Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Proc Natl Acad Sci U S A ; 121(37): e2406854121, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39231208

ABSTRACT

Alzheimer's disease (AD) is a prevalent neurodegenerative disease characterized by cognitive decline and learning/memory impairment associated with neuronal cell loss. Estrogen-related receptor α (ERRα) and ERRƎĀ³, which are highly expressed in the brain, have emerged as potential AD regulators, with unelucidated underlying mechanisms. Here, we identified genome-wide binding sites for ERRα and ERRƎĀ³ in human neuronal cells. They commonly target a subset of genes associated with neurodegenerative diseases, including AD. Notably, Dickkopf-1 (DKK1), a Wnt signaling pathway antagonist, was transcriptionally repressed by both ERRα and ERRƎĀ³ in human neuronal cells and brain. ERRα and ERRƎĀ³ repress RNA polymerase II (RNAP II) accessibility at the DKK1 promoter by modulating a specific active histone modification, histone H3 lysine acetylation (H3K9ac), with the potential contribution of their corepressor. This transcriptional repression maintains Wnt signaling activity, preventing tau phosphorylation and promoting a healthy neuronal state in the context of AD.


Subject(s)
Alzheimer Disease , ERRalpha Estrogen-Related Receptor , Intercellular Signaling Peptides and Proteins , Receptors, Estrogen , Animals , Humans , Mice , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Brain/metabolism , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Neurons/metabolism , Phosphorylation , Promoter Regions, Genetic , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , tau Proteins/metabolism , tau Proteins/genetics , Wnt Signaling Pathway/genetics
2.
BMC Cancer ; 24(1): 554, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698344

ABSTRACT

BACKGROUND: Prostate cancer is dependent on androgen receptor (AR) signaling, and androgen deprivation therapy (ADT) has proven effective in targeting prostate cancer. However, castration-resistant prostate cancer (CRPC) eventually emerges. AR signaling inhibitors (ARSI) have been also used, but resistance to these agents develops due to genetic AR alterations and epigenetic dysregulation. METHODS: In this study, we investigated the role of OCT1, a member of the OCT family, in an AR-positive CRPC patient-derived xenograft established from a patient with resistance to ARSI and chemotherapy. We conducted a genome-wide analysis chromatin immunoprecipitation followed by sequencing and bioinformatic analyses using public database. RESULTS: Genome-wide analysis of OCT1 target genes in PDX 201.1Ā A revealed distinct OCT1 binding sites compared to treatment-naĆÆve cells. Bioinformatic analyses revealed that OCT1-regulated genes were associated with cell migration and immune system regulation. In particular, C-terminal Binding Protein 2 (CTBP2), an OCT1/AR target gene, was correlated with poor prognosis and immunosuppressive effects in the tumor microenvironment. Metascape revealed that CTBP2 knockdown affects genes related to the immune response to bacteria. Furthermore, TISIDB analysis suggested the relationship between CTBP2 expression and immune cell infiltration in prostate cancer, suggesting that it may contribute to immune evasion in CRPC. CONCLUSIONS: Our findings shed light on the genome-wide network of OCT1 and AR in AR-positive CRPC and highlight the potential role of CTBP2 in immune response and tumor progression. Targeting CTBP2 may represent a promising therapeutic approach for aggressive AR-positive CRPC. Further validation will be required to explore novel therapeutic strategies for CRPC management.


Subject(s)
Alcohol Oxidoreductases , Co-Repressor Proteins , Gene Expression Regulation, Neoplastic , Octamer Transcription Factor-1 , Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen , Male , Humans , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Mice , Animals , Octamer Transcription Factor-1/metabolism , Octamer Transcription Factor-1/genetics , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Up-Regulation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Xenograft Model Antitumor Assays , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Tumor Microenvironment , Signal Transduction
3.
Mol Cell ; 64(1): 176-188, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27716482

ABSTRACT

How deregulation of chromatin modifiers causes malignancies is of general interest. Here, we show that histone H2A T120 is phosphorylated in human cancer cell lines and demonstrate that this phosphorylation is catalyzed by hVRK1. Cyclin D1 was one of ten genes downregulated upon VRK1 knockdown in two different cell lines and showed loss of H2A T120 phosphorylation and increased H2A K119 ubiquitylation of its promoter region, resulting in impaired cell growth. InĀ vitro, H2A T120 phosphorylation and H2A K119 ubiquitylation are mutually inhibitory, suggesting that histone phosphorylation indirectly activates chromatin. Furthermore, expression of aĀ phosphomimetic H2A T120D increased H3 K4 methylation. Finally, both VRK1 and the H2A T120D mutant histone transformed NIH/3T3 cells. These results suggest that histone H2A T120 phosphorylation by hVRK1 causes inappropriate gene expression, including upregulated cyclin D1, which promotes oncogenic transformation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic , Histones/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chromatin/chemistry , Chromatin/metabolism , Cyclin D1/metabolism , Drosophila Proteins , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , HeLa Cells , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Methylation , Mice , Oligopeptides/genetics , Oligopeptides/metabolism , Phosphorylation , Protamine Kinase/genetics , Protamine Kinase/metabolism , Protein Serine-Threonine Kinases/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Threonine/metabolism , Ubiquitination
4.
Int J Urol ; 31(6): 590-597, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38345202

ABSTRACT

Androgen deprivation therapy has achieved significant success in treating prostate cancer through strategies centered on the androgen receptor. However, the emergence of castration-resistant prostate cancer highlights this therapy limitation, underscoring the need to elucidate the mechanisms of treatment resistance. This review aimed to focus on multifaceted resistance mechanisms, including androgen receptor overexpression, splice variants, missense mutations, the involvement of the glucocorticoid receptor, and alterations in coregulators and transcription factors, revealing their roles in castration-resistant prostate cancer progression. These mechanisms promote cell survival and proliferation, depending on the androgen receptor signaling pathway, leading to resistance to conventional therapies. Amplification and mutations in the androgen receptor gene facilitate selective adaptation in treatment-resistant cells, consequently diminishing therapeutic efficacy. Furthermore, the activation of glucocorticoid receptors and aberrant regulation of specific coregulators and transcription factors contribute to the activation of androgen receptor-independent signaling pathways, promoting cell survival and proliferation. These findings hold promise for identifying new targets for treating castration-resistant prostate cancer and developing personalized treatment strategies. The development of future therapies will hinge on precisely targeting the androgen receptor signaling pathway, necessitating a deeper understanding of the molecular targets unique to castration-resistant prostate cancer.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen , Signal Transduction , Humans , Male , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/therapy , Prostatic Neoplasms, Castration-Resistant/drug therapy , Drug Resistance, Neoplasm/genetics , Cell Proliferation , Androgen Antagonists/therapeutic use , Gene Expression Regulation, Neoplastic , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Prostatic Neoplasms/therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics
5.
Biochem Biophys Res Commun ; 623: 9-16, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35868070

ABSTRACT

Androgens and androgen receptor (AR) have a central role in prostate cancer progression by regulating its downstream signaling. Although androgen depletion therapy (ADT) is the primary treatment for most prostate cancers, they acquires resistance to ADT and become castration resistant prostate cancers (CRPC). AR complex formation with multiple transcription factors is important for enhancer activity and transcriptional regulation, which can contribute to cancer progression and resistance to ADT. We previously demonstrated that OCT1 collaborates with AR in prostate cancer, and that a pyrrole-imidazole (PI) polyamide (PIP) targeting OCT1 inhibits cell and castration-resistant tumor growth (Obinata D etĀ al. Oncogene 2016). PIP can bind to DNA non-covalently without a drug delivery system unlike most DNA targeted therapeutics. In the present study, we developed a PIP modified with a DNA alkylating agent, chlorambucil (ChB) (OCT1-PIP-ChB). Then its effect on the growth of prostate cancer LNCaP, 22Rv1, and PC3 cells, pancreatic cancer BxPC3 cells, and colon cancer HCT116Ā cells, as well as non-cancerous MCF-10A epithelial cells, were analyzed. It was shown that the IC50s of OCT1-PIP-ChB for 22Rv1 and LNCaP were markedly lower compared to other cells, including non-cancerous MCF-10A cells. Comprehensive gene expression analysis of CRPC model 22Rv1 cells treated with IC50 concentrations of OCT1-PIP-ChB revealed that the gene group involved in DNA double-strand break repair was the most enriched among gene sets repressed by OCT1-PIP-ChB treatment. Importantly, inĀ vivo study using 22Rv1 xenografts, we showed that OCT1-PIP-ChB significantly reduced tumor growth compared to the control group without showing obvious adverse effects. Thus, the PIP combined with ChB can exert a significant inhibitory effect on prostate cancer cell proliferation and castration-resistant tumor growth, suggesting a potential role as a therapeutic agent.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Alkylating Agents , Cell Line, Tumor , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Male , Nylons/pharmacology , Prostatic Neoplasms, Castration-Resistant/pathology , Pyrroles/pharmacology , Pyrroles/therapeutic use , Receptors, Androgen/metabolism
6.
Proc Natl Acad Sci U S A ; 115(19): 4975-4980, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29686105

ABSTRACT

Androgen receptor (AR) signaling is essential for prostate cancer progression and acquiring resistance to hormone therapy. However, the molecular pathogenesis through AR activation has not been fully understood. We performed integrative transcriptomic analysis to compare the AR program in a castration-resistant prostate cancer (CRPC) model with that in their parental hormone-sensitive cells. We found that the gene cordon-bleu-like 1 (COBLL1) is highly induced by AR in CRPC model cells. The expression of COBLL1 that possesses an actin-binding domain is up-regulated in clinical prostate cancer tissues and is associated with a poor prognosis for prostate cancer patients. COBLL1 is involved in the cancer cell morphogenesis to a neuron-like cell shape observed in the CRPC model cells, promoting cell growth and migration. Moreover, nuclear COBLL1 interacts with AR to enhance complex formation with CDK1 and facilitates AR phosphorylation for genomic binding in CRPC model cells. Thus, our findings showed the mechanistic relevance of cordon-bleu proteins during the AR-mediated progression to CRPC.


Subject(s)
Cell Movement , Cell Nucleus/metabolism , Multiprotein Complexes/metabolism , Neoplasm Proteins/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Transcription Factors/metabolism , Animals , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/pathology , Gene Expression Profiling , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Multiprotein Complexes/genetics , Neoplasm Proteins/genetics , Prostatic Neoplasms, Castration-Resistant/diagnosis , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Domains , Receptors, Androgen/genetics , Transcription Factors/genetics
7.
Int J Mol Sci ; 22(21)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34768935

ABSTRACT

Octamer transcription factor 1 (OCT1) is a transcriptional factor reported to be a poor prognostic factor in various cancers. However, the clinical value of OCT1 in breast cancer is not fully understood. In the present study, an immunohistochemical study of OCT1 protein was performed using estrogen receptor (ER)-positive breast cancer tissues from 108 patients. Positive OCT1 immunoreactivity (IR) was associated with the shorter disease-free survival (DFS) of patients (p = 0.019). Knockdown of OCT1 inhibited cell proliferation in MCF-7 breast cancer cells as well as its derivative long-term estrogen-deprived (LTED) cells. On the other hand, the overexpression of OCT1 promoted cell proliferation in MCF-7 cells. Using microarray analysis, we identified the non-structural maintenance of chromosomes condensin I complex subunit H (NCAPH) as a novel OCT1-taget gene in MCF-7 cells. Immunohistochemical analysis showed that NCAPH IR was significantly positively associated with OCT1 IR (p < 0.001) and that positive NCAPH IR was significantly related to the poor DFS rate of patients (p = 0.041). The knockdown of NCAPH inhibited cell proliferation in MCF-7 and LTED cells. These results demonstrate that OCT1 and its target gene NCAPH are poor prognostic factors and potential therapeutic targets for patients with ER-positive breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Cycle Proteins/genetics , Cell Proliferation/genetics , Nuclear Proteins/genetics , Octamer Transcription Factor-1/genetics , Biomarkers, Tumor/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Female , HEK293 Cells , Humans , MCF-7 Cells , Middle Aged , Neoplasm Invasiveness/genetics , Nuclear Proteins/metabolism , Octamer Transcription Factor-1/metabolism , Prognosis , RNA Interference , RNA, Small Interfering/genetics , Receptors, Estrogen/metabolism
8.
Cancer Sci ; 111(1): 148-159, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31733123

ABSTRACT

The majority of breast cancers are primarily hormone-sensitive and can be managed by endocrine therapy, although therapy-resistant or hormone-refractory cancers need alternative treatments. Recently, increasing attention is being paid to RNA-binding proteins (RBP) in cancer pathophysiology. The precise role of RBP in breast cancer, however, remains to be clarified. We herein show that an RBP non-POU domain-containing octamer binding (NONO) plays a critical role in the pathophysiology of breast cancers regardless of their hormone dependency. Clinicopathological and immunohistochemical study of 127 breast cancer cases showed that NONO is a significant independent prognostic factor for breast cancer patients. Notably, siRNA-mediated NONO knockdown substantially repressed the proliferation of both hormone-sensitive MCF-7 and hormone-refractory MB-MDA-231 breast cancer cells. Integrative analysis combined with expression microarray and RIP-sequencing (RNA immunoprecipitation-sequencing) showed that NONO post-transcriptionally regulates the expression of cell proliferation-related genes by binding to their mRNAs, as exemplified by S-phase-associated kinase 2 and E2F transcription factor 8. Overall, these results suggest that NONO is a key regulator for breast cancer proliferation through the pre-mRNA splicing of cell proliferation-related genes and could be a potential new diagnostic and therapeutic target for advanced disease.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , RNA Processing, Post-Transcriptional/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , S-Phase Kinase-Associated Proteins/genetics , Cell Line, Tumor , Female , Gene Expression Regulation/genetics , Humans , Immunoprecipitation/methods , MCF-7 Cells , RNA, Messenger/genetics
9.
Cancer Sci ; 111(3): 881-890, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31883420

ABSTRACT

TRIM44 has oncogenic roles in various cancers. However, TRIM44 expression and its function in renal cell carcinoma (RCC) are still unknown. Here in this study, we investigated the clinical significance of TRIM44 and its biological function in RCC. TRIM44 overexpression was significantly associated with clinical M stage, histologic type (clear cell) and presence of lymphatic invasion (PĀ =Ā .047, PĀ =Ā .005, and PĀ =Ā .028, respectively). Moreover, TRIM44 overexpression was significantly associated with poor prognosis in terms of cancer-specific survival (PĀ =Ā .019). Gain-of-function and loss-of-function studies using TRIM44 and siTRIM44 transfection showed that TRIM44 promotes cell proliferation and cell migration in two RCC cell lines, Caki1 and 769P. To further investigate the role of TRIM44 in RCC, we performed integrated microarray analysis in Caki1 and 769P cells and explored the data in the Oncomine database. Interestingly, FRK was identified as a promising candidate target gene of TRIM44, which was downregulated in RCC compared with normal renal tissues. We found that cell proliferation was inhibited by TRIM44 knockdown and then recovered by siFRK treatment. Taken together, the present study revealed the association between high expression of TRIM44 and poor prognosis in RCC patients and that TRIM44 promotes cell proliferation by regulating FRK.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cell Movement/physiology , Cell Proliferation/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Neoplasms/metabolism , Neoplasm Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Tripartite Motif Proteins/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Disease Progression , Down-Regulation/physiology , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Kidney Neoplasms/pathology , Male , Middle Aged , Prognosis , Signal Transduction/physiology
10.
Cancer Sci ; 111(8): 2943-2953, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32495467

ABSTRACT

Neuroblastoma (NB) is a childhood malignancy originating from the sympathetic nervous system, and accounts for approximately 15% of all pediatric cancer-related deaths. As the 5-y survival rate of patients with high-risk NB is <50%, novel therapeutic strategies for NB patients are urgently required. Nonaethylene glycol mono('4-iodo-4-biphenyl)ester (9bw) is a polyethylene glycol derivative, synthesized by modifying a compound originally extracted from filamentous bacteria. Although 9bw shows remarkable inhibition of tumor cell growth, the underlying mechanisms remain unclear. Here, we examined the efficacy of 9bw on human NB-derived cells, and investigated the molecular mechanisms underlying the cytotoxic effects of 9bw on these cells. Our results indicated that 9bw induced cell death in NB cells by decreasing the production of ATP. Metabolome analysis and measurement of oxygen consumption indicated that 9bw markedly suppressed oxidative phosphorylation (OXPHOS). Further analyses indicated that 9bw inhibited the activity of mitochondrial respiratory complex I. Moreover, we showed that 9bw inhibited growth of NB in vivo. Based on the results of the present study, 9bw is a good candidate as a novel agent for treatment of NB.


Subject(s)
Antineoplastic Agents/pharmacology , Electron Transport Complex I/antagonists & inhibitors , Esters/pharmacology , Neuroblastoma/drug therapy , Oxidative Phosphorylation/drug effects , Polyethylene Glycols/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Electron Transport Complex I/metabolism , Esters/chemistry , Esters/therapeutic use , Female , Humans , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Neuroblastoma/pathology , Polyethylene Glycols/chemistry , Polyethylene Glycols/therapeutic use , Xenograft Model Antitumor Assays
11.
Genes Cells ; 24(11): 719-730, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31541592

ABSTRACT

Amyloid precursor protein (APP) is a representative gene related to Alzheimer's disease (AD). Androgens function by binding to the androgen receptor (AR). Both androgen and RNA-binding protein PSF play a role in the pathology of AD. However, the involvement of AR and PSF in APP regulation in neuron has not been investigated. Here, we explored the regulatory mechanism of APP expression by AR and PSF using neuron-derived cells. We demonstrated that androgen up-regulates the production of APP at the mRNA and protein levels. This induction is enhanced by AR over-expression and inhibited by its silencing. One candidate AR-binding region was identified in the intron region of APP and validated its activity as AR-dependent enhancer by the luciferase assay. Furthermore, the public transcriptome data of brain tissues of mice indicated that APP is regulated by PSF post-transcriptionally. We observed a decreased expression of APP after PSF knockdown and interaction of PSF with the APP transcript. Moreover, we revealed that silencing of PSF inhibited the stability of the APP mRNA. Thus, these results presented a new regulatory mechanism of APP expression by androgen through AR-mediated transcription and PSF at the post-transcriptional level that might be associated with the occurrence of AD.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Androgens/genetics , Androgens/metabolism , Neurons/metabolism , RNA-Binding Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , Cell Line , Disease Models, Animal , Gene Knockdown Techniques , Humans , Mice , PTB-Associated Splicing Factor/genetics , Protein Binding , RNA, Messenger/metabolism , Receptors, Androgen , Transcriptome , Up-Regulation
12.
Proc Natl Acad Sci U S A ; 114(39): 10461-10466, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28893982

ABSTRACT

Developing therapeutic approaches are necessary for treating hormone-refractory prostate cancer. Activation of androgen receptor (AR) and its variants' expression along with the downstream signals are mostly important for disease progression. However, the mechanism for marked increases of AR signals and its expression is still unclear. Here, we revealed that various spliceosome genes are aberrantly induced by RNA-binding protein PSF, leading to enhancement of the splicing activities for AR expression. Our high-speed sequence analyses identified global PSF-binding transcripts. PSF was shown to stabilize and activate key long noncoding RNAs and AR-regulated gene expressions in prostate cancer cells. Interestingly, mRNAs of spliceosome-related genes are putative primary targets of PSF. Their gene expressions are up-regulated by PSF in hormone-refractory prostate cancer. Moreover, PSF coordinated these spliceosome proteins to form a complex to promote AR splicing and expression. Thus, targeting PSF and its related pathways implicates the therapeutic possibility for hormone-refractory prostate cancer.


Subject(s)
PTB-Associated Splicing Factor/biosynthesis , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , RNA, Long Noncoding/genetics , Receptors, Androgen/biosynthesis , Spliceosomes/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , PTB-Associated Splicing Factor/genetics , Prostatic Neoplasms, Castration-Resistant/therapy , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptors, Androgen/genetics , Transplantation, Heterologous
13.
Cancer Sci ; 110(11): 3476-3485, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31454442

ABSTRACT

Octamer transcription factor 1 (OCT1) is an androgen receptor (AR)-interacting partner and regulates the expression of target genes in prostate cancer cells. However, the function of OCT1 in castration-resistant prostate cancer (CRPC) is not fully understood. In the present study, we used 22Rv1 cells as AR-positive CRPC model cells to analyze the role of OCT1 in CRPC. We showed that OCT1 knockdown suppressed cell proliferation and migration of 22Rv1 cells. Using microarray analysis, we identified four AR and OCT1-target genes, disks large-associated protein 5 (DLGAP5), kinesin family member 15 (KIF15), non-SMC condensin I complex subunit G (NCAPG), and NDC80 kinetochore complex component (NUF2) in 22Rv1 cells. We observed that knockdown of DLGAP5 and NUF2 suppresses growth and migration of 22Rv1 cells. Furthermore, immunohistochemical analysis showed that positive expression of DLGAP5 in prostate cancer specimens is related to poor cancer-specific survival rates of patients. Notably, enhanced expression of DLGAP5 was observed in CRPC tissues of patients. Thus, our findings suggest that these four genes regulated by the AR/OCT1 complex could have an important role in CRPC progression.


Subject(s)
Cell Cycle Proteins/genetics , Kinesins/genetics , Neoplasm Proteins/genetics , Octamer Transcription Factor-1/physiology , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/genetics , Aged , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cytoskeletal Proteins , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Silencing , Humans , Male , Microarray Analysis , Nuclear Proteins/genetics , Octamer Transcription Factor-1/genetics , Prostatic Neoplasms, Castration-Resistant/mortality , Receptors, Androgen/metabolism , Survival Rate , Up-Regulation
14.
Cancer Sci ; 109(12): 3840-3852, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30238687

ABSTRACT

Tripartite motif 36 (TRIM36) belongs to the TRIM family, most members of which are involved in ubiquitination and degradation of target proteins by functioning as E3 ubiquitin ligases. The function of TRIM36 has not been well documented, therefore, we investigated the clinical significance and function of TRIM36 in human prostate cancer (PC). Multivariate logistic regression analysis showed that TRIM36 immunoreactivity was an independent predictor of cancer-specific survival of PC patients. Gain-of-function study revealed that overexpression of TRIM36 suppressed cell proliferation and migration of LNCaP, 22Rv1, and DU145 cells. Moreover, TRIM36 knockdown using siRNA suppressed apoptosis and promoted cell proliferation and migration in LNCaP and 22Rv1 cells. Furthermore, our microarray analysis revealed that the apoptosis-related pathway was significantly upregulated by TRIM36 overexpression. The TUNEL assay showed that apoptosis promoted by docetaxel treatment was alleviated in siTRIM36-treated LNCaP and 22Rv1 cells. Taken together, these results suggest that high expression of TRIM36 is associated with favorable prognosis and that TRIM36 plays a tumor-suppressive role by inhibiting cell proliferation and migration as well as promoting apoptosis in PC.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Prostatic Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Grading , Oligonucleotide Array Sequence Analysis , Prognosis , Progression-Free Survival , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Signal Transduction , Up-Regulation
16.
J Biol Chem ; 291(34): 17861-80, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27342777

ABSTRACT

Long noncoding RNAs (lncRNA) have been associated with the development of cancer. However, the interplay between lncRNAs and androgen receptor (AR) signaling in prostate cancer is still unclear. Here, we identified lncRNAs induced by androgen in AR-positive prostate cancer cells, where induction was abolished by AR knockdown as well as an anti-androgen, bicalutamide. By combining these data, we identified an androgen-regulated lncRNA, suppressor of cytokine signaling 2-antisense transcript 1 (SOCS2-AS1), the expression of which was higher in castration-resistant prostate cancer model cells, i.e long-term androgen-deprived (LTAD) cells, than in parental androgen-dependent LNCaP cells. SOCS2-AS1 promoted castration-resistant and androgen-dependent cell growth. We found that SOCS2-AS1 knockdown up-regulated genes related to the apoptosis pathway, including tumor necrosis factor superfamily 10 (TNFSF10), and sensitized prostate cancer cells to docetaxel treatment. Moreover, we also demonstrated that SOCS2-AS1 promotes androgen signaling by modulating the epigenetic control for AR target genes including TNFSF10 These findings suggest that SOCS2-AS1 plays an important role in the development of castration-resistant prostate cancer by repressing apoptosis.


Subject(s)
Androgens/pharmacology , Apoptosis/drug effects , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Prostatic Neoplasms, Castration-Resistant/metabolism , RNA, Long Noncoding/biosynthesis , RNA, Neoplasm/biosynthesis , Up-Regulation/drug effects , Cell Line, Tumor , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , TNF-Related Apoptosis-Inducing Ligand/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/genetics , Up-Regulation/genetics
17.
Cancer Sci ; 108(11): 2107-2114, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28796922

ABSTRACT

Long non-coding RNAs (lncRNAs) are RNA transcripts larger than 200 nucleotides that do not code for proteins the aberrant expression of which has been documented in various types of cancer, including prostate cancer. Lack of appropriate sensitive and specific biomarkers for prostate cancer has led to overdiagnosis and overtreatment, making lncRNAs promising novel biomarkers as well as therapeutic targets for the disease. The present review attempts to summarize the current knowledge of lncRNA expression patterns and mechanisms in prostate cancer, which contribute to carcinogenesis. In particular, we focused on lncRNAs regulated by androgen receptor and expressed in castration-resistant prostate cancer.


Subject(s)
Biomarkers, Tumor/genetics , Prostatic Neoplasms/genetics , RNA, Long Noncoding/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Medical Overuse , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/pathology
18.
Cancer Sci ; 108(7): 1386-1393, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28474805

ABSTRACT

The proliferation of prostate cancer cells is controlled by the androgen receptor (AR) signaling pathway. However, the function of AR target genes has not been fully elucidated. In previous studies, we have identified global AR binding sites and AR target genes in prostate cancer cells. Here, we focused on Claudin 8 (CLDN8), a protein constituting tight junctions in cell membranes. We found one AR binding site in the promoter region and two functional androgen-responsive elements in the sequence. Reporter assay revealed that transcriptional activation of the CLDN8 promoter by androgen is dependent on these androgen-responsive elements. Furthermore, CLDN8 mRNA is induced by androgen time-dependently and the induction is blocked by AR inhibitor, suggesting that AR is involved in the transcriptional activation. In addition, our functional analyses by overexpression and knockdown of CLDN8 mRNA indicate that CLDN8 promotes prostate cancer cell proliferation and migration. Claudin 8 was overexpressed in prostate cancer clinical samples compared to benign tissues. Furthermore, we found that CLDN8 regulates intracellular signal transduction and stabilizes the cytoskeleton. Taken together, these results indicate that CLDN8 functions as an AR downstream signal to facilitate the progression of prostate cancer. Claudin 8 may be a novel molecular target for prostate cancer therapy.


Subject(s)
Cell Movement , Cell Proliferation , Claudins/genetics , Gene Expression Regulation, Neoplastic/genetics , Prostatic Neoplasms/pathology , Blotting, Western , Cell Movement/genetics , Cell Proliferation/genetics , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Humans , Immunohistochemistry , Male , Polymerase Chain Reaction , Receptors, Androgen/metabolism , Response Elements/genetics , Transcriptional Activation
19.
Cancer Sci ; 108(3): 373-379, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28032932

ABSTRACT

Although long non-coding RNAs (lncRNAs) have been associated with a variety of cancers, the interplay between lncRNAs and androgen receptor signaling in prostate cancer is still unclear. We identified an androgen-dependent lncRNA, POTEF-AS1, whose expression was regulated by androgen receptor in two androgen-dependent cells by using directional RNA sequencing analysis. POTEF-AS1 promoted cell growth, repressed genes related to the Toll-like receptor signaling and apoptosis pathways, and inhibited apoptosis in docetaxel-treated LNCaP cells. These findings suggest that POTEF-AS1 would play a key role in the progression of prostate cancer by repressing Toll-like receptor signaling.


Subject(s)
Antigens, Neoplasm/genetics , Apoptosis/genetics , Cell Survival/genetics , Prostatic Neoplasms/genetics , RNA, Long Noncoding/genetics , Receptors, Androgen/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/genetics , Docetaxel , Humans , Male , Prostatic Neoplasms/pathology , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/genetics , Taxoids/pharmacology , Toll-Like Receptors/antagonists & inhibitors
20.
Cancer Sci ; 108(10): 2011-2021, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28771887

ABSTRACT

Long-chain acyl-coenzyme A (CoA) synthetase 3 (ACSL3) is an androgen-responsive gene involved in the generation of fatty acyl-CoA esters. ACSL3 is expressed in both androgen-sensitive and castration-resistant prostate cancer (CRPC). However, its role in prostate cancer remains elusive. We overexpressed ACSL3 in androgen-dependent LNCaP cells and examined the downstream effectors of ACSL3. Furthermore, we examined the role of ACSL3 in the androgen metabolism of prostate cancer. ACSL3 overexpression led to upregulation of several genes such as aldo-keto reductase 1C3 (AKR1C3) involved in steroidogenesis, which utilizes adrenal androgen dehydroepiandrosterone sulfate (DHEAS) as substrate, and downregulated androgen-inactivating enzyme UDP-glucuronosyltransferase 2 (UGT2B). Exposure to DHEAS significantly increased testosterone levels and cell proliferative response in ACSL3-overexpressing cells when compared to that in control cells. A public database showed that ACSL3 level was higher in CRPC than in hormone-sensitive prostate cancer. CRPC cells showed an increased expression of ACSL3 and an expression pattern of AKR1C3 and UGT2B similar to ACSL3-overexpressing cells. DHEAS stimulation significantly promoted the proliferation of CRPC cells when compared to that of LNCaP cells. These findings suggest that ACSL3 contributes to the growth of CRPC through intratumoral steroidogenesis (i.e. promoting androgen synthesis from DHEAS and preventing the catabolism of active androgens).


Subject(s)
Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Dehydroepiandrosterone Sulfate/pharmacology , Prostatic Neoplasms, Castration-Resistant/metabolism , Testosterone/metabolism , 3-Hydroxysteroid Dehydrogenases/metabolism , Aldo-Keto Reductase Family 1 Member C3 , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glucuronosyltransferase/metabolism , Humans , Hydroxyprostaglandin Dehydrogenases/metabolism , Lipogenesis , Male , Prostatic Neoplasms, Castration-Resistant/genetics
SELECTION OF CITATIONS
SEARCH DETAIL