Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cell ; 148(4): 651-63, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22341440

ABSTRACT

To assess telomerase as a cancer therapeutic target and determine adaptive mechanisms to telomerase inhibition, we modeled telomerase reactivation and subsequent extinction in T cell lymphomas arising in Atm(-/-) mice engineered with an inducible telomerase reverse transcriptase allele. Telomerase reactivation in the setting of telomere dysfunction enabled full malignant progression with alleviation of telomere dysfunction-induced checkpoints. These cancers possessed copy number alterations targeting key loci in human T cell lymphomagenesis. Upon telomerase extinction, tumor growth eventually slowed with reinstatement of telomere dysfunction-induced checkpoints, yet growth subsequently resumed as tumors acquired alternative lengthening of telomeres (ALT) and aberrant transcriptional networks centering on mitochondrial biology and oxidative defense. ALT+ tumors acquired amplification/overexpression of PGC-1ß, a master regulator of mitochondrial biogenesis and function, and they showed marked sensitivity to PGC-1ß or SOD2 knockdown. Genetic modeling of telomerase extinction reveals vulnerabilities that motivate coincidental inhibition of mitochondrial maintenance and oxidative defense mechanisms to enhance antitelomerase cancer therapy.


Subject(s)
Mitochondria , Telomerase/antagonists & inhibitors , Telomere Homeostasis , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Genes, cdc , Humans , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Mice , Mitochondria/metabolism , Neoplasm Invasiveness/pathology , Neoplasms/genetics , Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/metabolism , Receptors, Estrogen/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Telomerase/genetics , Telomerase/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics
2.
Cell ; 148(5): 896-907, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22341455

ABSTRACT

To determine the role of telomere dysfunction and telomerase reactivation in generating pro-oncogenic genomic events and in carcinoma progression, an inducible telomerase reverse transcriptase (mTert) allele was crossed onto a prostate cancer-prone mouse model null for Pten and p53 tumor suppressors. Constitutive telomerase deficiency and associated telomere dysfunction constrained cancer progression. In contrast, telomerase reactivation in the setting of telomere dysfunction alleviated intratumoral DNA-damage signaling and generated aggressive cancers with rearranged genomes and new tumor biological properties (bone metastases). Comparative oncogenomic analysis revealed numerous recurrent amplifications and deletions of relevance to human prostate cancer. Murine tumors show enrichment of the TGF-ß/SMAD4 network, and genetic validation studies confirmed the cooperative roles of Pten, p53, and Smad4 deficiencies in prostate cancer progression, including skeletal metastases. Thus, telomerase reactivation in tumor cells experiencing telomere dysfunction enables full malignant progression and provides a mechanism for acquisition of cancer-relevant genomic events endowing new tumor biological capabilities.


Subject(s)
Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Telomerase/metabolism , Telomere/metabolism , Animals , Bone Neoplasms/secondary , Cell Line, Tumor , Crosses, Genetic , DNA Copy Number Variations , Disease Models, Animal , Female , Genomic Instability , Humans , Male , Mice , Tumor Suppressor Protein p53/metabolism
3.
Nature ; 470(7333): 269-73, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21289624

ABSTRACT

Effective clinical management of prostate cancer (PCA) has been challenged by significant intratumoural heterogeneity on the genomic and pathological levels and limited understanding of the genetic elements governing disease progression. Here, we exploited the experimental merits of the mouse to test the hypothesis that pathways constraining progression might be activated in indolent Pten-null mouse prostate tumours and that inactivation of such progression barriers in mice would engender a metastasis-prone condition. Comparative transcriptomic and canonical pathway analyses, followed by biochemical confirmation, of normal prostate epithelium versus poorly progressive Pten-null prostate cancers revealed robust activation of the TGFß/BMP-SMAD4 signalling axis. The functional relevance of SMAD4 was further supported by emergence of invasive, metastatic and lethal prostate cancers with 100% penetrance upon genetic deletion of Smad4 in the Pten-null mouse prostate. Pathological and molecular analysis as well as transcriptomic knowledge-based pathway profiling of emerging tumours identified cell proliferation and invasion as two cardinal tumour biological features in the metastatic Smad4/Pten-null PCA model. Follow-on pathological and functional assessment confirmed cyclin D1 and SPP1 as key mediators of these biological processes, which together with PTEN and SMAD4, form a four-gene signature that is prognostic of prostate-specific antigen (PSA) biochemical recurrence and lethal metastasis in human PCA. This model-informed progression analysis, together with genetic, functional and translational studies, establishes SMAD4 as a key regulator of PCA progression in mice and humans.


Subject(s)
Disease Progression , Neoplasm Metastasis/pathology , Prostatic Neoplasms/pathology , Smad4 Protein/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/physiology , Humans , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Mice , Mice, Transgenic , Models, Biological , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/genetics , Osteopontin/genetics , Osteopontin/metabolism , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Penetrance , Prognosis , Prostate/metabolism , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , Smad4 Protein/deficiency , Smad4 Protein/genetics , Transforming Growth Factor beta
4.
Prostate ; 75(16): 1926-33, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26469352

ABSTRACT

BACKGROUND: We previously identified a protein tumor signature of PTEN, SMAD4, SPP1, and CCND1 that, together with clinical features, was associated with lethal outcomes among prostate cancer patients. In the current study, we sought to validate the molecular model using time-dependent measures of AUC and predictive values for discriminating lethal from non-lethal prostate cancer. METHODS: Using data from the initial study, we fit survival models for men with prostate cancer who were participants in the Physicians' Health Study (PHS; n = 276). Based on these models, we generated prognostic risk scores in an independent population, the Health Professionals Follow-up Study (HPFS; n = 347) to evaluate external validity. In each cohort, men were followed prospectively from cancer diagnosis through 2011 for development of distant metastasis or cancer mortality. We measured protein tumor expression of PTEN, SMAD4, SPP1, and CCND1 on tissue microarrays. RESULTS: During a median of 11.9 and 14.3 years follow-up in the PHS and HPFS cohorts, 24 and 32 men (9%) developed lethal disease. When used as a prognostic factor in a new population, addition of the four markers to clinical variables did not improve discriminatory accuracy through 15 years of follow-up. CONCLUSIONS: Although the four markers have been identified as key biological mediators in metastatic progression, they do not provide independent, long-term prognostic information beyond clinical factors when measured at diagnosis. This finding may underscore the broad heterogeneity in aggressive prostate tumors and highlight the challenges that may result from overfitting in discovery-based research.


Subject(s)
Cyclin D1/metabolism , Osteopontin/metabolism , PTEN Phosphohydrolase/metabolism , Prostate/metabolism , Prostatic Neoplasms/diagnosis , Smad4 Protein/metabolism , Aged , Aged, 80 and over , Area Under Curve , Biomarkers, Tumor/metabolism , Disease Progression , Follow-Up Studies , Gene Expression Profiling , Humans , Male , Middle Aged , Neoplasm Invasiveness/pathology , Prognosis , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/mortality
5.
Nature ; 455(7216): 1129-33, 2008 Oct 23.
Article in English | MEDLINE | ID: mdl-18948956

ABSTRACT

Glioblastoma (GBM) is a highly lethal brain tumour presenting as one of two subtypes with distinct clinical histories and molecular profiles. The primary GBM subtype presents acutely as a high-grade disease that typically harbours mutations in EGFR, PTEN and INK4A/ARF (also known as CDKN2A), and the secondary GBM subtype evolves from the slow progression of a low-grade disease that classically possesses PDGF and TP53 events. Here we show that concomitant central nervous system (CNS)-specific deletion of p53 and Pten in the mouse CNS generates a penetrant acute-onset high-grade malignant glioma phenotype with notable clinical, pathological and molecular resemblance to primary GBM in humans. This genetic observation prompted TP53 and PTEN mutational analysis in human primary GBM, demonstrating unexpectedly frequent inactivating mutations of TP53 as well as the expected PTEN mutations. Integrated transcriptomic profiling, in silico promoter analysis and functional studies of murine neural stem cells (NSCs) established that dual, but not singular, inactivation of p53 and Pten promotes an undifferentiated state with high renewal potential and drives increased Myc protein levels and its associated signature. Functional studies validated increased Myc activity as a potent contributor to the impaired differentiation and enhanced renewal of NSCs doubly null for p53 and Pten (p53(-/-) Pten(-/-)) as well as tumour neurospheres (TNSs) derived from this model. Myc also serves to maintain robust tumorigenic potential of p53(-/-) Pten(-/-) TNSs. These murine modelling studies, together with confirmatory transcriptomic/promoter studies in human primary GBM, validate a pathogenetic role of a common tumour suppressor mutation profile in human primary GBM and establish Myc as an important target for cooperative actions of p53 and Pten in the regulation of normal and malignant stem/progenitor cell differentiation, self-renewal and tumorigenic potential.


Subject(s)
Brain Neoplasms/pathology , Cell Differentiation , Glioma/pathology , Neoplastic Stem Cells/pathology , Neurons/pathology , PTEN Phosphohydrolase/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Brain Neoplasms/genetics , Cell Proliferation , Gene Expression Regulation , Glioblastoma/genetics , Glioblastoma/pathology , Glioma/genetics , Humans , Immunohistochemistry , Mice , Neoplastic Stem Cells/metabolism , Neurons/metabolism , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Protein p53/genetics
6.
Mol Cell Biol ; 23(1): 250-8, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12482978

ABSTRACT

mi-er1 (previously called er1) was first isolated from Xenopus laevis embryonic cells as a novel fibroblast growth factor-regulated immediate-early gene. Xmi-er1 was shown to encode a nuclear protein with an N-terminal acidic transcription activation domain. The human orthologue of mi-er1 (hmi-er1) displays 91% similarity to the Xenopus sequence at the amino acid level and was shown to be upregulated in breast carcinoma cell lines and tumors. Alternative splicing at the 3' end of hmi-er1 produces two major isoforms, hMI-ER1alpha and hMI-ER1beta, which contain distinct C-terminal domains. In this study, we investigated the role of hMI-ER1alpha and hMI-ER1beta in the regulation of transcription. Using fusion proteins of hMI-ER1alpha or hMI-ER1beta tethered to the GAL4 DNA binding domain, we show that both isoforms, when recruited to the G5tkCAT minimal promoter, function to repress transcription. We demonstrate that this repressor activity is due to interaction and recruitment of a trichostatin A-sensitive histone deacetylase 1 (HDAC1). Furthermore, deletion analysis revealed that recruitment of HDAC1 to hMI-ER1alpha and hMI-ER1beta occurs through their common ELM2 domain. The ELM2 domain was first described in the Caenorhabditis elegans Egl-27 protein and is present in a number of SANT domain-containing transcription factors. This is the first report of a function for the ELM2 domain, highlighting its role in the regulation of transcription.


Subject(s)
Caenorhabditis elegans Proteins , DNA-Binding Proteins , Histone Deacetylases/metabolism , Repressor Proteins/metabolism , Transcription Factors , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Carcinoma/genetics , Carcinoma/metabolism , Conserved Sequence , Enzyme Inhibitors/pharmacology , Female , Helminth Proteins/chemistry , Histone Deacetylase 1 , Histone Deacetylase Inhibitors , Histone Deacetylases/genetics , Humans , Hydroxamic Acids/pharmacology , Mice , Molecular Sequence Data , Protein Isoforms , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Transcription, Genetic , Tumor Cells, Cultured , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
7.
Oncotarget ; 8(70): 114526-114539, 2017 Dec 29.
Article in English | MEDLINE | ID: mdl-29383099

ABSTRACT

Hepatocellular carcinoma (HCC) represents a serious public health challenge with few therapeutic options available to cancer patients.Wnt/ß-catenin pathway is thought to play a significant role in HCC pathogenesis. In this study, we confirmed high frequency of CTNNB1 (ß-catenin) mutations in two independent cohorts of HCC patients and demonstrated significant upregulation of ß-catenin protein in the overwhelming majority of HCC patient samples, patient-derived xenografts (PDX) and established cell lines. Using genetic tools validated for target specificity through phenotypic rescue experiments, we went on to investigate oncogenic dependency on ß-catenin in an extensive collection of human HCC cells lines. Our results demonstrate that dependency on ß-catenin generally tracks with its activation status. HCC cell lines that harbored activating mutations in CTNNB1 or displayed elevated levels of non-phosphorylated (active) ß-catenin were significantly more sensitive to ß-catenin siRNA treatment than cell lines with wild-type CTNNB1 and lower active ß-catenin. Finally, significant therapeutic benefit of ß-catenin knock-down was demonstrated in established HCC tumor xenografts using doxycycline-inducible shRNA system. ß-catenin downregulation and tumor growth inhibition was associated with reduction in AXIN2, direct transcriptional target of ß-catenin, and decreased cancer cell proliferation as measured by Ki67 staining. Taken together, our data highlight fundamental importance of aberrant ß-catenin signaling in the maintenance of oncogenic phenotype in HCC.

8.
Cancer Discov ; 6(1): 80-95, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26701088

ABSTRACT

UNLABELLED: The signaling mechanisms between prostate cancer cells and infiltrating immune cells may illuminate novel therapeutic approaches. Here, utilizing a prostate adenocarcinoma model driven by loss of Pten and Smad4, we identify polymorphonuclear myeloid-derived suppressor cells (MDSC) as the major infiltrating immune cell type, and depletion of MDSCs blocks progression. Employing a novel dual reporter prostate cancer model, epithelial and stromal transcriptomic profiling identified CXCL5 as a cancer-secreted chemokine to attract CXCR2-expressing MDSCs, and, correspondingly, pharmacologic inhibition of CXCR2 impeded tumor progression. Integrated analyses identified hyperactivated Hippo-YAP signaling in driving CXCL5 upregulation in cancer cells through the YAP-TEAD complex and promoting MDSC recruitment. Clinicopathologic studies reveal upregulation and activation of YAP1 in a subset of human prostate tumors, and the YAP1 signature is enriched in primary prostate tumor samples with stronger expression of MDSC-relevant genes. Together, YAP-driven MDSC recruitment via heterotypic CXCL5-CXCR2 signaling reveals an effective therapeutic strategy for advanced prostate cancer. SIGNIFICANCE: We demonstrate a critical role of MDSCs in prostate tumor progression and discover a cancer cell nonautonomous function of the Hippo-YAP pathway in regulation of CXCL5, a ligand for CXCR2-expressing MDSCs. Pharmacologic elimination of MDSCs or blocking the heterotypic CXCL5-CXCR2 signaling circuit elicits robust antitumor responses and prolongs survival.


Subject(s)
Chemokine CXCL5/genetics , Myeloid Cells/immunology , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms/immunology , Smad4 Protein/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Chemokine CXCL5/metabolism , Disease Progression , Hippo Signaling Pathway , Humans , Male , Mice , Phosphoproteins/genetics , Phosphoproteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Signal Transduction , Transcription Factors , YAP-Signaling Proteins
9.
J Vis Exp ; (98)2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25938254

ABSTRACT

Telomere dysfunction-induced loss of genome integrity and its associated DNA damage signaling and checkpoint responses are well-established drivers that cause tissue degeneration during ageing. Cancer, with incidence rates greatly increasing with age, is characterized by short telomere lengths and high telomerase activity. To study the roles of telomere dysfunction and telomerase reactivation in ageing and cancer, the protocol shows how to generate two murine inducible telomerase knock-in alleles 4-Hydroxytamoxifen (4-OHT)-inducible TERT-Estrogen Receptor (mTERT-ER) and Lox-Stopper-LoxTERT (LSL-mTERT). The protocol describes the procedures to induce telomere dysfunction and reactivate telomerase activity in mTERT-ER and LSL-mTERT mice in vivo. The representative data show that reactivation of telomerase activity can ameliorate the tissue degenerative phenotypes induced by telomere dysfunction. In order to determine the impact of telomerase reactivation on tumorigenesis, we generated prostate tumor model G4 PB-Cre4 Pten(L/L) p53(L/L) LSL-mTERT(L/L) and thymic T-cell lymphoma model G4 Atm(-/-) mTERT(ER/ER). The representative data show that telomerase reactivation in the backdrop of genomic instability induced by telomere dysfunction can greatly enhance tumorigenesis. The protocol also describes the procedures used to isolate neural stem cells (NSCs) from mTERT-ER and LSL-mTERT mice and reactivate telomerase activity in NSCs in vitro. The representative data show that reactivation of telomerase can enhance the self-renewal capability and neurogenesis in vitro. Finally, the protocol describes the procedures for performing telomere FISH (Fluorescence In Situ Hybridization) on both mouse FFPE (Formalin Fixed and Paraffin Embedded) brain tissues and metaphase chromosomes of cultured cells.


Subject(s)
Alleles , Neoplasms/genetics , Regeneration/genetics , Telomerase/genetics , Animals , Cells, Cultured , Female , Gene Knock-In Techniques/methods , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred C57BL , Neoplasms/pathology , Neural Stem Cells/cytology , Neural Stem Cells/enzymology , Telomere/metabolism
10.
Gene ; 295(1): 79-88, 2002 Jul 24.
Article in English | MEDLINE | ID: mdl-12242014

ABSTRACT

mi-er1 (previously called er1) is a fibroblast growth factor-inducible early response gene activated during mesoderm induction in Xenopus embryos and encoding a nuclear protein that functions as a transcriptional activator. The human orthologue of mi-er1 was shown to be upregulated in breast carcinoma cell lines and breast tumours when compared to normal breast cells. In this report, we investigate the structure of the human mi-er1 (hmi-er1) gene and characterize the alternatively spliced transcripts and protein isoforms. hmi-er1 is a single copy gene located at 1p31.2 and spanning 63 kb. It contains 17 exons and includes one skipped exon, a facultative intron and three polyadenylation signals to produce 12 transcripts encoding six distinct proteins. hmi-er1 transcripts were expressed at very low levels in most human adult tissues and the mRNA isoform pattern varied with the tissue. The 12 transcripts encode proteins containing a common internal sequence with variable N- and C-termini. Three distinct N- and two distinct C-termini were identified, giving rise to six protein isoforms. The two C-termini differ significantly in size and sequence and arise from alternate use of a facultative intron to produce hMI-ER1alpha and hMI-ER1beta. In all tissues except testis, transcripts encoding the beta isoform were predominant. hMI-ER1alpha lacks the predicted nuclear localization signal and transfection assays revealed that, unlike hMI-ER1beta, it is not a nuclear protein, but remains in the cytoplasm. Our results demonstrate that alternate use of a facultative intron regulates the subcellular localization of hMI-ER1 proteins and this may have important implications for hMI-ER1 function.


Subject(s)
Alternative Splicing , Immediate-Early Proteins/genetics , Nuclear Proteins/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins , Female , Gene Expression , Genes/genetics , Humans , Introns/genetics , Male , Mice , Molecular Sequence Data , Protein Isoforms/genetics , Sequence Homology, Amino Acid , Transcription Factors , Transcription, Genetic
11.
Biochem Pharmacol ; 63(8): 1415-21, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11996882

ABSTRACT

Sanguinarine, a benzophenanthrine alkaloid, is potentially antineoplastic through induction of cell death pathways. The development of multidrug resistance (MDR) is a major obstacle to the success of chemotherapeutic agents. The aim of this study was to investigate whether sanguinarine is effective against uterine cervical MDR and, if so, by which mechanism. The effects of treatment with sanguinarine on human papillomavirus (HPV) type 16-immortalized endocervical cells and their MDR counterpart cells were compared. Trypan blue exclusion assays and clonogenic survival assays demonstrated that MDR human cervical cells are as sensitive as their drug-sensitive parental cells to death induced by sanguinarine. Upon treatment of both types of cells with sanguinarine, two distinct concentration-dependent modes of cell death were observed. Treatment with 2.12 or 4.24 microM sanguinarine induced death in most cells that was characterized as apoptosis using the criteria of cell surface blebbing, as determined by light and scanning electron microscopy, and proteolytic activation of caspase-3 and cleavage of the caspase-3 substrate poly(ADP-ribose) polymerase (PARP), as detected by Western blot analysis. However, 8.48 and 16.96 microM sanguinarine caused a second mode of cell death, oncosis, distinguished by cell surface blistering, and neither caspase-3 activation nor PARP cleavage. This study provides the first evidence that sanguinarine is effective against MDR in cervical cells via bimodal cell death, which displays alternative mechanisms involving different morphologies and caspase-3 activation status.


Subject(s)
Alkaloids/pharmacology , Apoptosis , Cervix Uteri/cytology , Intercalating Agents/pharmacology , Benzophenanthridines , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Resistance, Multiple , Enzyme Activation/drug effects , Female , Humans , Isoquinolines
12.
Cancer Cell ; 18(5): 472-84, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21075312

ABSTRACT

mTORC1 is a validated therapeutic target for renal cell carcinoma (RCC). Here, analysis of Tsc1-deficient (mTORC1 hyperactivation) mice uncovered a FoxO-dependent negative feedback circuit constraining mTORC1-mediated renal tumorigenesis. We document robust FoxO activation in Tsc1-deficient benign polycystic kidneys and FoxO extinction on progression to murine renal tumors; murine renal tumor progression on genetic deletion of both Tsc1 and FoxOs; and downregulated FoxO expression in most human renal clear cell and papillary carcinomas, yet continued expression in less aggressive RCCs and benign renal tumor subtypes. Mechanistically, integrated analyses revealed that FoxO-mediated block operates via suppression of Myc through upregulation of the Myc antagonists, Mxi1-SRα and mir-145, establishing a FoxO-Mxi1-SRα/mir-145 axis as a major progression block in renal tumor development.


Subject(s)
Carcinoma, Renal Cell/metabolism , Forkhead Transcription Factors/physiology , Kidney Neoplasms/metabolism , Proteins/physiology , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/physiology , Carcinoma, Renal Cell/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , MicroRNAs/physiology , Multiprotein Complexes , Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/physiology , Signal Transduction , TOR Serine-Threonine Kinases , Transcriptional Activation , Tuberous Sclerosis Complex 1 Protein , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/physiology
13.
Cell Stem Cell ; 5(5): 540-53, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19896444

ABSTRACT

The PI3K-AKT-FoxO pathway is integral to lifespan regulation in lower organisms and essential for the stability of long-lived cells in mammals. Here, we report the impact of combined FoxO1, 3, and 4 deficiencies on mammalian brain physiology with a particular emphasis on the study of the neural stem/progenitor cell (NSC) pool. We show that the FoxO family plays a prominent role in NSC proliferation and renewal. FoxO-deficient mice show initial increased brain size and proliferation of neural progenitor cells during early postnatal life, followed by precocious significant decline in the NSC pool and accompanying neurogenesis in adult brains. Mechanistically, integrated transcriptomic, promoter, and functional analyses of FoxO-deficient NSC cultures identified direct gene targets with known links to the regulation of human brain size and the control of cellular proliferation, differentiation, and oxidative defense. Thus, the FoxO family coordinately regulates diverse genes and pathways to govern key aspects of NSC homeostasis in the mammalian brain.


Subject(s)
Brain/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Multipotent Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Animals , Brain/pathology , Calmodulin-Binding Proteins , Cell Differentiation , Cell Proliferation , Cells, Cultured , Forkhead Transcription Factors/genetics , Mice , Mice, Knockout , Multipotent Stem Cells/cytology , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Organ Size/genetics , Oxygen/metabolism , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , Wnt Proteins/genetics
14.
PLoS One ; 3(12): e3949, 2008.
Article in English | MEDLINE | ID: mdl-19079609

ABSTRACT

Prostate cancer is the second leading cause of cancer-related deaths in men. Activation of MAP kinase signaling pathway has been implicated in advanced and androgen-independent prostate cancers, although formal genetic proof has been lacking. In the course of modeling malignant melanoma in a tyrosinase promoter transgenic system, we developed a genetically-engineered mouse (GEM) model of invasive prostate cancers, whereby an activating mutation of BRAF(V600E)--a mutation found in approximately 10% of human prostate tumors--was targeted to the epithelial compartment of the prostate gland on the background of Ink4a/Arf deficiency. These GEM mice developed prostate gland hyperplasia with progression to rapidly growing invasive adenocarcinoma without evidence of AKT activation, providing genetic proof that activation of MAP kinase signaling is sufficient to drive prostate tumorigenesis. Importantly, genetic extinction of BRAF(V600E) in established prostate tumors did not lead to tumor regression, indicating that while sufficient to initiate development of invasive prostate adenocarcinoma, BRAF(V600E) is not required for its maintenance.


Subject(s)
Prostatic Neoplasms/pathology , Proto-Oncogene Proteins B-raf/metabolism , Androgens , Animals , Biomarkers, Tumor/metabolism , Castration , Cell Lineage , Cell Proliferation , Epithelial Cells/enzymology , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Hyperplasia , Male , Mice , Mice, Transgenic , Neoplasm Invasiveness , Phosphoproteins/metabolism , Prostate/enzymology , Prostate/pathology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Trans-Activators/metabolism , Transgenes , Urothelium/embryology , Urothelium/pathology
15.
Cell ; 128(2): 309-23, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17254969

ABSTRACT

Activated phosphoinositide 3-kinase (PI3K)-AKT signaling appears to be an obligate event in the development of cancer. The highly related members of the mammalian FoxO transcription factor family, FoxO1, FoxO3, and FoxO4, represent one of several effector arms of PI3K-AKT signaling, prompting genetic analysis of the role of FoxOs in the neoplastic phenotypes linked to PI3K-AKT activation. While germline or somatic deletion of up to five FoxO alleles produced remarkably modest neoplastic phenotypes, broad somatic deletion of all FoxOs engendered a progressive cancer-prone condition characterized by thymic lymphomas and hemangiomas, demonstrating that the mammalian FoxOs are indeed bona fide tumor suppressors. Transcriptome and promoter analyses of differentially affected endothelium identified direct FoxO targets and revealed that FoxO regulation of these targets in vivo is highly context-specific, even in the same cell type. Functional studies validated Sprouty2 and PBX1, among others, as FoxO-regulated mediators of endothelial cell morphogenesis and vascular homeostasis.


Subject(s)
Cell Lineage/genetics , Cell Transformation, Neoplastic/genetics , Endothelial Cells/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/genetics , Tumor Suppressor Proteins/genetics , Animals , Cell Cycle Proteins , Cell Differentiation/genetics , Cell Transformation, Neoplastic/metabolism , Drosophila Proteins , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Hemangioma/genetics , Hemangioma/metabolism , Homeodomain Proteins/genetics , Homeostasis/genetics , Lymphoma/genetics , Lymphoma/metabolism , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , Nerve Tissue Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Pre-B-Cell Leukemia Transcription Factor 1 , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/metabolism
16.
J Biol Chem ; 279(27): 28009-16, 2004 Jul 02.
Article in English | MEDLINE | ID: mdl-15117948

ABSTRACT

To gain insight into the regulation of hmi-er1 expression, we cloned a human genomic DNA fragment containing one of the two hmi-er1 promoters and consisting of 1460 bp upstream of the translation initiation codon of hMI-ER1. Computer-assisted sequence analysis revealed that the hmi-er1 promoter region contains a CpG island but lacks an identifiable TATA element, initiator sequence and downstream promoter element. This genomic DNA was able to direct transcription of a luciferase reporter gene in a variety of human cell lines, and the minimal promoter was shown to be located within-68/+144 bp. Several putative Sp1 binding sites were identified, and we show that Sp1 can bind to the hmi-er1 minimal promoter and increase transcription, suggesting that the level of hmi-er1 expression may depend on the availability of Sp1 protein. Functional analysis revealed that hMI-ER1 represses Sp1-activated transcription from the minimal promoter by a histone deacetylase-independent mechanism. Chromatin immunoprecipitation analysis demonstrated that both Sp1 and hMI-ER1 are associated with the chromatin of the hmi-er1 promoter and that overexpression of hMI-ER1 in cell lines that allow Tet-On-inducible expression resulted in loss of detectable Sp1 from the endogenous hmi-er1 promoter. The mechanism by which this occurs does not involve binding of hMI-ER1 to cis-acting elements. Instead, we show that hMI-ER1 physically associates with Sp1 and that endogenous complexes containing the two proteins could be detected in vivo. Furthermore, hMI-ER1 specifically interferes with binding of Sp1 to the hmi-er1 minimal promoter as well as to an Sp1 consensus oligonucleotide. Deletion analysis revealed that this interaction occurs through a region containing the SANT domain of hMI-ER1. Together, these data reveal a functional role for the SANT domain in the action of co-repressor regulatory factors and suggest that the association of hMI-ER1 with Sp1 represents a novel mechanism for the negative regulation of Sp1 target promoters.


Subject(s)
Immediate-Early Proteins/chemistry , Nuclear Proteins/chemistry , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Base Sequence , Binding Sites , Cell Line , Chromatin/metabolism , Cloning, Molecular , CpG Islands , DNA/genetics , DNA-Binding Proteins , Dose-Response Relationship, Drug , Down-Regulation , Genes, Reporter , Glutathione Transferase/metabolism , HeLa Cells , Histone Deacetylases/metabolism , Humans , Immediate-Early Proteins/metabolism , Molecular Sequence Data , Nuclear Proteins/metabolism , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Transcription Factors , Transcription, Genetic , Transfection
17.
Exp Cell Res ; 272(2): 185-91, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11777343

ABSTRACT

Telomerase, a ribonucleoprotein complex of hTERT, hTR, and TP1, has been reported to be associated with carcinogenesis and multidrug resistance (MDR). This study used our in vitro human cervical multistep carcinogenesis/MDR model system in which normal human ectocervical and endocervical (HEN) cells were immortalized by HPV18 or 16, respectively, and subsequently transformed. The first evidence was found that immortalization and telomerase activation were correlated with increased expression specifically of two of the hTERT alternatively spliced mRNAs, one encoding wild-type protein containing the full-length functional reverse transcriptase (RT) region and one encoding a defective RT protein. Expression of neither hTERT mRNA containing full-length functional or defective RT motif was affected by transformation/MDR. All-trans-retinoic acid (ATRA) treatment of HPV-immortalized HEN-16-2 cells and transformed/MDR HEN-16-2/CDDP cells inhibited telomerase activity and downregulated expression of hTERT mRNAs containing full-length functional and a defective RT motif, but there were no changes in hTR and TP1 expression. Moreover, ATRA inhibited cell growth rate of HEN-16-2 and HEN-16-2/CDDP cells equally. These results provided the first evidence that ATRA equally in both immortalized and transformed/MDR cell lines inhibits telomerase activity and downregulates expression, but not splicing, of hTERT, and this is correlated with cell growth rate inhibition; the potential is implicated for applying ATRA to hTERT-targeted treatment of cervical cell carcinogenesis/MDR.


Subject(s)
Alternative Splicing/drug effects , Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic , Down-Regulation/drug effects , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Telomerase/antagonists & inhibitors , Telomerase/genetics , Tretinoin/pharmacology , Uterine Cervical Neoplasms/metabolism , Carrier Proteins/genetics , Cell Division , Cell Line, Transformed , Cells, Cultured , Cervix Uteri/cytology , DNA-Binding Proteins , Enzyme Activation , Female , Gene Expression Profiling , HeLa Cells , Humans , Models, Biological , RNA-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL