Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Res ; 16(12): 1940-1951, 2018 12.
Article in English | MEDLINE | ID: mdl-30042175

ABSTRACT

Prostate cancer is a prevalent public health problem, especially because noncutaneous advanced malignant forms significantly affect the lifespan and quality of life of men worldwide. New therapeutic targets and approaches are urgently needed. The current study reports elevated expression of R1 (CDCA7L/RAM2/JPO2), a c-Myc-interacting protein and transcription factor, in human prostate cancer tissue specimens. In a clinical cohort, high R1 expression is associated with disease recurrence and decreased patient survival. Overexpression and knockdown of R1 in human prostate cancer cells indicate that R1 induces cell proliferation and colony formation. Moreover, silencing R1 dramatically reduces the growth of prostate tumor xenografts in mice. Mechanistically, R1 increases c-Myc protein stability by inhibiting ubiquitination and proteolysis through transcriptional suppression of HUWE1, a c-Myc-targeting E3 ligase, via direct interaction with a binding element in the promoter. Moreover, transcriptional repression is supported by a negative coexpression correlation between R1 and HUWE1 in a prostate cancer clinical dataset. Collectively, these findings, for the first time, characterize the contribution of R1 to prostate cancer pathogenesis. IMPLICATIONS: These findings provide evidence that R1 is a novel regulator of prostate tumor growth by stabilizing c-Myc protein, meriting further investigation of its therapeutic and prognostic potential.


Subject(s)
Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Up-Regulation , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mutation , Neoplasm Transplantation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Stability , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/genetics , Survival Analysis
2.
Cancer Cell ; 31(3): 368-382, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28292438

ABSTRACT

Metastasis is a predominant cause of death for prostate cancer (PCa) patients; however, the underlying mechanisms are poorly understood. We report that monoamine oxidase A (MAOA) is a clinically and functionally important mediator of PCa bone and visceral metastases, activating paracrine Shh signaling in tumor-stromal interactions. MAOA provides tumor cell growth advantages in the bone microenvironment by stimulating interleukin-6 (IL6) release from osteoblasts, and triggers skeletal colonization by activating osteoclastogenesis through osteoblast production of RANKL and IL6. MAOA inhibitor treatment effectively reduces metastasis and prolongs mouse survival by disengaging the Shh-IL6-RANKL signaling network in stromal cells in the tumor microenvironment. These findings provide a rationale for targeting MAOA and its associated molecules to treat PCa metastasis.


Subject(s)
Cell Communication , Hedgehog Proteins/physiology , Interleukin-6/physiology , Monoamine Oxidase/physiology , Prostatic Neoplasms/pathology , RANK Ligand/physiology , Signal Transduction/physiology , Animals , Bone Neoplasms/secondary , Humans , Male , Mice , Mice, SCID , Monoamine Oxidase/analysis , Osteoblasts/physiology , Stromal Cells/physiology , Tumor Microenvironment
3.
Oncotarget ; 7(51): 84645-84657, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27835867

ABSTRACT

Lethal progression of prostate cancer metastasis can be improved by developing animal models that recapitulate the clinical conditions. We report here that cytokeratin 13 (KRT13), an intermediate filament protein, plays a directive role in prostate cancer bone, brain, and soft tissue metastases. KRT13 expression was elevated in bone, brain, and soft tissue metastatic prostate cancer cell lines and in primary and metastatic clinical prostate, lung, and breast cancer specimens. When KRT13 expression was determined at a single cell level in primary tumor tissues of 44 prostate cancer cases, KRT13 level predicted bone metastasis and the overall survival of prostate cancer patients. Genetically enforced KRT13 expression in human prostate cancer cell lines drove metastases toward mouse bone, brain and soft tissues through a RANKL-independent mechanism, as KRT13 altered the expression of genes associated with EMT, stemness, neuroendocrine/neuromimicry, osteomimicry, development, and extracellular matrices, but not receptor activator NF-κB ligand (RANKL) signaling networks in prostate cancer cells. Our results suggest new inhibitors targeting RANKL-independent pathways should be developed for the treatment of prostate cancer bone and soft tissue metastases.


Subject(s)
Adenocarcinoma/metabolism , Bone Neoplasms/metabolism , Brain Neoplasms/metabolism , Keratin-13/metabolism , Prostatic Neoplasms/metabolism , Adenocarcinoma/secondary , Animals , Bone Neoplasms/secondary , Brain Neoplasms/secondary , Cell Line, Tumor , Cell Movement , Cellular Reprogramming , Gene Expression Regulation, Neoplastic , Humans , Keratin-13/genetics , Male , Mice , Mice, SCID , Prognosis , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , RANK Ligand/metabolism , Survival Analysis , Transcriptome , Up-Regulation , Xenograft Model Antitumor Assays
4.
Oncotarget ; 6(42): 44072-83, 2015 Dec 29.
Article in English | MEDLINE | ID: mdl-26624980

ABSTRACT

FYN is a SRC family kinase (SFK) that has been shown to be up-regulated in human prostate cancer (PCa) tissues and cell lines. In this study, we observed that FYN is strongly up-regulated in human neuroendocrine PCa (NEPC) tissues and xenografts, as well as cells derived from a NEPC transgenic mouse model. In silico analysis of FYN expression in prostate cancer cell line databases revealed an association with the expression of neuroendocrine (NE) markers such as CHGA, CD44, CD56, and SYP. The loss of FYN abrogated the invasion of PC3 and ARCaPM cells in response to MET receptor ligand HGF. FYN also contributed to the metastatic potential of NEPC cells in two mouse models of visceral metastasis with two different cell lines (PC3 and TRAMPC2-RANKL). The activation of MET appeared to regulate neuroendocrine (NE) features as evidenced by increased expression of NE markers in PC3 cells with HGF. Importantly, the overexpression of FYN protein in DU145 cells was directly correlated with the increase of CHGA. Thus, our data demonstrated that the neuroendocrine differentiation that occurs in PCa cells is, at least in part, regulated by FYN kinase. Understanding the role of FYN in the regulation of NE markers will provide further support for ongoing clinical trials of SFK and MET inhibitors in castration-resistant PCa patients.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Differentiation , Cell Movement , Liver Neoplasms/enzymology , Neuroendocrine Tumors/enzymology , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins c-fyn/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Chromogranin A/metabolism , Computer Simulation , Databases, Genetic , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/pharmacology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Male , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Neoplasm Invasiveness , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/secondary , Phenotype , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , Time Factors , Transfection , Tumor Burden , Up-Regulation
5.
Transl Androl Urol ; 4(4): 438-54, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26816842

ABSTRACT

Prostate cancer (PCa) metastasizes to bone and soft tissues, greatly decreasing quality of life, causing bone pain, skeletal complications, and mortality in PCa patients. While new treatment strategies are being developed, the molecular and cellular basis of PCa metastasis and the "cross-talk" between cancer cells and their microenvironment and crucial cell signaling pathways need to be successfully dissected for intervention. In this review, we introduce a new concept of the mechanism of PCa metastasis, the recruitment and reprogramming of bystander and dormant cells (DCs) by a population of metastasis-initiating cells (MICs). We provide evidence that recruited and reprogrammed DCs gain MICs phenotypes and can subsequently metastasize to bone and soft tissues. We show that MICs can also recruit and reprogram circulating tumor cells (CTCs) and this could contribute to cancer cell evolution and the acquisition of therapeutic resistance. We summarize relevant molecular signaling pathways, including androgen receptors (ARs) and their variants and growth factors (GFs) and cytokines that could contribute to the predilection of PCa for homing to bone and soft tissues. To understand the etiology and the biology of PCa and the effectiveness of therapeutic targeting, we briefly summarize the animal and cell models that have been employed. We also report our experience in the use of three-dimensional (3-D) culture and co-culture models to understand cell signaling networks and the use of these attractive tools to conduct drug screening exercises against already-identified molecular targets. Further research into PCa growth and metastasis will improve our ability to target cancer metastasis more effectively and provide better rationales for personalized oncology.

6.
Mol Cancer Ther ; 9(12): 3289-301, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21159612

ABSTRACT

As p53 loss of function (LOF) confers high-level drug resistance in neuroblastoma, p53-independent therapies might have superior activity in recurrent neuroblastoma. We tested the activity of vorinostat, a histone deacetylase inhibitor, and flavopiridol, a pan-Cdk inhibitor, in a panel of multidrug-resistant neuroblastoma cell lines that included lines with wild-type (wt) and transcriptionally active TP53 (n = 3), mutated (mt), and LOF TP53 (n = 4) or p14(ARF) deletion (n = 1). The combination of vorinostat and flavopiridol was synergistic and significantly more cytotoxic (P < 0.001) in cell lines with p53-LOF and in the clones stably transfected with dominant-negative p53 plasmids. Cell cycle analysis by flow cytometry showed prominent cell-cycle arrest in G(2)/M (37%) for a cell line with wt TP53 (SK-N-RA) at 16 to 20 hours, while cells with mt TP53 (CHLA-90) slipped into sub-G(1) at 6 to 24 hours (25%-40% specific cell death). The morphological hallmarks of mitotic cell death, including defective spindle formation and abnormal cytokinesis, were detected by confocal microscopy after the treatment with vorinostat + flavopiridol combination in CHLA-90. The combination caused reduction in the expression of G(2)/M proteins (cyclin B1, Mad2, MPM2) in 2 cell lines with mt TP53 but not in those with wt TP53. Plk1 expression was reduced in all treated lines. Small interfering RNA knockdown of Mad2 and cyclin B1 or Plk1 synergistically reduced the clonogenicity of CHLA-90 cells. The combination of HDAC inhibitor and flavopiridol may be a unique approach to treating neuroblastomas with p53 LOF, one that evokes induction of mitotic failure.


Subject(s)
Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Flavonoids/pharmacology , Hydroxamic Acids/pharmacology , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Piperidines/pharmacology , Tumor Suppressor Protein p53/genetics , Biomarkers, Tumor/metabolism , Cell Death/drug effects , Cell Line, Tumor , Drug Synergism , Flavonoids/chemistry , Flavonoids/therapeutic use , G2 Phase/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/therapeutic use , Mitosis/drug effects , Mutant Proteins/metabolism , Mutation/genetics , Piperidines/chemistry , Piperidines/therapeutic use , Protein Kinase Inhibitors/pharmacology , Vorinostat
7.
J Biol Chem ; 280(17): 16821-8, 2005 Apr 29.
Article in English | MEDLINE | ID: mdl-15664986

ABSTRACT

TFII-I is a signal-induced multi-functional transcription factor that has recently been implicated as a regulatory component of the endoplasmic reticulum (ER) stress response. TFII-I acts through ER stress-induced binding to the ER stress element, which is highly conserved in promoters of ER stress-inducible genes such as Grp78/BiP. Interestingly, its tyrosine phosphorylation sites are required for its activation of the Grp78 promoter. Toward understanding the link between TFII-I, the tyrosine kinase signaling pathway, and Grp78 activation, we discovered that Tg stress induces a dramatic increase of TFII-I phosphorylation at Tyr248 and localization of this form of TFII-I to the nucleus. Chromatin immunoprecipitation analysis further reveals enhanced TFII-I binding to the Grp78 promoter in vivo upon ER stress. Previously, we reported that genistein, a general inhibitor of tyrosine kinase, could suppress ER stress induction of Grp78 by inhibiting complex formation on the ER stress element; however, the mechanism is not known. Consistent with TFII-I being a target of genistein suppression, we observed that genistein could suppress Tg stress-induced phosphorylation of TFII-I. We further demonstrate that c-Src, which is one of kinases identified to mediate phosphorylation of TFII-I at Tyr248, is activated by Tg stress and is able to stimulate the Grp78 promoter activity. Lastly, using stable cell lines with suppressed TFII-I levels, we show that TFII-I is required for optimal induction of Grp78 by ER stress. Our studies provide a molecular link that connects the c-Src tyrosine kinase transduction pathway to ER stress-induced transcriptional activation of Grp78 mediated by TFII-I.


Subject(s)
Endoplasmic Reticulum/metabolism , Heat-Shock Proteins/physiology , Molecular Chaperones/physiology , Transcription Factors, TFII/physiology , Transcription, Genetic , Tyrosine/metabolism , Animals , Blotting, Northern , Blotting, Western , CSK Tyrosine-Protein Kinase , Cell Line , Chromatin Immunoprecipitation , Endoplasmic Reticulum Chaperone BiP , Genes, Reporter , Genistein/pharmacology , Heat-Shock Proteins/metabolism , Humans , Kinetics , Mice , Microscopy, Fluorescence , Models, Biological , Models, Genetic , Molecular Chaperones/metabolism , NIH 3T3 Cells , Phosphorylation , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Time Factors , Transcriptional Activation , Transfection , Tyrosine/chemistry , src-Family Kinases
8.
J Biol Chem ; 279(12): 11354-63, 2004 Mar 19.
Article in English | MEDLINE | ID: mdl-14699159

ABSTRACT

ATF6 is a key transcriptional activator of the unfolded protein response (UPR), which allows mammalian cells to maintain cellular homeostasis when they are subjected to a variety of environmental and physiological stresses that target the endoplasmic reticulum (ER). ATF6, a 90-kDa ER transmembrane protein, contains three evolutionarily conserved N-linked glycosylation sites within its carboxyl luminal domain. Although it is well established that p90ATF6 activation requires transit from the ER to the Golgi, where it is cleaved by the S1P/S2P protease system to generate a nuclear form p60ATF6 that acts as a transcriptional activator, the functional significance of p90ATF6 N-linked glycosylation is unknown. Here we show that ER Ca(2+) depletion stress, a triggering mechanism for the UPR, induces the formation of ATF6(f), which represents de novo partial glycosylation of newly synthesized p90ATF6. By mutating a single amino acid within the N-linked glycosylation site closest to the carboxyl terminus of p90ATF6, we recreated ATF6(f). This mutation sharply reduces p90ATF6 association with calreticulin, a major Ca(2+)-binding chaperone for N-glycoprotein. We further determined that ATF6(f) exhibits a faster rate of constitutive transport to the Golgi, resulting in a higher level of p60ATF6 in the nucleus and stronger transactivating activity in the absence of ER stress. Additional analysis of p90ATF6 mutants targeting single or multiple N-glycosylation sites also showed higher constitutive transactivating activity than wild type ATF6. Because accumulation of underglycosylated proteins in the ER is a potent inducer for the UPR, these studies uncover a novel mechanism whereby the glycosylation status of p90ATF6 can serve as a sensor for ER homeostasis, resulting in ATF6 activation to trigger the UPR.


Subject(s)
DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Activating Transcription Factor 6 , Animals , Base Sequence , Calcium/metabolism , Cell Line , DNA Primers , DNA-Binding Proteins/genetics , Endoplasmic Reticulum/metabolism , Fluorescent Antibody Technique , Glycosylation , Homeostasis , Humans , Mutation , Protein Denaturation , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
9.
Biochem Biophys Res Commun ; 306(4): 912-7, 2003 Jul 11.
Article in English | MEDLINE | ID: mdl-12821128

ABSTRACT

A novel truncated form of Bcl-2, termed Bcl-2psi, was discovered in invasive prostate cancer cells, using laser capture microdissection, RNA-polymerase cycling reaction, and microarray analysis. The expression of Bcl-2psi increased prior to metastasis in higher-grade prostate cancer. The immunoreactive Bcl-2psi was specifically identified in higher-grade prostate cancer cells. These findings suggest that Bcl-2psi may be a potential pre-metastatic marker for detection, diagnosis, and therapy during the initiation of metastasis in prostate cancer.


Subject(s)
Biomarkers, Tumor , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/chemistry , Base Sequence , Blotting, Northern , Blotting, Western , Dissection , Electrophoresis, Polyacrylamide Gel , Gene Library , Humans , Immunohistochemistry , Lasers , Male , Molecular Sequence Data , Mutation , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Protein Structure, Tertiary , RNA/metabolism , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...