Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Hepatology ; 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39348425

ABSTRACT

BACKGROUND AND AIMS: Patients with aggressive hepatocellular carcinoma (HCC) have limited therapeutic options. Therefore, a better understanding of HCC pathogenesis is needed to improve treatment. Genomic studies of HCC have improved our understanding of cancer biology. However, the ubiquitomic characteristics of HCC remain poorly understood. We aimed to reveal the ubiquitomic characteristics of HCC and provide clinical feature biomarkers of the aggressive HCC that may be used for diagnosis or therapy in the clinic. APPROACH AND RESULTS: The comprehensive proteomic, phosphoproteomic, and ubiquitomic analyses were performed on tumors and adjacent normal liver tissues from 85 HCC patients. HCCs displayed overexpression of drugable targets CBR1-S151 and CPNE1-S55. COL4A1, LAMC1 and LAMA4 were highly expressed in the DFS poor patients. Phosphoproteomic and ubiquitomic features of HCC revealed crosstalk in metabolism and metastasis. Ubiquitomics predicted diverse prognosis and clarified HCC subtype-specific proteomic signatures. Expression of biomarkers TUBA1A, BHMT2, BHMT, and ACY1 exhibited differential ubiquitination levels and displayed high prognostic risk scores, suggesting that targeting these proteins or their modified forms may be beneficial for future clinical treatment. We validated that TUBA1A K370 deubiquitination drove severe HCC and labeled an aggressive subtype of HCCs. TUBA1A K370 deubiquitination was at least partly attributed to AKT-mediated USP14 activation in HCC. Notably, targeting AKT-USP14-TUBA1A complex promoted TUBA1A degradation and blocked liver tumorigenesis in vivo. CONCLUSIONS: This study expands our knowledge of ubiquitomic signatures, biomarkers, and potential therapeutic targets in HCC.

2.
Heliyon ; 10(7): e28877, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38596087

ABSTRACT

Objective: To develop and validate nomograms for predicting the OS and CSS of patients with Solitary Hepatocellular Carcinoma (HCC). Methods: Using the TRIPOD guidelines, this study identified 5206 patients in the Surveillance, Epidemiology, and End Results (SEER) 17 registry database. All patients were randomly divided in a ratio of 7:3 into a training cohort (n = 3646) and a validation cohort (n = 1560), and the Chinese independent cohort (n = 307) constituted the external validation group. The prognosis-related risk factors were selected using univariate Cox regression analysis, and the independent prognostic factors of OS and CSS were identified using the Lasso-Cox regression model. The nomograms for predicting the OS and CSS of the patients were constructed based on the identified prognostic factors. Their prediction ability was evaluated using the concordance index (C-index), receiver operating characteristic (ROC) curve, and calibration curve in both the training and validation cohorts. Results: We identified factors that predict OS and CSS and constructed two nomograms based on the data. The ROC analysis, C-index analysis, and calibration analysis indicated that the two nomograms performed well over the 1, 3, and 5-year OS and CSS periods in both the training and validation cohorts. Additionally, these results were confirmed in the external validation group. Decision curve analysis (DCA) demonstrated that the two nomograms were clinically valuable and superior to the TNM stage system. Conclusion: We established and validated nomograms to predict 1,3, and 5-year OS and CSS in solitary HCC patients, and our results may also be helpful for clinical decision-making.

3.
Cancer Lett ; 566: 216257, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37277019

ABSTRACT

The incidence rate of human hepatocellular carcinoma (HCC) is approximately three times higher in males than in females. A better understanding of the mechanisms underlying HCC development in males could lead to more effective therapies for HCC. Our previous study found that FBXW10 played a critical role in promoting HCC development in male mice and patients, but the mechanism remains unknown. Here, we found that FBXW10 promoted K63-linked ANXA2 polyubiquitination and activation in HCC tissues from males, and this process was required for S6K1-mediated phosphorylation. Activated ANXA2 further translocated from the cytoplasm to the cell membrane to bind KRAS and then activated the MEK/ERK pathway, leading to HCC proliferation and lung metastasis. Interfering with ANXA2 significantly blocked FBXW10-driven HCC growth and lung metastasis in vitro and in vivo. Notably, membrane ANXA2 was upregulated and positively correlated with FBXW10 expression in male HCC patients. These findings offer new insights into the regulation and function of FBXW10 signaling in HCC tumorigenesis and metastasis and suggest that the FBXW10-S6K1-ANXA2-KRAS-ERK axis may serve as a potential biomarker and therapeutic target in male HCC patients with high FBXW10 expression.


Subject(s)
Annexin A2 , Carcinoma, Hepatocellular , F-Box Proteins , Liver Neoplasms , Lung Neoplasms , Female , Humans , Male , Animals , Mice , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Annexin A2/genetics , Annexin A2/metabolism , F-Box Proteins/genetics , F-Box Proteins/metabolism
4.
J Biol Chem ; 284(36): 24006-16, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19592489

ABSTRACT

Interleukin (IL)-23, a new member of the IL-12 family, plays a central role in the Th17 immune response and in autoimmune diseases. It is clear that activated macrophages and dendritic cells produce IL-23, but the molecular mechanisms whereby inflammatory signals stimulate IL-23 expression are not fully understood. We demonstrate that induction of IL-23 p19 gene expression by LPS depends on the TLR4 and MyD88 pathways. All three MAPK pathways (ERK, JNK, and p38) that are activated by lipopolysaccharide (LPS) stimulation were shown to exert a positive effect on p19 expression. We cloned a 1.3-kb putative p19 promoter and defined its transcription initiation sites by the 5'-rapid amplification of cDNA ends method. By analyzing IL-23 p19 promoter mutants, we have identified a promoter region (-413 to +10) that contains several important elements, including NF-kappaB and AP-1. In addition to NF-kappaB, we have demonstrated that the proximal AP-1 site is important for p19 promoter activation. Mutation of the AP-1 site resulted in the loss of p19 promoter activation. Electrophoretic mobility shift assay (EMSA) analysis showed that c-Jun and c-Fos bind to the AP-1 site, which was confirmed by a chromatin immunoprecipitation assay. Furthermore, co-transfection of c-Jun and ATF2 synergistically induced p19 promoter activation, and c-Jun and ATF2 formed a protein complex, demonstrated by co-immunoprecipitation. Finally, LPS-stimulated peritoneal macrophages from IL-10-deficient mice expressed significantly higher IL-23 p19 than macrophages from wild type mice, and the addition of recombinant IL-10 strongly inhibited LPS-induced p19 expression. Thus, this study suggests that MyD88-dependent Toll-like receptor signaling induces IL-23 p19 gene expression through both MAPKs and NF-kappaB.


Subject(s)
Gene Expression Regulation/physiology , Interleukin-23 Subunit p19/biosynthesis , Macrophages, Peritoneal/metabolism , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptor 4/metabolism , Transcription Factor AP-1/metabolism , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/immunology , Activating Transcription Factor 2/metabolism , Animals , Autoimmune Diseases , Cell Line , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation/drug effects , Interleukin-10/immunology , Interleukin-10/pharmacology , Interleukin-23 Subunit p19/genetics , Interleukin-23 Subunit p19/immunology , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/immunology , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/immunology , Proto-Oncogene Proteins c-jun/metabolism , Response Elements/genetics , Response Elements/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology
5.
Eur J Immunol ; 38(7): 1807-13, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18506885

ABSTRACT

IL-17 and IL-22 are typical cytokines produced by the Th17 T cell subset, but it is unclear if Th17 cytokines can be produced by other cell types. We demonstrate that IL-10-deficient and IL-10R-deficient macrophages stimulated with lipopolysaccharide produce high levels of IL-17 and IL-22. Addition of exogenous IL-10 to IL-10-deficient macrophages abolished IL-17 production. When IL-10-deficient and IL-10R-deficient splenocytes were cultured under Th17 polarizing conditions, the population of IL-17-producing cells was increased and the cultures produced significantly higher levels of IL-17 and IL-22. The addition of recombinant IL-10 to IL-10-deficient splenocytes significantly decreased the percentage of IL-17-producing CD4(+) T cells. Finally, the mRNA for the Th17 transcription factor retinoic acid-related orphan receptor (ROR)gammat was significantly elevated in IL-10-deficient spleen cells and macrophages. These data demonstrate that Th17 cytokines and RORgammat are also expressed in macrophages and that IL-10 negatively regulates the expression of Th17 cytokines and RORgammat by both macrophages and T cells.


Subject(s)
Interleukin-10/metabolism , Interleukin-17/metabolism , Interleukins/metabolism , Macrophages/immunology , T-Lymphocyte Subsets/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Interleukin-10/deficiency , Interleukin-10/immunology , Interleukin-17/immunology , Interleukins/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3 , Receptors, Interleukin-10/deficiency , Receptors, Interleukin-10/immunology , Receptors, Interleukin-10/metabolism , Receptors, Retinoic Acid/immunology , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/immunology , Receptors, Thyroid Hormone/metabolism , T-Lymphocyte Subsets/metabolism , Interleukin-22
6.
Exp Cell Res ; 314(1): 92-102, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17900565

ABSTRACT

Understanding prostate stem cells may provide insight into the origin of prostate cancer. Primary cells have been cultured from human prostate tissue but they usually survive only 15-20 population doublings before undergoing senescence. We report here that RC-170N/h/clone 7 cells, a clonal cell line from hTERT-immortalized primary non-malignant tissue-derived human prostate epithelial cell line (RC170N/h), retain multipotent stem cell properties. The RC-170N/h/clone 7 cells expressed a human embryonic stem cell marker, Oct-4, and potential prostate epithelial stem cell markers, CD133, integrin alpha2beta1(hi) and CD44. The RC-170N/h/clone 7 cells proliferated in KGM and Dulbecco's Modified Eagle Medium with 10% fetal bovine serum and 5 microg/ml insulin (DMEM+10% FBS+Ins.) medium, and differentiated into epithelial stem cells that expressed epithelial cell markers, including CK5/14, CD44, p63 and cytokeratin 18 (CK18); as well as the mesenchymal cell markers, vimentin, desmin; the neuron and neuroendocrine cell marker, chromogranin A. Furthermore the RC170 N/h/clone 7 cells differentiated into multi tissues when transplanted into the sub-renal capsule and subcutaneously of NOD-SCID mice. The results indicate that RC170N/h/clone 7 cells retain the properties of multipotent stem cells and will be useful as a novel cell model for studying the mechanisms of human prostate stem cell differentiation and transformation.


Subject(s)
Cell Differentiation/genetics , Epithelial Cells/metabolism , Multipotent Stem Cells/metabolism , Prostate/metabolism , Telomerase/genetics , Aged , Animals , Antigens, Differentiation/biosynthesis , Biomarkers/analysis , Biomarkers/metabolism , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Line, Transformed , Cell Lineage/drug effects , Cell Lineage/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cellular Senescence/drug effects , Cellular Senescence/genetics , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/metabolism , Culture Media/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Graft Survival/drug effects , Graft Survival/genetics , Humans , Male , Mice , Mice, SCID , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Phenotype , Prostate/cytology , Stem Cell Transplantation/methods , Telomerase/metabolism , Telomerase/pharmacology , Transplantation, Heterologous/physiology
7.
Int J Oncol ; 31(3): 537-44, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17671680

ABSTRACT

Although epidemiological studies have suggested a positive correlation between environmental radon exposure and prostate cancer, the mechanism involved is not clear. In the present study, we examined the oncogenic transforming potency of alpha-particles using non-tumorigenic, telomerase-immortalized human benign prostate epithelial cells. We report the malignant transformation of human benign prostate epithelial cells after a single exposure to 0.6 Gy dose of alpha-particles. Transformed cells showed anchorage-independent growth in soft agar and induced progressively growing tumors when transplanted into SCID mice. The tumors were characterized histologically as poorly differentiated adenocarcinomas. The cell line derived from tumor (SCID 5015), like the unirradiated cells, expressed cytokeratin 5, 8 and 18, NKX3.1 and AMACR. The malignant cells showed increased secretion of MMP2. Stepwise chromosomal changes in the progression to tumorigenicity were observed. Chromosome abnormalities were identified in both irradiated and tumorigenic cells relative to the non-irradiated control cells. Prominent changes in chromosomes 6, 11 and 16, as well as mutations and deletions of the p53 gene were observed in the tumor outgrowth and tumor cells. These findings provide the first evidence of malignant transformation of human benign prostate epithelial cells exposed to a single dose of alpha-particles. This model provides an opportunity to study the cellular and molecular alterations that occur in radiation carcinogenesis in human prostate cells.


Subject(s)
Adenocarcinoma/diagnosis , Adenocarcinoma/etiology , Alpha Particles , Cell Transformation, Neoplastic , Linear Energy Transfer , Neoplasms, Radiation-Induced/etiology , Prostate/pathology , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/etiology , Adenocarcinoma/pathology , Animals , Cell Line, Transformed , Chromosome Aberrations , Humans , Male , Mice , Mice, SCID , Neoplasm Transplantation , Prostatic Neoplasms/pathology
8.
Cancer Res ; 67(7): 3153-61, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17409422

ABSTRACT

Understanding normal and cancer stem cells may provide insight into the origin of and new therapeutics for prostate cancer. Normal and cancer stem cells in prostate have recently been identified with a CD44(+)/alpha(2)beta(1)(high)/CD133(+) phenotype. Stromal cell-derived factor-1 (SDF-1) and its receptor, CXCR4, have multiple essential functions, including homing of stem cells and metastasis of cancer cells. We show here that human telomerase reverse transcriptase (hTERT)-immortalized primary nonmalignant (RC-165N/hTERT) and malignant (RC-92a/hTERT) tumor-derived human prostate epithelial cell lines retain stem cell properties with a CD133(+)/CD44(+)/alpha(2)beta(1)(+)/34betaE12(+)/CK18(+)/p63(-)/androgen receptor (AR)(-)/PSA(-) phenotype. Higher CD133 expression was detected in the hTERT-immortalized cells than in primary prostate cells. These immortalized cells exhibited "prostaspheres" in nonadherent culture systems and also maintained higher CD133 expression. The CD133(+) cells from these immortalized cell lines had high proliferative potential and were able to differentiate into AR(+) phenotype. In three-dimensional culture, the CD133(+) cells from RC-165N/hTERT cells produced branched structures, whereas the CD133(+) cells from RC-92a/hTERT cells produced large irregular spheroids with less branched structures. SDF-1 induced, but anti-CXCR4 antibody inhibited, migration of CD133(+) cells from RC-92a/hTERT cells, which coexpressed CXCR4. CXCR4/SDF-1 may sustain tumor chemotaxis in cancer stem cells. Furthermore, immunostaining of clinical prostate specimens showed that CD133 expression was detected in a subpopulation of prostate cancer cells and corresponded to the loss of AR. Expression of CXCR4 was also detected in CD133(+) cancer cells. These novel in vitro models may offer useful tools for the study of the biological features and functional integration of normal and cancer stem cells in prostate.


Subject(s)
Antigens, CD/biosynthesis , Glycoproteins/biosynthesis , Neoplastic Stem Cells/metabolism , Prostatic Neoplasms/metabolism , Receptors, CXCR4/biosynthesis , Telomerase/metabolism , AC133 Antigen , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cell Movement/physiology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Male , Neoplastic Stem Cells/pathology , Peptides , Prostatic Neoplasms/pathology , Tumor Cells, Cultured
9.
Exp Cell Res ; 312(6): 831-43, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16413016

ABSTRACT

In vitro human prostate cell culture models are critical for clarifying the mechanism of prostate cancer progression and for testing preventive and therapeutic agents. Cell lines ideal for the study of human primary prostate tumors would be those derived from spontaneously immortalized tumor cells; unfortunately, explanted primary prostate cells survive only short-term in culture, and rarely immortalize spontaneously. Therefore, we recently have generated five immortal human prostate epithelial cell cultures derived from both the benign and malignant tissues of prostate cancer patients with telomerase, a gene that prevents cellular senescence. Examination of these cell lines for their morphologies and proliferative capacities, their abilities to grow in low serum, to respond to androgen stimulation, to grow above the agar layer, to form tumors in SCID mice, suggests that they may serve as valid, useful tools for the elucidation of early events in prostate tumorigenesis. Furthermore, the chromosome alterations observed in these immortalized cell lines expressing aspects of the malignant phenotypes imply that these cell lines accurately recapitulate the genetic composition of primary tumors. These novel in vitro models may offer unique models for the study of prostate carcinogenesis and also provide the means for testing both chemopreventive and chemotherapeutic agents.


Subject(s)
DNA-Binding Proteins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Telomerase/metabolism , Tumor Cells, Cultured , Adult , Aged , Animals , Cell Aggregation/physiology , Cell Proliferation , Chromosomes, Human/genetics , Cytogenetic Analysis , Dihydrotestosterone/pharmacology , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Mice , Mice, SCID , Middle Aged , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Phenotype , Racemases and Epimerases/genetics , Receptors, Androgen/drug effects , Receptors, Androgen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
10.
Anticancer Res ; 25(1A): 1-8, 2005.
Article in English | MEDLINE | ID: mdl-15816512

ABSTRACT

The generation of suitable in vitro models is critical for understanding the process associated with the development and progression of prostate cancer in high-risk African-American men. However, the generation of long-term human prostate epithelial cell lines derived from primary human prostate epithelium have been unsuccessful due to the absence of in vitro immortalization. We have successfully established an immortal human prostate epithelial cell line from primary benign tissues of African-American prostate cancer patients by using telomerase. The actively proliferating secondary African-American prostate epithelial RC-165N cells, derived from benign prostate tissue of a radical prostatectomy specimen, were transduced through infection with a retrovirus vector expressing the human telomerase catalytic subunit (hTERT). A high level of telomerase activity was detected in RC-165N/hTERT cells but not in RC-165N cells. RC-165N/hTERT cells are currently growing well at passage 50 whereas RC-165N cells senesced within passage 3. RC-165N/hTERT cells exhibit epithelial morphology. These immortalized cells showed no cell growth in soft agar, and no tumor formation in SCID mice. The RC-165N/hTERT cells express androgen-regulated prostate-specific homobox gene. NKX 3.1 and epithelial cell specific cytokeratin 8, androgen receptor (AR), prostate stem cell antigen and p16, but not PSA. AR protein was detected by Western blot analysis.


Subject(s)
Androgen Antagonists/pharmacology , Androgen Receptor Antagonists , Black People , Cell Line , Prostate/cytology , Prostatic Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , DNA-Binding Proteins , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/physiology , Flutamide/pharmacology , Humans , Karyotyping , Male , Prostate/drug effects , Prostate/enzymology , Prostate/physiology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Telomerase/genetics , Telomerase/metabolism , Transfection
11.
Int J Oncol ; 25(4): 1057-64, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15375556

ABSTRACT

Understanding of molecular genetic mechanisms underlying prostate carcinogenesis would be greatly advanced by in vitro models of prostate tumors representing primary tumors. We have successfully established a neoplastic immortalized human prostate epithelial (HPE) clonal culture derived from a primary tumor of a prostate cancer patient (RC-58T) with hTERT, the catalytic subunit of telomerase. The early passage RC-58T cells derived from a radical prostatectomy specimen of a 52-year-old white male patient was transduced through infection with a retrovirus vector expressing the hTERT for the establishment of the RC-58T/hTERT cell line. One clonal line, soft-agar derived from the RC-58T/hTERT cell line, was isolated and further characterized phenotypically and genetically. These clonal (RC-58T/hTERT SA#4) cells are currently growing well at passage 70 and exhibit transformed morphology. The RC-58T/hTERT SA#4 line expressed a high level of telomerase activity and showed anchorage-independent growth in soft agar. The clonal line like the untransduced RC-58T cells (passage 3) expressed prostate specific antigen (PSA), androgen receptor (AR), prostate stem cell antigen (PSCA), and an androgen-regulated prostate specific gene NKX3.1, P16, and cytokeratin (CK) 8. Growth is slightly stimulated by dihydrotestosterone (DHT), and lyates are immunoreactive with AR antibody by Western blot analysis. More importantly, this clonal line produced adenocarcinomas when transplanted into SCID mice. A number of chromosome alterations were observed including the loss of chromosome Y, 1q, 2p, 3p, 4q, 8p, 11p, 14p, 17p and 18q. Our results demonstrate that this primary tumor-derived HPE cell line retained its neoplastic phenotypes and its prostate specific markers and should allow elucidating molecular and genetic alterations involved in prostate cancer. This is the first documented case of an AR and PSA expressing telomerase established human prostate cancer cell line with neoplastic phenotypes from a primary tumor of a prostate cancer patient.


Subject(s)
Prostatic Neoplasms/genetics , Telomerase/genetics , Animals , Blotting, Western , Cell Division , Cell Line, Tumor , Chromosome Aberrations , DNA-Binding Proteins , Dihydrotestosterone/pharmacology , Humans , Male , Mice , Mice, SCID , Middle Aged , Phenotype , Prostatic Neoplasms/pathology , Receptors, Androgen/analysis , Reverse Transcriptase Polymerase Chain Reaction
12.
Int J Oncol ; 22(6): 1311-7, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12738999

ABSTRACT

Research into molecular and genetic mechanisms underlying prostate carcinogenesis would be greatly advanced by in vitro models of prostate tumors representing primary tumors. The generation of immortalized primary prostate cancer cells that will accurately reflect the in situ characteristics of malignant epithelium is greatly needed. We have successfully established a neoplastic immortalized human prostate epithelial (HPE) cell culture derived from a primary tumor. The RC-9 cells transduced through infection with a retrovirus vector expressing the E6 and E7 genes (E6E7) of human papilloma virus-16 (HPV-16) are currently growing well at passage 40, whereas RC-9 cells senesced at passage 7. RC-9/E6E7 cells exhibit epithelial morphology and high level of telomerase activity. More importantly, these immortalized cells produced tumors (SCID5038D) when inoculated into SCID mice. RC-9/E6E7 cells and SCID-5038D cells exhibit a high level of telomerase activity and androgen-responsiveness when treated with R1881. Expression of prostate specific antigen (PSA), androgen receptor (AR), prostate stem cell antigen (PSCA), an androgen-regulated prostate specific gene (NKX3.1), p16, cytokeratins 8, 15 and HPV-16 E6 gene was detected in both of these cells. RC-9/E6E7 and SCID5038D cells also showed growth inhibition when exposed to retinoic acid and transforming growth factor (TGF)-beta1, potent inhibitors of prostate epithelial cell growth. A number of chromosome alterations were observed including the loss of chromosomes 2p, 3p, 8p, 13, 14, 16, 17, 18, 21 and the gain of 7 and 20 in the tumor cell line (SCID5038D). These results demonstrate that this primary tumor-derived HPE cell line retained its neoplastic phenotypes and its prostate-specific markers and should allow studies to elucidate molecular and genetic alterations involved in prostate cancer. This is the first documented case of a malignant AR and PSA positive established human prostate cancer cell line from a primary tumor of a prostate cancer patient.


Subject(s)
Epithelial Cells/cytology , Prostate/cytology , Prostatic Neoplasms/pathology , Animals , Cell Culture Techniques/methods , Cell Division/drug effects , Epithelial Cells/pathology , Humans , Karyotyping , Male , Mice , Mice, SCID , Middle Aged , Prostate/pathology , Prostatectomy , Prostatic Neoplasms/genetics , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/genetics , Transplantation, Heterologous , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL