Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Transl Res ; 237: 1-15, 2021 11.
Article in English | MEDLINE | ID: mdl-34004371

ABSTRACT

The 7 members of the A3 family of cytidine deaminases (A3A to A3H) share a conserved catalytic activity that converts cytidines in single-stranded (ss) DNA into uridines, thereby inducing mutations. After their initial identification as cell-intrinsic defenses against HIV and other retroviruses, A3s were also found to impair many additional viruses. Moreover, some of the A3 proteins (A3A, A3B, and A3H haplotype I) are dysregulated in cancer cells, thereby causing chromosomal mutations that can be selected to fuel progression of malignancy. Viral mechanisms that increase transcription of A3 genes or induce proteasomal degradation of A3 proteins have been characterized. However, only a few underlying biological mechanisms regulating levels of A3s in uninfected cells have been described. Here, we characterize that the von Hippel-Lindau tumor suppressor (pVHL), via its CRLpVHL, induces degradation of all 7 A3 proteins. Two independent lines of evidence supported the conclusion that the multiprotein CRLpVHL complex is necessary for A3 degradation. CRLpVHL more effectively induced degradation of nuclear, procancer A3 (A3B) than the cytoplasmic, antiretroviral A3 (A3G). These results identify specific cellular factors that regulate A3s post-translationally.


Subject(s)
APOBEC Deaminases/metabolism , Gene Expression Regulation, Enzymologic/physiology , Ubiquitin-Protein Ligases/metabolism , APOBEC Deaminases/genetics , Cell Line, Tumor , Down-Regulation , Humans , Ubiquitin-Protein Ligases/genetics
2.
Curr Opin Pharmacol ; 53: 66-76, 2020 08.
Article in English | MEDLINE | ID: mdl-32777746

ABSTRACT

CD73 (ecto-5'-nucleotidase) is a novel immunoinhibitory protein that plays a key role for tumor growth and metastasis. Its main function is to convert extracellular ATP to immunosuppressive adenosine in concert with CD39 in normal tissues to limit excessive immune response. However, tumors take advantage of the CD73-mediated adenosinergic mechanism to protect them from immune attack. In particular, inducible expression of CD73 along with other adenosinergic molecules on both cancer cells and host cells sustains immunosuppressive tumor microenvironment by affecting multiple aspects of the immune response. Owing to its multifaceted capacity to tumor promotion as an emerging immune checkpoint, CD73 is an ideal therapeutic target for cancer treatment especially in combination with conventional therapy and/or other immune checkpoint inhibitors. In this review, we will discuss the roles of CD73 on tumor and immune cells and will highlight the therapeutic value of CD73 for combination therapy.


Subject(s)
5'-Nucleotidase/immunology , Immunotherapy , Neoplasms/therapy , Animals , Combined Modality Therapy , Humans , Neoplasms/immunology
3.
Oncotarget ; 8(31): 51264-51276, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28881646

ABSTRACT

Genomic studies are rapidly identifying genetic alterations in human cancer, but functional validation of such alterations has been slow. Here, using human prostate cancer as a model, we have assessed the feasibility of engineering defined genetic alterations in well-known cancer driver genes to transform benign prostate epithelial organoids derived from African American men. Benign human prostate organoids were transduced with lentiviruses expressing MYC, shPTEN, shTP53 and AR, alone and in various combinations, to recapitulate prostate cancer development. Organoids expressing MYC, shPTEN, shTP53 and AR (denoted MPPA); MYC, shPTEN and shTP53 (MPP); or MYC (M) were significantly larger, had higher proliferation rates and demonstrated pathologically transformed morphology compared to organoids transduced with control lentivirus. Alterations in MYC, PTEN and TP53 also affected the rate of organoid basal-to-luminal differentiation in vitro. MPPA and MPP organoids expressed the clinical prostate cancer marker AMACR and developed prostate adenocarcinoma when grafted under the renal capsule in mice. These data indicate that genetic alterations commonly observed in human prostate cancer can be rapidly modeled in human organoid culture. Prostate cancer organoids provide a useful pre-clinical model for the evaluation of new candidate cancer genes, cancer disparities, and potentially for testing of novel therapeutic agents.

4.
Nat Commun ; 7: 12943, 2016 10 05.
Article in English | MEDLINE | ID: mdl-27703144

ABSTRACT

Identification of defined cell populations with stem/progenitor properties is key for understanding prostate development and tumorigenesis. Here we show that the polycomb repressor protein Bmi1 marks a population of castration-resistant luminal epithelial cells enriched in the mouse proximal prostate. We employ lineage tracing to show that these castration-resistant Bmi1-expressing cells (or CARBs) are capable of tissue regeneration and self-renewal. Notably, CARBs are distinct from the previously described luminal castration-resistant Nkx3.1-expressing cells (CARNs). CARBs can serve as a prostate cancer cell-of-origin upon Pten deletion, yielding luminal prostate tumours. Clonal analysis using the R26R-confetti allele indicates preferential tumour initiation from CARBs localized to the proximal prostate. These studies identify Bmi1 as a marker for a distinct population of castration-resistant luminal epithelial cells enriched in the proximal prostate that can serve as a cell of origin for prostate cancer.


Subject(s)
Polycomb Repressive Complex 1/metabolism , Prostate/cytology , Prostatic Neoplasms, Castration-Resistant/metabolism , Proto-Oncogene Proteins/metabolism , Regeneration , Alleles , Androgens/metabolism , Animals , Cell Lineage , Cell Transformation, Neoplastic/pathology , Epithelial Cells/metabolism , Male , Mice , Neoplastic Stem Cells/metabolism , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Recombination, Genetic
5.
Cancer Res ; 76(13): 3802-12, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27197174

ABSTRACT

Sirtuins participate in sensing nutrient availability and directing metabolic activity to match energy needs with energy production and consumption. However, the pivotal targets for sirtuins in cancer are mainly unknown. In this study, we identify the M2 isoform of pyruvate kinase (PKM2) as a critical target of the sirtuin SIRT2 implicated in cancer. PKM2 directs the synthesis of pyruvate and acetyl-CoA, the latter of which is transported to mitochondria for use in the Krebs cycle to generate ATP. Enabled by a shotgun mass spectrometry analysis founded on tissue culture models, we identified a candidate SIRT2 deacetylation target at PKM2 lysine 305 (K305). Biochemical experiments including site-directed mutants that mimicked constitutive acetylation suggested that acetylation reduced PKM2 activity by preventing tetramerization to the active enzymatic form. Notably, ectopic overexpression of a deacetylated PKM2 mutant in Sirt2-deficient mammary tumor cells altered glucose metabolism and inhibited malignant growth. Taken together, our results argued that loss of SIRT2 function in cancer cells reprograms their glycolytic metabolism via PKM2 regulation, partially explaining the tumor-permissive phenotype of mice lacking Sirt2 Cancer Res; 76(13); 3802-12. ©2016 AACR.


Subject(s)
Breast Neoplasms/pathology , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Glucose/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Neoplasm Recurrence, Local/pathology , Sirtuin 2/physiology , Thyroid Hormones/chemistry , Thyroid Hormones/metabolism , Acetylation , Animals , Blotting, Western , Breast Neoplasms/metabolism , Cell Proliferation , Female , Fluorescent Antibody Technique , Glycolysis , Humans , Immunoenzyme Techniques , Mice , Mice, Knockout , Neoplasm Recurrence, Local/metabolism , Neoplasm Staging , Prognosis , Tissue Array Analysis , Tumor Cells, Cultured , Thyroid Hormone-Binding Proteins
6.
Clin Cancer Res ; 20(12): 3211-21, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24771642

ABSTRACT

PURPOSE: To identify genes whose depletion is detrimental to Pim1-overexpressing prostate cancer cells and to validate this finding in vitro and in vivo. EXPERIMENTAL DESIGN: RNAi screening was used to identify genes whose depletion is detrimental to Pim1-overexpressing cells. Our finding was validated using shRNA or PLK1-specific inhibitor BI 2536. Xenograft studies were performed using both PLK1-knockdown cells and BI 2536 to investigate the effects of PLK1 inhibition on tumorigenesis in Pim1-overexpressing cells. Finally, PLK1 and PIM1 expression patterns in human prostate tumors were examined by immunohistochemistry using tissue microarrays. RESULTS: We identified the mitotic regulator polo-like kinase (PLK1) as a gene whose depletion is particularly detrimental to the viability of Pim1-overexpressing prostate cancer. Inhibition of PLK1 by shRNA or BI 2536 in Pim1-overexpressing prostate cancer xenograft models resulted in a dramatic inhibition of tumor progression. Notably, Pim1-overexpressing cells were more prone to mitotic arrest followed by apoptosis due to PLK1 inhibition than control cells. Furthermore, inhibition of PLK1 led to the reduction of MYC protein levels both in vitro and in vivo. Our data also suggest that PIM1 and PLK1 physically interact and PIM1 might phosphorylate PLK1. Finally, PLK1 and PIM1 are frequently co-expressed in human prostate tumors, and co-expression of PLK1 and PIM1 was significantly correlated to higher Gleason grades. CONCLUSIONS: Our findings demonstrate that PIM1-overexpressing cancer cells are particularly sensitive to PLK1 inhibition, suggesting that PIM1 might be used as a marker for identifying patients who will benefit from PLK1 inhibitor treatment.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , RNA Interference/physiology , RNA, Small Interfering/genetics , Animals , Blotting, Western , Cell Cycle Proteins/metabolism , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Immunoprecipitation , Male , Mice , Mice, Nude , Mitosis/physiology , Neoplasm Grading , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-pim-1/genetics , Tissue Array Analysis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
7.
J Clin Invest ; 122(5): 1907-19, 2012 May.
Article in English | MEDLINE | ID: mdl-22484818

ABSTRACT

Cooperativity between oncogenic mutations is recognized as a fundamental feature of malignant transformation, and it may be mediated by synergistic regulation of the expression of pro- and antitumorigenic target genes. However, the mechanisms by which oncogenes and tumor suppressors coregulate downstream targets and pathways remain largely unknown. Here, we used ChIP coupled to massively parallel sequencing (ChIP-seq) and gene expression profiling in mouse prostates to identify direct targets of the tumor suppressor Nkx3.1. Further analysis indicated that a substantial fraction of Nkx3.1 target genes are also direct targets of the oncoprotein Myc. We also showed that Nkx3.1 and Myc bound to and crossregulated shared target genes in mouse and human prostate epithelial cells and that Nkx3.1 could oppose the transcriptional activity of Myc. Furthermore, loss of Nkx3.1 cooperated with concurrent overexpression of Myc to promote prostate cancer in transgenic mice. In human prostate cancer patients, dysregulation of shared NKX3.1/MYC target genes was associated with disease relapse. Our results indicate that NKX3.1 and MYC coregulate prostate tumorigenesis by converging on, and crossregulating, a common set of target genes. We propose that coregulation of target gene expression by oncogenic/tumor suppressor transcription factors may represent a general mechanism underlying the cooperativity of oncogenic mutations during tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/physiology , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/physiology , Transcription Factors/physiology , Animals , Base Sequence , Binding Sites , Cell Line , Consensus Sequence , Gene Regulatory Networks , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Neoplasm Transplantation , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Rats , Sequence Analysis, DNA , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
8.
J Cell Physiol ; 227(2): 801-12, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21503880

ABSTRACT

Polyploidy has been linked to tumorigenicity mainly due to the chromosomal aberrations. Elevated reactive oxygen species (ROS) generation, on the other hand, has also been associated with oncogenic transformation in most cancer cells. However, a possible link between ploidy and ROS is largely unexplored. Here we have examined the role of ROS in the tumorigenicity of polyploid cells. We show that polyploid prostate and mammary epithelial cells contain higher levels of ROS due to their higher mitochondrial contents. ROS levels and mitochondrial mass are also higher in dihydrocytochalasin B (DCB)-induced polyploid cells, suggesting that higher levels of ROS observed in polyploid cell can occur due to cytokinesis failure. Interestingly, polyploid cells were more sensitive to the inhibitory effect of the antioxidant, N-Acetyl-L-cysteine (NAC), than control diploid cells. Treatment of polyploid/diploid cells with NAC led to the selective elimination of polyploid cells over time and abrogated the tumorigenicity of polyploid cells. This effect was partially mediated via the Akt signaling pathway. We next explored a possible role for ROS in promoting chromosomal instability by analyzing the effects of ROS on the mitotic stage of the cell cycle. Enhancing ROS levels by treating cells with hydrogen peroxide delayed not only entry into and but also exit from mitosis. Furthermore, increasing ROS levels significantly increased taxol resistance. Our results indicated that increased ROS in polyploid cells can contribute to tumorigenicity and highlight the therapeutic potential of antioxidants by selectively targeting the tumorigenic polyploid cells and by reversing taxol resistance.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Polyploidy , Reactive Oxygen Species/metabolism , Antineoplastic Agents/pharmacology , Cell Line , Drug Resistance, Neoplasm , Female , Humans , Hydrogen Peroxide , Male , Mammary Glands, Human/cytology , Mitochondria/physiology , Mitosis/drug effects , Mitosis/physiology , Paclitaxel/pharmacology , Prostate/cytology
9.
BMC Cancer ; 10: 248, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20515470

ABSTRACT

BACKGROUND: The serine/threonine kinase PIM1 has been implicated as an oncogene in various human cancers including lymphomas, gastric, colorectal and prostate carcinomas. In mouse models, Pim1 is known to cooperate with c-Myc to promote tumorigenicity. However, there has been limited analysis of the tumorigenic potential of Pim1 overexpression in benign and malignant human prostate cancer cells in vivo. METHODS: We overexpressed Pim1 in three human prostate cell lines representing different disease stages including benign (RWPE1), androgen-dependent cancer (LNCaP) and androgen-independent cancer (DU145). We then analyzed in vitro and in vivo tumorigenicity as well as the effect of Pim1 overexpression on c-MYC transcriptional activity by reporter assays and gene expression profiling using an inducible MYC-ER system. To validate that Pim1 induces tumorigenicity and target gene expression by modulating c-MYC transcriptional activity, we inhibited c-MYC using a small molecule inhibitor (10058-F4) or RNA interference. RESULTS: Overexpression of Pim1 alone was not sufficient to convert the benign RWPE1 cell to malignancy although it enhanced their proliferation rates when grown as xenografts in vivo. However, Pim1 expression enhanced the in vitro and in vivo tumorigenic potentials of the human prostate cancer cell lines LNCaP and DU145. Reporter assays revealed increased c-MYC transcriptional activity in Pim1-expressing cells and mRNA expression profiling demonstrated that a large fraction of c-MYC target genes were also regulated by Pim1 expression. The c-MYC inhibitor 10058-F4 suppressed the tumorigenicity of Pim1-expressing prostate cancer cells. Interestingly, 10058-F4 treatment also led to a reduction of Pim1 protein but not mRNA. Knocking-down c-MYC using short hairpin RNA reversed the effects of Pim1 on Pim1/MYC target genes. CONCLUSION: Our results suggest an in vivo role of Pim1 in promoting prostate tumorigenesis although it displayed distinct oncogenic activities depending on the disease stage of the cell line. Pim1 promotes tumorigenicity at least in part by enhancing c-MYC transcriptional activity. We also made the novel discovery that treatment of cells with the c-MYC inhibitor 10058-F4 leads to a reduction in Pim1 protein levels.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Transcriptional Activation , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-pim-1/genetics , RNA Interference , RNA, Messenger/metabolism , Receptors, Androgen/metabolism , Thiazoles/pharmacology , Time Factors , Transfection , Tumor Burden , Up-Regulation
11.
PLoS Genet ; 5(7): e1000542, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19578399

ABSTRACT

In human somatic tumorigenesis, mutations are thought to arise sporadically in individual cells surrounded by unaffected cells. This contrasts with most current transgenic models where mutations are induced synchronously in entire cell populations. Here we have modeled sporadic oncogene activation using a transgenic mouse in which c-MYC is focally activated in prostate luminal epithelial cells. Focal c-MYC expression resulted in mild pathology, but prostate-specific deletion of a single allele of the Pten tumor suppressor gene cooperated with c-MYC to induce high grade prostatic intraepithelial neoplasia (HGPIN)/cancer lesions. These lesions were in all cases associated with loss of Pten protein expression from the wild type allele. In the prostates of mice with concurrent homozygous deletion of Pten and focal c-MYC activation, double mutant (i.e. c-MYC+;Pten-null) cells were of higher grade and proliferated faster than single mutant (Pten-null) cells within the same glands. Consequently, double mutant cells outcompeted single mutant cells despite the presence of increased rates of apoptosis in the former. The p53 pathway was activated in Pten-deficient prostate cells and tissues, but c-MYC expression shifted the p53 response from senescence to apoptosis by repressing the p53 target gene p21(Cip1). We conclude that c-MYC overexpression and Pten deficiency cooperate to promote prostate tumorigenesis, but a p53-dependent apoptotic response may present a barrier to further progression. Our results highlight the utility of inducing mutations focally to model the competitive interactions between cell populations with distinct genetic alterations during tumorigenesis.


Subject(s)
Mutation , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Animals , Apoptosis , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Transgenic , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/physiopathology , Protein Binding , Proto-Oncogene Proteins c-myc/metabolism
12.
PLoS One ; 3(7): e2572, 2008 Jul 02.
Article in English | MEDLINE | ID: mdl-18596907

ABSTRACT

BACKGROUND: Polyploidy is a prominent feature of many human cancers, and it has long been hypothesized that polyploidy may contribute to tumorigenesis by promoting genomic instability. In this study, we investigated whether polyploidy per se induced by a relevant oncogene can promote genomic instability and tumorigenicity in human epithelial cells. PRINCIPAL FINDINGS: When the oncogenic serine-threonine kinase Pim-1 is overexpressed in immortalized, non-tumorigenic human prostate and mammary epithelial cells, these cells gradually converted to polyploidy and became tumorigenic. To assess the contribution of polyploidy to tumorigenicity, we obtained sorted, matched populations of diploid and polyploid cells expressing equivalent levels of the Pim-1 protein. Spectral karyotyping revealed evidence of emerging numerical and structural chromosomal abnormalities in polyploid cells, supporting the proposition that polyploidy promotes chromosomal instability. Polyploid cells displayed an intact p53/p21 pathway, indicating that the viability of polyploid cells in this system is not dependent on the inactivation of the p53 signaling pathway. Remarkably, only the sorted polyploid cells were tumorigenic in vitro and in vivo. CONCLUSIONS: Our results support the notion that polyploidy can promote chromosomal instability and the initiation of tumorigenesis in human epithelial cells.


Subject(s)
Breast Neoplasms/genetics , Epithelial Cells/metabolism , Mammary Glands, Human/metabolism , Polyploidy , Prostate/metabolism , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Carcinoma/genetics , Flow Cytometry , Genomic Instability , Humans , Male , Mammary Glands, Human/pathology , Mice , Proto-Oncogene Proteins c-pim-1/genetics , Rats , Rats, Sprague-Dawley , Telomerase/metabolism
13.
Genesis ; 44(10): 447-53, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17013838

ABSTRACT

The transcription factor c-MYC and the serine-threonine kinase Pim-1 have multiple roles in development and cancer, including in lymphomagenesis and prostate tumorigenesis. In some cancers, MYC and Pim-1 oncogenes are co-expressed and show marked cooperativity. To facilitate the analysis of the pathological roles of MYC and Pim-1 in specific cell types and developmental stages, we generated mice carrying Cre-inducible MYC/Pim-1 transgenes. The mice carry a constitutively expressed lacZ marker and silent MYC/Pim-1 genes. Cre-mediated recombination results in deletion of the lacZ marker and concurrent activation of the MYC/Pim-1 transgene. In addition, the Pim-1 mice harbor an alkaline phosphatase gene as a positive marker for recombination. Mouse lines for each gene were established, which show distinct patterns of expression in multiple tissues. In vivo recombination was confirmed for all lines by breeding to Cre transgenic mice. These mice provide a valuable resource for investigating the significance of MYC and Pim-1 overexpression in various tissues.


Subject(s)
Cell Transformation, Neoplastic/genetics , Integrases/metabolism , Proto-Oncogene Proteins c-myc/physiology , Proto-Oncogene Proteins c-pim-1/physiology , Animals , Female , Gene Expression Regulation, Neoplastic , Integrases/genetics , Lac Operon , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-pim-1/genetics , Recombination, Genetic , Tissue Distribution , Transgenes/physiology
14.
J Biol Chem ; 280(49): 40568-77, 2005 Dec 09.
Article in English | MEDLINE | ID: mdl-16221667

ABSTRACT

Overexpression of the oncogenic serine/threonine kinase Pim-1 has been shown to induce chromosomal missegregation and polyploidy in prostate epithelial cell lines (1). Here we demonstrated that Pim-1-induced polyploidy develops in a passage-dependent manner in culture consistent with a stochastic mode of progression. Induction of chromosomal instability by Pim-1 was not restricted to prostate cells as it was also observed in telomerase-immortalized normal human mammary epithelial cells. Elevated levels of cyclin B1 protein, but not its messenger RNA, were evident in early passage Pim-1 overexpressing cells, suggesting that increased cyclin B1 levels contribute to the development of polyploidy. Furthermore, regulation of cyclin B1 protein and cyclin B1/CDK1 activity after treatment with anti-microtubule agents was impaired. Small interfering RNA targeting cyclin B1 reversed the cytokinesis delay but not the mitotic checkpoint defect in Pim-1 overexpressing cells. These results indicated that chronic Pim-1 overexpression dysregulates cyclin B1 protein expression, which contributes to the development of polyploidy by delaying cytokinesis.


Subject(s)
Chromosomes, Human/drug effects , Cyclin B/genetics , Gene Expression Regulation/drug effects , Animals , CDC2 Protein Kinase/metabolism , Cattle , Cell Cycle , Cell Division , Cell Line, Tumor , Cyclin B/analysis , Cyclin B/physiology , Cyclin B1 , Cytokinesis , Gene Expression , Humans , Immunoblotting , Male , Mitosis , Nocodazole/pharmacology , Polyploidy , Prostatic Neoplasms , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/pharmacology , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transfection
15.
Biochim Biophys Acta ; 1743(3): 231-42, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15843037

ABSTRACT

Wild-type transforming growth factor alpha (TGFalpha) expression in lactotrope cells in the pituitary gland led to lactotrope-specific pituitary hyperplasia and adenomata. To indicate whether the EGF receptor is involved in this TGFalpha-mediated phenotype, we bred TGFalpha mice with mice expressing the cytoplasmic truncated-EGF receptor (EGFR-tr), which is dominant-negative in other models. These bitransgenic mice developed pituitary pathology despite expression of the dominant-negative receptor. To further characterize this observation, we generated two lineages of transgenic mice that overexpress mutant forms of TGFalpha: a processed soluble form (s TGFalpha) and a cytoplasmic-deleted form (TGFalphaDeltaC). While sTGFalpha expression in lactotrope cells failed to induce autocrine lactotrope hyperplasia, the pituitary became very enlarged due to proliferation of neighboring interstitial cells. In contrast, the TGFalphaDeltaC mice did not develop a phenotype, although the mRNA and protein were present in the pituitary and this form of TGFalpha was confirmed to be biologically active and targeted properly to the plasma membrane of cultured CHO cells. The results suggest that the cytoplasmic domain of TGFalpha is required for autocrine parenchymal tumor formation in the pituitary gland. This signal cannot be inhibited by the EGFR-tr. Conversely, the released form of TGFalpha appears to have primarily paracrine activity.


Subject(s)
ErbB Receptors/genetics , Pituitary Gland/metabolism , Transforming Growth Factor alpha/genetics , Animals , CHO Cells , Cell Division , Cell Membrane/metabolism , Coloring Agents , Cricetinae , Cytoplasm/metabolism , ErbB Receptors/biosynthesis , ErbB Receptors/metabolism , Hyperplasia , Mice , Mice, Transgenic , Mitotic Index , Phenotype , Pituitary Gland/pathology , Protein Structure, Tertiary , Signal Transduction , Solubility , Transforming Growth Factor alpha/biosynthesis , Transforming Growth Factor alpha/metabolism
16.
Cancer Res ; 63(23): 8079-84, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14678956

ABSTRACT

Aneuploidy and chromosomal aberrations are hallmarks of most human epithelial malignancies. Here we show that overexpression of the oncogenic kinase Pim-1 in human prostate epithelial cells induces genomic instability by subverting the mitotic spindle checkpoint. Cells overexpressing Pim-1 have a defect in the mitotic spindle checkpoint, abnormal mitotic spindles, centrosome amplification, and chromosome missegregation. Polyploidy and aneuploidy ensue due to a delay in completing cytokinesis. These results define a novel role for elevated Pim-1 expression in promoting genomic instability in human prostate tumors.


Subject(s)
Chromosome Segregation , Polyploidy , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Cell Cycle/physiology , Cell Line, Tumor , Epithelial Cells/enzymology , Epithelial Cells/pathology , Humans , Male , Mitosis/genetics , Prostate/enzymology , Prostate/physiology , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-pim-1 , Spindle Apparatus/genetics
17.
Mol Cell Endocrinol ; 194(1-2): 135-46, 2002 Aug 30.
Article in English | MEDLINE | ID: mdl-12242036

ABSTRACT

Type 2 diabetes mellitus results from a complex interaction between nutritional excess and multiple genes. Whereas pancreatic beta-cells normally respond to glucose challenge by rapid insulin release (first phase insulin secretion), there is a loss of this acute response in virtually all of the type 2 diabetes patients with significant fasting hyperglycemia. Our previous studies demonstrated that irreversible intracellular accumulation of a glucose metabolite, protein O-linked N-acetylglucosamine modification (O-GlcNAc), is associated with pancreatic beta-cell apoptosis. In the present study, we show that streptozotocin (STZ), a non-competitive chemical blocker of O-GlcNAcase, induces an insulin secretory defect in isolated rat islet cells. In contrast, transgenic mice with down-regulated glucose to glucosamine metabolism in beta-cells exhibited an enhanced insulin secretion capacity. Interestingly, the STZ blockade of O-GlcNAcase activity is also associated with a growth hormone secretory defect and impairment of intracellular secretory vesicle trafficking. These results provide evidence for the roles of O-GlcNAc in the insulin secretion and possible involvement of O-GlcNAc in general glucose-regulated hormone secretion pathways.


Subject(s)
Acetylglucosaminidase/antagonists & inhibitors , Growth Hormone/drug effects , Insulin Antagonists/pharmacology , Insulin/metabolism , Streptozocin/pharmacology , Acetylglucosamine/metabolism , Acetylglucosamine/physiology , Animals , Glucose/metabolism , Growth Hormone/metabolism , Histone Acetyltransferases , Insulin Secretion , Islets of Langerhans/cytology , Male , Mice , Mice, Transgenic , Multienzyme Complexes , Rats , Rats, Sprague-Dawley , Secretory Vesicles , beta-N-Acetylhexosaminidases
SELECTION OF CITATIONS
SEARCH DETAIL
...