitors (PARPis) have been shown to benefit men with mCRPC harboring DDR defects due to mutations in BRCA1/2 and ATM, additional treatments are necessary because the effects are not durable. EXPERIMENTAL DESIGN: We performed transcriptomic analysis of publicly available mCRPC cases, comparing BRCA2 null with BRCA2 wild-type. We generated BRCA2-null prostate cancer cells using CRISPR/Cas9 and treated these cells with PARPis and SRC inhibitors. We also assessed the antiproliferative effects of combination treatment in 3D prostate cancer organoids. RESULTS: We observed significant enrichment of the SRC signaling pathway in BRCA2-altered mCRPC. BRCA2-null prostate cancer cell lines had increased SRC phosphorylation and higher sensitivity to SRC inhibitors (e.g., dasatinib, bosutinib, and saracatinib) relative to wild-type cells. Combination treatment with PARPis and SRC inhibitors was antiproliferative and had a synergistic effect in BRCA2-null prostate cancer cells, mCRPC organoids, and Trp53/Rb1-null prostate cancer cells. Inhibition of SRC signaling by dasatinib augmented DNA damage in BRCA2-null prostate cancer cells. Moreover, SRC knockdown increased PARPi sensitivity in BRCA2-null prostate cancer cells. CONCLUSIONS: This work suggests that SRC activation may be a potential mechanism of PARPi resistance and that treatment with SRC inhibitors may overcome this resistance. Our preclinical study demonstrates that combining PARPis and SRC inhibitors may be a promising therapeutic strategy for patients with BRCA2-null mCRPC.


Subject(s)
Antineoplastic Agents/pharmacology , BRCA2 Protein/genetics , Gene Expression Regulation, Neoplastic/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Synthetic Lethal Mutations , src-Family Kinases/antagonists & inhibitors , Animals , Apoptosis , Cell Proliferation , Drug Synergism , Drug Therapy, Combination , Humans , Male , Mice , Mice, Nude , Prognosis , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Mol Oncol ; 14(8): 1881-1897, 2020 08.
Article in English | MEDLINE | ID: mdl-32385899

ABSTRACT

Although there are molecularly distinct subtypes of prostate cancer, no molecular classification system is used clinically. The ribonucleotide reductase small subunit M2 (RRM2) gene plays an oncogenic role in many cancers. Our previous study elucidated comprehensive molecular mechanisms of RRM2 in prostate cancer (PC). Given the potent functions of RRM2, we set out to determine whether the RRM2 signature can be used to identify aggressive subtypes of PC. We applied gene ontology and pathway analysis in RNA-seq datasets from PC cells overexpressing RRM2. We refined the RRM2 signature by integrating it with two molecular classification systems (PCS and PAM50 subtypes) that define aggressive PC subtypes (PCS1 and luminal B) and correlated signatures with clinical outcomes in six published cohorts comprising 4000 cases of PC. Increased expression of genes in the RRM2 signature was significantly correlated with recurrence, high Gleason score, and lethality of PC. Patients with high RRM2 levels showed higher PCS1 score, suggesting the aggressive PC feature. Consistently, RRM2-regulated genes were highly enriched in the PCS1 signature from multiple PC cohorts. A simplified RRM2 signature (12 genes) was identified by intersecting the RRM2 signature, PCS1 signature, and the PAM50 classifier. Intriguingly, inhibition of RRM2 specifically targets PCS1 and luminal B genes. Furthermore, 11 genes in the RRM2 signature were correlated with enzalutamide resistance by using a single-cell RNA-seq dataset from PC circulating tumor cells. Finally, high expression of RRM2 was associated with an immunosuppressive tumor-immune microenvironment in both primary prostate cancer and metastatic prostate cancer using CIBERSORT analysis and LM22, a validated leukocyte gene signature matrix. These data demonstrate that RRM2 is a driver of aggressive prostate cancer subtypes and contributes to immune escape, suggesting that RRM2 inhibition may be of clinical benefit for patients with PC.


Subject(s)
Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Neoplasm Recurrence, Local/metabolism , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Ribonucleoside Diphosphate Reductase/metabolism , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cohort Studies , Computational Biology , Databases, Genetic , Gene Expression Profiling , Gene Ontology , Gene Silencing , Humans , Male , Neoplasm Grading , Neoplasm Recurrence, Local/genetics , Neoplasm Staging , Nitriles/pharmacology , Phenylthiohydantoin/pharmacology , Prognosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA Interference , RNA-Seq , Ribonucleoside Diphosphate Reductase/antagonists & inhibitors , Ribonucleoside Diphosphate Reductase/genetics , Single-Cell Analysis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Up-Regulation
7.
Food Chem ; 326: 126969, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32438229

ABSTRACT

Biocompatible magnetic molecularly imprinted polymers (BMMIPs) were prepared with Zein for the first time, and were used to enrich tetracycline compounds selectively. Innovative combination of BMMIPs and electrochemistry to obtain lower detection line to satisfy industrial detection demands. Using Zein as the crosslinking agent, the polymers were synthesized on the surface of Fe3O4 particles. The scanning electron microscope, transmission electron microscope and X-ray diffraction technologies were used to characterize BMMIPs. Through optimization, BMMIPs attained large adsorption capacity (236.40 mg/g) with fast kinetics (40 min) and followed the Langmuir isotherm and pseudo-second-order kinetic models. BMMIPs had good recognition ability, the selective factors of oxytetracycline, chlortetracycline, doxycycline were 4.78, 4.23, and 3.39, respectively. Excellent linearity was attained in the range of 0.025-500 µg/mL, with low detection limits and low quantitation limits of 0.025 and 0.083 µg/mL. According to our exploring, BMMIPs was ideal materials for enrichment of tetracycline in complex biological samples.


Subject(s)
Biocompatible Materials/chemistry , Food Contamination/analysis , Milk/chemistry , Molecular Imprinting/methods , Tetracyclines/analysis , Adsorption , Animals , Anti-Bacterial Agents/analysis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Electrochemical Techniques , Food Analysis/methods , Limit of Detection , Magnetic Phenomena , Magnetite Nanoparticles/chemistry , Polymers/chemistry , Tetracycline/analysis , Tetracycline/chemistry , Tetracycline/isolation & purification , Tetracyclines/chemistry , Tetracyclines/isolation & purification , X-Ray Diffraction , Zein/chemistry
8.
Clin Cancer Res ; 26(8): 2047-2064, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31796516

ABSTRACT

PURPOSE: Previous sequencing studies revealed that alterations of genes associated with DNA damage response (DDR) are enriched in men with metastatic castration-resistant prostate cancer (mCRPC). BRCA2, a DDR and cancer susceptibility gene, is frequently deleted (homozygous and heterozygous) in men with aggressive prostate cancer. Here we show that patients with prostate cancer who have lost a copy of BRCA2 frequently lose a copy of tumor suppressor gene RB1; importantly, for the first time, we demonstrate that co-loss of both genes in early prostate cancer is sufficient to induce a distinct biology that is likely associated with worse prognosis. EXPERIMENTAL DESIGN: We prospectively investigated underlying molecular mechanisms and genomic consequences of co-loss of BRCA2 and RB1 in prostate cancer. We used CRISPR-Cas9 and RNAi-based methods to eliminate these two genes in prostate cancer cell lines and subjected them to in vitro studies and transcriptomic analyses. We developed a 3-color FISH assay to detect genomic deletions of BRCA2 and RB1 in prostate cancer cells and patient-derived mCRPC organoids. RESULTS: In human prostate cancer cell lines (LNCaP and LAPC4), loss of BRCA2 leads to the castration-resistant phenotype. Co-loss of BRCA2-RB1 in human prostate cancer cells induces an epithelial-to-mesenchymal transition, which is associated with invasiveness and a more aggressive disease phenotype. Importantly, PARP inhibitors attenuate cell growth in human mCRPC-derived organoids and human CRPC cells harboring single-copy loss of both genes. CONCLUSIONS: Our findings suggest that early identification of this aggressive form of prostate cancer offers potential for improved outcomes with early introduction of PARP inhibitor-based therapy.See related commentary by Mandigo and Knudsen, p. 1784.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , BRCA2 Protein , Biomarkers, Tumor , Genes, BRCA2 , Humans , Male , Phenotype , Prostatic Neoplasms, Castration-Resistant/genetics
9.
Mol Oncol ; 13(9): 1944-1958, 2019 09.
Article in English | MEDLINE | ID: mdl-31225930

ABSTRACT

Epigenetic silencing of miRNA is a primary mechanism of aberrant miRNA expression in cancer, and hypermethylation of miRNA promoters has been reported to contribute to prostate cancer initiation and progression. Recent data have shown that the miR-193b promoter is hypermethylated in prostate cancer compared with normal tissue, but studies assessing its functional significance have not been performed. We aimed to elucidate the function of miR-193b and identify its critical targets in prostate cancer. We observed an inverse correlation between miR-193b level and methylation of its promoter in The Cancer Genome Atlas (TCGA) cohort. Overexpression of miR-193b in prostate cancer cell lines inhibited invasion and induced apoptosis. We found that a majority of the top 150 genes downregulated when miR-193b was overexpressed in liposarcoma are overexpressed in metastatic prostate cancer and that 41 miR-193b target genes overlapped with the 86 genes in the aggressive prostate cancer subtype 1 (PCS1) signature. Overexpression of miR-193b led to the inhibition of the majority of the 41 genes in prostate cancer cell lines. High expression of the 41 genes was correlated with recurrence of prostate cancer. Knockdown of miR-193b targets FOXM1 and RRM2 in prostate cancer cells phenocopied overexpression of miR-193b. Dual treatment with DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors decreased miR-193b promoter methylation and restored inhibition of FOXM1 and RRM2. Our data suggest that silencing of miR-193b through promoter methylation may release the inhibition of PCS1 genes, contributing to prostate cancer progression and suggesting a possible therapeutic strategy for aggressive prostate cancer.


Subject(s)
DNA Methylation , DNA, Neoplasm/metabolism , Gene Silencing , MicroRNAs/metabolism , Promoter Regions, Genetic , Prostatic Neoplasms/metabolism , RNA, Neoplasm/metabolism , DNA, Neoplasm/genetics , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism , Humans , Male , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , PC-3 Cells , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Neoplasm/genetics , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/metabolism
10.
Clin Cancer Res ; 25(14): 4480-4492, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30996073

ABSTRACT

PURPOSE: Defects in genes in the DNA repair pathways significantly contribute to prostate cancer progression. We hypothesize that overexpression of DNA repair genes may also drive poorer outcomes in prostate cancer. The ribonucleotide reductase small subunit M2 (RRM2) is essential for DNA synthesis and DNA repair by producing dNTPs. It is frequently overexpressed in cancers, but very little is known about its function in prostate cancer. EXPERIMENTAL DESIGN: The oncogenic activity of RRM2 in prostate cancer cells was assessed by inhibiting or overexpressing RRM2. The molecular mechanisms of RRM2 function were determined. The clinical significance of RRM2 overexpression was evaluated in 11 prostate cancer clinical cohorts. The efficacy of an RRM2 inhibitor (COH29) was assessed in vitro and in vivo. Finally, the mechanism underlying the transcriptional activation of RRM2 in prostate cancer tissue and cells was determined. RESULTS: Knockdown of RRM2 inhibited its oncogenic function, whereas overexpression of RRM2 promoted epithelial mesenchymal transition in prostate cancer cells. The prognostic value of RRM2 RNA levels in prostate cancer was confirmed in 11 clinical cohorts. Integrating the transcriptomic and phosphoproteomic changes induced by RRM2 unraveled multiple oncogenic pathways downstream of RRM2. Targeting RRM2 with COH29 showed excellent efficacy. Thirteen putative RRM2-targeting transcription factors were bioinformatically identified, and FOXM1 was validated to transcriptionally activate RRM2 in prostate cancer. CONCLUSIONS: We propose that increased expression of RRM2 is a mechanism driving poor patient outcomes in prostate cancer and that its inhibition may be of significant therapeutic value.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Proliferation , DNA Repair , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/pathology , Ribonucleoside Diphosphate Reductase/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cells, Cultured , Cohort Studies , Forkhead Box Protein M1/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Prognosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Ribonucleoside Diphosphate Reductase/genetics , Survival Rate , Transcriptional Activation , Transcriptome , Xenograft Model Antitumor Assays
11.
Sci Rep ; 9(1): 3197, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30824765

ABSTRACT

Liposarcoma is the most common soft tissue sarcoma. Molecularly targeted therapeutics have had limited efficacy in liposarcomas, in part because of inadequate knowledge of the complex molecular alterations in these tumors. Our recent study revealed the tumor suppressive function of miR-193b in liposarcoma. Considering the biological and clinical heterogeneity of liposarcoma, here, we confirmed the under-expression of miR-193b in additional patient liposarcoma samples and cell lines. Based on STRING analysis of protein-protein interactions among the reported putative miR-193b targets, we validated three: PDGFRß, SMAD4, and YAP1, belonging to strongly interacting pathways (focal adhesion, TGFß, and Hippo, respectively). We show that all three are directly targeted by miR-193b in liposarcoma. Inhibition of PDGFRß reduces liposarcoma cell viability and increases adipogenesis. Knockdown of SMAD4 promotes adipogenic differentiation. miR-193b targeting of the Hippo signaling effector YAP1 indirectly inhibits Wnt/ß-catenin signaling. Both a PDGFR inhibitor (CP-673451) and a Wnt/ ß-catenin inhibitor (ICG-001) had potent inhibitory effects on liposarcoma cells, suggesting their potential application in liposarcoma treatment. In summary, we demonstrate that miR-193b controls cell growth and differentiation in liposarcoma by targeting multiple key components (PDGFRß, SMAD4, and YAP1) in several oncogenic signaling pathways.


Subject(s)
Carcinogenesis/genetics , Liposarcoma/genetics , MicroRNAs/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing/metabolism , Adipogenesis/genetics , Base Sequence , Cell Differentiation/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Liposarcoma/pathology , MicroRNAs/genetics , Smad4 Protein/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway/genetics , YAP-Signaling Proteins
12.
Cancer Epidemiol Biomarkers Prev ; 28(4): 707-714, 2019 04.
Article in English | MEDLINE | ID: mdl-30602500

ABSTRACT

BACKGROUND: 4%-9% of prostate cancers harbor homozygous deletions of the androgen-induced tumor suppressor gene, promyelocytic leukemia zinc finger (PLZF, ZBTB16). PLZF loss induces an in vitro phenotype of castration resistance and enzalutamide resistance. The association of low expression of PLZF and clinical outcomes is unclear. METHODS: We assessed PLZF mRNA expression in patients diagnosed with primary prostate cancer during prospective follow-up of the Health Professionals Follow-up Study (HPFS; n = 254) and the Physicians' Health Study (PHS; n = 150), as well as in The Cancer Genome Atlas (n = 333). We measured PTEN status (using copy numbers and IHC) and transcriptional activation of the MAPK pathway. Patients from HPFS and PHS were followed for metastases and prostate cancer-specific mortality (median, 15.3 years; 113 lethal events). RESULTS: PLZF mRNA expression was lower in tumors with PLZF deletions. There was a strong, positive association between intratumoral androgen receptor (AR) signaling and PLZF expression. PLZF expression was also lower in tumors with PTEN loss. Low PLZF expression was associated with higher MAPK signaling. Patients in the lowest quartile of PLZF expression compared with those in the highest quartile were more likely to develop lethal prostate cancer, independent of clinicopathologic features, Gleason score, and AR signaling (odds ratio, 3.17; 95% confidence interval, 1.32-7.60). CONCLUSIONS: Low expression of the tumor suppressor gene PLZF is associated with a worse prognosis in primary prostate cancer. IMPACT: Suppression of PLZF as a consequence of androgen deprivation may be undesirable. PLZF should be tested as a predictive marker for resistance to androgen deprivation therapy.


Subject(s)
Genes, Tumor Suppressor/physiology , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Prostatic Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Humans , Male , Middle Aged
13.
J Clin Invest ; 128(7): 2979-2995, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29863497

ABSTRACT

Epigenetic modifications control cancer development and clonal evolution in various cancer types. Here, we show that loss of the male-specific histone demethylase lysine-specific demethylase 5D (KDM5D) encoded on the Y chromosome epigenetically modifies histone methylation marks and alters gene expression, resulting in aggressive prostate cancer. Fluorescent in situ hybridization demonstrated that segmental or total deletion of the Y chromosome in prostate cancer cells is one of the causes of decreased KDM5D mRNA expression. The result of ChIP-sequencing analysis revealed that KDM5D preferably binds to promoter regions with coenrichment of the motifs of crucial transcription factors that regulate the cell cycle. Loss of KDM5D expression with dysregulated H3K4me3 transcriptional marks was associated with acceleration of the cell cycle and mitotic entry, leading to increased DNA-replication stress. Analysis of multiple clinical data sets reproducibly showed that loss of expression of KDM5D confers a poorer prognosis. Notably, we also found stress-induced DNA damage on the serine/threonine protein kinase ATR with loss of KDM5D. In KDM5D-deficient cells, blocking ATR activity with an ATR inhibitor enhanced DNA damage, which led to subsequent apoptosis. These data start to elucidate the biological characteristics resulting from loss of KDM5D and also provide clues for a potential novel therapeutic approach for this subset of aggressive prostate cancer.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Histone Demethylases/deficiency , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Chromosomes, Human, Y/genetics , DNA Damage , Epigenesis, Genetic , Gene Dosage , Gene Knockdown Techniques , Histone Code/genetics , Histone Demethylases/genetics , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred NOD , Mice, SCID , Minor Histocompatibility Antigens/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/enzymology , Prostatic Neoplasms, Castration-Resistant/genetics , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays
14.
Cancer Res ; 77(21): 5728-5740, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28882999

ABSTRACT

Well-differentiated and dedifferentiated liposarcomas (WDLS/DDLS) account for approximately 13% of all soft tissue sarcoma in adults and cause substantial morbidity or mortality in the majority of patients. In this study, we evaluated the functions of miRNA (miR-193b) in liposarcoma in vitro and in vivo Deep RNA sequencing on 93 WDLS, 145 DDLS, and 12 normal fat samples demonstrated that miR-193b was significantly underexpressed in DDLS compared with normal fat. Reintroduction of miR-193b induced apoptosis in liposarcoma cells and promoted adipogenesis in human adipose-derived stem cells (ASC). Integrative transcriptomic and proteomic analysis of miR-193b-target networks identified novel direct targets, including CRK-like proto-oncogene (CRKL) and focal adhesion kinase (FAK). miR-193b was found to regulate FAK-SRC-CRKL signaling through CRKL and FAK. miR-193b also stimulated reactive oxygen species signaling by targeting the antioxidant methionine sulfoxide reductase A to modulate liposarcoma cell survival and ASC differentiation state. Expression of miR-193b in liposarcoma cells was downregulated by promoter methylation, resulting at least in part from increased expression of the DNA methyltransferase DNMT1 in WDLS/DDLS. In vivo, miR-193b mimetics and FAK inhibitor (PF-562271) each inhibited liposarcoma xenograft growth. In summary, miR-193b not only functions as a tumor suppressor in liposarcoma but also promotes adipogenesis in ASC. Furthermore, this study reveals key tyrosine kinase and DNA methylation pathways in liposarcoma, some with immediate implications for therapeutic exploration. Cancer Res; 77(21); 5728-40. ©2017 AACR.


Subject(s)
Adipogenesis/genetics , Gene Expression Regulation, Neoplastic , Liposarcoma/genetics , MicroRNAs/genetics , Signal Transduction/genetics , Stem Cells/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adipose Tissue/cytology , Animals , Cell Line, Tumor , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Gene Expression Profiling/methods , Genes, Tumor Suppressor , Humans , Indoles/pharmacology , Liposarcoma/drug therapy , Liposarcoma/pathology , Methionine Sulfoxide Reductases/genetics , Methionine Sulfoxide Reductases/metabolism , Mice, Inbred ICR , Mice, SCID , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Mas , Reactive Oxygen Species/metabolism , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays
15.
Am J Chin Med ; 44(6): 1221-1236, 2016.
Article in English | MEDLINE | ID: mdl-27744729

ABSTRACT

Drug-induced liver injury (DILI) is the most common cause of acute liver failure. Disruption of the Th17/Treg balance can lead to hepatic inflammation, which causes the main symptoms of DILI. Here we investigate the protective mechanisms of (-)-Epigallocatechin-3-gallate (EGCG) on triptolide (TP)-induced DILI that shows the Th17/Treg imbalance. Pretreatment with EGCG (5[Formula: see text]mg/kg) for 10 days before TP (0.5[Formula: see text]mg/kg) administration in mice significantly reduced the increased alanine aminotransferase (ALT) level ([Formula: see text]) induced by TP treatment. The hepatic histology analysis further proved that EGCG protected mice from TP-induced liver injury. The imbalance of Th17/Treg was induced by TP treatment, as shown by the upregulation of TLR4 and downregulation of Tim3 expression. EGCG pretreatment can maintain the expression of TLR4 and Tim3 at normal levels to restore the Th17/Treg imbalance. In addition, EGCG can block the TP-induced expression of the downstream targets of TLR4, including MyD88, NF[Formula: see text]B, and retinoid related orphan receptor (ROR-[Formula: see text]t), while EGCG can restore the TP inhibition of forkhead/winged-helix family transcriptional repressor p3 (FoxP3) that is the downstream target of Tim3. Consequently, EGCG pretreatment can effectively inhibit the Th17-related pro-inflammatory cytokine (e.g. IL-17 and IL-6) upregulation induced by TP treatment. However, TP inhibition of Treg-related anti-inflammatory cytokine IL-10 production was restored by EGCG pretreatment. Taken together, these results suggest that EGCG possesses significant protective properties against TP-induced hepatic inflammatory injury, and that these properties are carried out via the restoration of the Th17/Treg imbalance by the inhibition of the TLR4 signaling pathway and the enhanced activation of the Tim3 signaling pathway.


Subject(s)
Catechin/analogs & derivatives , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/prevention & control , Diterpenes/adverse effects , Immunosuppressive Agents/adverse effects , Phenanthrenes/adverse effects , Phytotherapy , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Catechin/administration & dosage , Catechin/pharmacology , Chemical and Drug Induced Liver Injury/etiology , Cytokines/metabolism , Disease Models, Animal , Epoxy Compounds/adverse effects , Female , Hepatitis A Virus Cellular Receptor 2 , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Toll-Like Receptor 4 , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...