Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Mol Cancer Res ; 21(11): 1176-1185, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37477641

ABSTRACT

Prostate-specific membrane antigen (PSMA) expression increases with prostate cancer grade and progression; however, the role of PSMA in prostate cancer progression remains poorly understood. Telomere stability is essential for the survival and genome stability of cancer cells. We found massive telomere DNA damage in PSMA-negative prostate cancer cells (PC-3 and DU145) compared with PSMA-positive prostate cancer (LNCaP) cells. The ectopic expression of PSMA suppressed telomere DNA damage in PC3 cells. PSMA inhibitor, 2-PMPA, and PSMA knockdown induced telomere DNA damage in PSMA-positive LNCaP cells but not in PSMA-negative PC-3 cells, suggesting that PSMA plays a critical role in telomere stability in prostate cancer cells. In addition, we observed that inhibition of PSMA or inhibition of glutamate receptor, which mediates PSMA-dependent activation of AKT, suppressed AKT phosphorylation, and caused telomere DNA damage. Furthermore, 2-PMPA-induced telomere DNA damage in LNCaP cells was associated with telomere aberrations, such as telomere-telomere fusions, sister-chromatid telomere fusions, and telomere breakages. AKT is reported to promote cell growth by stabilizing telomere association with telomere-binding proteins TRF1 and TPP1. We observed that TRF1 and TPP1 transfection of LNCaP cells attenuated the inhibitory effect of 2-PMPA on cell growth and telomere DNA damage. Together, these observations indicate that PSMA role in maintaining telomere stability in prostate cancer cells is mediated by AKT. Thus, these studies reveal an important role of PSMA in maintaining telomere stability that can promote cell survival and, thereby, prostate cancer progression. IMPLICATIONS: Role of PSMA in telomere stability suggests a strong correlation between PSMA expression and prostate cancer progression.


Subject(s)
Prostate , Prostatic Neoplasms , Male , Humans , Prostate/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Antigens, Surface/genetics , Glutamate Carboxypeptidase II/genetics , Prostatic Neoplasms/metabolism , Phosphorylation , Telomere/genetics , Telomere/metabolism , Cell Line, Tumor
2.
Prostate ; 82(2): 182-192, 2022 02.
Article in English | MEDLINE | ID: mdl-34672379

ABSTRACT

BACKGROUND: Despite multiple treatment advances for castration-resistant prostate cancer (CRPC), there are currently no curative therapies and patients ultimately to succumb to the disease. Docetaxel (DTX) is the standard first-line chemotherapy for patients with metastatic CRPC; however, drug resistance is inevitable and often develops rapidly, leading to disease progression in nearly all patients. In contrast, when DTX is deployed with androgen deprivation therapy in castration-sensitive disease, more durable responses and improved outcomes are observed, suggesting that aberrant androgen receptor (AR) signaling accelerates DTX resistance in CRPC. In this study, we demonstrate that AR dysregulates the mitotic checkpoint, a critical pathway involved in the anticancer action of DTX. METHODS: Androgen-dependent and independent cell lines were used to evaluate the role of AR in DTX resistance. Impact of drug treatment on cell viability, survival, and cell-cycle distribution were determined by plate-based viability assay, clonogenic assay, and cell-cycle analysis by flow cytometry, respectively. Mitotic checkpoint kinase signal transduction and apoptosis activation was evaluated by Western blotting. Pathway gene expression analysis was evaluated by RT-PCR. A Bliss independence model was used to calculate synergy scores for drug combination studies. RESULTS: Activation of AR in hormone-sensitive cells induces a rescue phenotype by increasing cell viability and survival and attenuating G2/M arrest in response to DTX. Analysis of mitotic checkpoint signaling shows that AR negatively regulates spindle checkpoint signaling, resulting in premature mitotic progression and evasion of apoptosis. This phenotype is characteristic of mitotic slippage and is also observed in CRPC cell lines where we demonstrate involvement of AR splice variant AR-v7 in dysregulation of checkpoint signaling. Our findings suggest that DTX resistance is mediated through mechanisms that drive premature mitotic exit. Using pharmacologic inhibitors of anaphase-promoting complex/cyclosome and polo-like kinase 1, we show that blocking mitotic exit induces mitotic arrest, apoptosis, and synergistically inhibits cell survival in combination with DTX. CONCLUSION: Our results suggest that targeting the mechanisms of dysregulated mitotic checkpoint signaling in AR-reactivated tumors has significant clinical potential to extend treatment benefit with DTX and improve outcomes in patients with lethal prostate cancer.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Cell Cycle Checkpoints , Docetaxel/pharmacology , Prostatic Neoplasms, Castration-Resistant , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Male , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/metabolism
3.
PLoS One ; 14(5): e0211090, 2019.
Article in English | MEDLINE | ID: mdl-31083651

ABSTRACT

Telomere stability is important for cell viability, as cells with telomere DNA damage that is not repaired do not survive. We reported previously that androgen receptor (AR) antagonist induces telomere DNA damage in androgen-sensitive LNCaP prostate cancer cells; this triggers a DNA damage response (DDR) at telomeres that includes activation of ATM, and blocking ATM activation prevents telomere DNA repair and leads to cell death. Remarkably, AR antagonist induces telomere DNA damage and triggers ATM activation at telomeres also in 22Rv1 castration-resistant prostate cancer (CRPC) cells that are not growth inhibited by AR antagonist. Treatment with AR antagonist enzalutamide (ENZ) or ATM inhibitor (ATMi) by itself had no effect on growth in vitro or in vivo, but combined treatment with ENZ plus ATMi significantly inhibited cell survival in vitro and tumor growth in vivo. By inducing telomere DNA damage and activating a telomere DDR, an opportunity to inhibit DNA repair and promote cell death was created, even in CRPC cells. 22Rv1 cells express both full-length AR and AR splice variant AR-V7, but full-length AR was found to be the predominant form of AR associated with telomeres and required for telomere stability. Although 22Rv1 growth of untreated 22Rv1 cells appears to be driven by AR-V7, it is, ironically, expression of full-length AR that makes them sensitive to growth inhibition by combined treatment with ENZ plus ATMi. Notably, this combined treatment approach to induce telomere DNA damage and inhibit the DDR was effective in inducing cell death also in other CRPC cell lines (LNCaP/AR and C4-2B). Thus, the use of ENZ in combination with a DDR inhibitor, such as ATMi, may be effective in prolonging disease-free survival of patients with AR-positive metastatic CRPC, even those that co-express AR splice variant.


Subject(s)
DNA Damage , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Stress, Physiological , Telomere/genetics , Alternative Splicing , Animals , Antineoplastic Agents/pharmacology , Cell Death , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Heterografts , Humans , Male , Mice , Prostatic Neoplasms, Castration-Resistant/pathology , RNA Interference , Receptors, Androgen/genetics
4.
J Biol Chem ; 290(42): 25522-33, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26336104

ABSTRACT

Androgen receptor (AR) plays a role in maintaining telomere stability in prostate cancer cells, as AR inactivation induces telomere dysfunction within 3 h. Since telomere dysfunction in other systems is known to activate ATM (ataxia telangiectasia mutated)-mediated DNA damage response (DDR) signaling pathways, we investigated the role of ATM-mediated DDR signaling in AR-inactivated prostate cancer cells. Indeed, the induction of telomere dysfunction in cells treated with AR-antagonists (Casodex or MDV3100) or AR-siRNA was associated with a dramatic increase in phosphorylation (activation) of ATM and its downstream effector Chk2 and the presenceof phosphorylated ATM at telomeres, indicating activation of DDR signaling at telomeres. Moreover, Casodex washout led to the reversal of telomere dysfunction, indicating repair of damaged telomeres. ATM inhibitor blocked ATM phosphorylation, induced PARP cleavage, abrogated cell cycle checkpoint activation and attenuated the formation of γH2AX foci at telomeres in AR-inactivated cells, suggesting that ATM inhibitor induces apoptosis in AR-inactivated cells by blocking the repair of damaged DNA at telomeres. Finally, colony formation assay revealed a dramatic decrease in the survival of cells co-treated with Casodex and ATM inhibitor as compared with those treated with either Casodex or ATM inhibitor alone. These observations indicate that inhibitors of DDR signaling pathways may offer a unique opportunity to enhance the potency of AR-targeted therapies for the treatment of androgen-sensitive as well as castration-resistant prostate cancer.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Cell Death/physiology , Prostatic Neoplasms/pathology , Receptors, Androgen/physiology , Telomere , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Enzyme Activation , Humans , Male , Prostatic Neoplasms/genetics
5.
Oncotarget ; 6(8): 6136-50, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25704883

ABSTRACT

There is a critical need for therapeutic agents that can target the amino-terminal domain (NTD) of androgen receptor (AR) for the treatment of castration-resistant prostate cancer (CRPC). Calmodulin (CaM) binds to the AR NTD and regulates AR activity. We discovered that Hydrazinobenzoylcurcumin (HBC), which binds exclusively to CaM, inhibited AR activity. HBC abrogated AR interaction with CaM, suppressed phosphorylation of AR Serine81, and blocked the binding of AR to androgen-response elements. RNA-Seq analysis identified 57 androgen-regulated genes whose expression was significantly (p ≤ 0.002) altered in HBC treated cells as compared to controls. Oncomine analysis revealed that genes repressed by HBC are those that are usually overexpressed in prostate cancer (PCa) and genes stimulated by HBC are those that are often down-regulated in PCa, suggesting a reversing effect of HBC on androgen-regulated gene expression associated with PCa. Ingenuity Pathway Analysis revealed a role of HBC affected genes in cellular functions associated with proliferation and survival. HBC was readily absorbed into the systemic circulation and inhibited the growth of xenografted CRPC tumors in nude mice. These observations demonstrate that HBC inhibits AR activity by targeting the AR NTD and suggest potential usefulness of HBC for effective treatment of CRPC.


Subject(s)
Curcumin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/drug therapy , Pyrazoles/pharmacology , Receptors, Androgen/metabolism , Animals , Cell Proliferation/drug effects , Curcumin/pharmacology , Gene Expression , Humans , Male , Mice , Mice, Nude , NIH 3T3 Cells , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Random Allocation , Xenograft Model Antitumor Assays
6.
Aging (Albany NY) ; 5(1): 3-17, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23363843

ABSTRACT

Telomeres protect the ends of linear chromosomes from being recognized as damaged DNA, and telomere stability is required for genome stability. Here we demonstrate that telomere stability in androgen receptor (AR)-positive LNCaP human prostate cancer cells is dependent on AR and androgen, as AR inactivation by AR antagonist bicalutamide (Casodex), AR-knockdown, or androgen-depletion caused telomere dysfunction, and the effect of androgen-depletion or Casodex was blocked by the addition of androgen. Notably, neither actinomycin D nor cycloheximide blocked the DNA damage response to Casodex, indicating that the role of AR in telomere stability is independent of its role in transcription. We also demonstrate that AR is a component of telomeres, as AR-bound chromatin contains telomeric DNA, and telomeric chromatin contains AR. Importantly, AR inactivation by Casodex caused telomere aberrations, including multiple abnormal telomere signals, remindful of a fragile telomere phenotype that has been described previously to result from defective telomere DNA replication. We suggest that AR plays an important role in telomere stability and replication of telomere DNA in prostate cancer cells, and that AR inactivation-mediated telomere dysfunction may contribute to genomic instability and progression of prostate cancer cells.


Subject(s)
Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Telomere/metabolism , Androgen Antagonists/pharmacology , Anilides/pharmacology , Cell Line, Tumor , Chromatin/metabolism , Humans , Male , Nitriles/pharmacology , Tosyl Compounds/pharmacology , Transcription, Genetic
7.
PLoS One ; 8(2): e56692, 2013.
Article in English | MEDLINE | ID: mdl-23437213

ABSTRACT

BACKGROUND: The androgen receptor (AR) plays a critical role in the proliferation of prostate cancer cells. However, its mechanism of action in proliferation remains unknown. An understanding of the mechanism of AR action in proliferation may lead to the development of effective strategies for the treatment of prostate cancer. METHODOLOGY/PRINCIPAL FINDINGS: In this study we report that pulse treatment of synchronized LNCaP cells with Casodex, an AR-antagonist, for 4 hours in mid-G(1) phase was sufficient to prevent cells from entering S phase. Since the assembly of pre-replication complex (pre-RC) in G(1) is required for the progression of cells from G(1) to S phase, the effect of Casodex during mid-G(1) suggested that the role of AR in proliferation might be to regulate the assembly of pre-RC. To test this possibility, we investigated the interaction between AR and Cdc6, an essential component of pre-RC in LNCaP cells. AR co-localized and co-immunoprecipitated with Cdc6, and Casodex treatment disrupted this interaction. AR-immunoprecipitate (AR-IP) also contained cyclin E and cyclin A, which play a critical role in pre-RC assembly and cell cycle entry into S phase, and DNA polymerase-α, PCNA, and ribonucleotide reductase, which are essential for the initiation of DNA synthesis. In addition, in cells in S phase, AR co-sedimented with components of the DNA replication machinery of cells that entered S phase. CONCLUSIONS/SIGNIFICANCE: Together, these observations suggest a novel role of AR as a component of the pre-RC to exert control over progression of LNCaP cells from G(1) to S phase through a mechanism that is independent of its role as a transcription factor.


Subject(s)
Androgen Receptor Antagonists/administration & dosage , Anilides/administration & dosage , Cell Transformation, Neoplastic/drug effects , Nitriles/administration & dosage , Prostatic Neoplasms/drug therapy , Receptors, Androgen/genetics , Tosyl Compounds/administration & dosage , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin A/metabolism , Cyclin E/metabolism , DNA Replication/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Male , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism
8.
9.
Biochem Biophys Res Commun ; 422(4): 561-7, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22609405

ABSTRACT

Methyl-2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate (CDDO-Me) is a multifunctional oleanane synthetic triterpenoid with potent anti-inflammatory and antitumorigenic properties. The mechanisms of the antisurvival and apoptosis-inducing activities of CDDO-Me and related derivatives of oleanolic acid have been defined; however, to date, no study has been carried out on the effect of CDDOs on human telomerase reverse transcriptase (hTERT) gene or telomerase activity. Here we report for the first time that inhibition of cell proliferation and induction of apoptosis by CDDO-Me in pancreatic cancer cell lines is associated with the inhibition of hTERT gene expression, hTERT telomerase activity and a number of proteins that regulate hTERT expression and activity. Furthermore, abrogation or overexpression of hTERT protein altered the susceptibility of tumor cells to CDDO-Me. These findings suggest that telomerase (hTERT) is a relevant target of CDDO-Me in pancreatic cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Oleanolic Acid/analogs & derivatives , Pancreatic Neoplasms/enzymology , Suppression, Genetic , Telomerase/antagonists & inhibitors , Cell Line, Tumor , Humans , Oleanolic Acid/pharmacology , Pancreatic Neoplasms/genetics , Telomerase/genetics
10.
J Cell Physiol ; 226(7): 1889-96, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21506119

ABSTRACT

Although inactivation of the androgen receptor (AR) by androgen-ablation or anti-androgen treatment has been frontline therapy for disseminated prostate cancer for over 60 years, it is not curative because castration-resistant prostate cancer cells retain AR activity. Therefore, curative strategy should include targeted elimination of AR protein. Since AR binds to calmodulin (CaM), and since CaM-binding proteins are targets of calpain (Cpn)-mediated proteolysis, we studied the role of CaM and Cpn in AR breakdown in prostate cancer cells. Whereas the treatment of prostate cancer cells individually with anti-CaM drug or calcimycin, which increases intracellular Ca(++) and activates Cpn, led to minimal AR breakdown, combined treatment led to a precipitous decrease in AR protein levels. This decrease in AR protein occurred without noticeable changes in AR mRNA levels, suggesting an increase in AR protein turnover rather than inhibition of AR mRNA expression. Thus, CaM inactivation seems to sensitize AR to Cpn-mediated breakdown in prostate cancer cells. Consistent with this possibility, purified recombinant human AR (rhAR) underwent proteolysis in the presence of purified Cpn, and the addition of purified CaM to the incubation blocked rhAR proteolysis. Together, these observations demonstrate that AR is a Cpn target and AR-bound CaM plays an important role in protecting AR from Cpn-mediated breakdown in prostate cancer cells. These observations raise an intriguing possibility that anti-CaM drugs in combination with Cpn-activating agents may offer a curative strategy for the treatment of prostate cancer, which relies on AR for growth and survival.


Subject(s)
Calmodulin/metabolism , Calpain/metabolism , Prostatic Neoplasms/metabolism , Protein Processing, Post-Translational , Receptors, Androgen/metabolism , Antineoplastic Agents/pharmacology , Calcimycin/pharmacology , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calmodulin/antagonists & inhibitors , Calmodulin/genetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Ionophores/pharmacology , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , RNA Interference , RNA, Messenger/metabolism , Receptors, Androgen/genetics , Recombinant Proteins/metabolism , Sulfonamides/pharmacology , Time Factors , Transfection , Trifluoperazine/pharmacology
11.
J Cell Sci ; 124(Pt 1): 68-81, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21118958

ABSTRACT

DNA damage can induce a tumor suppressive response termed cellular senescence. Damaged senescent cells permanently arrest growth, secrete inflammatory cytokines and other proteins and harbor persistent nuclear foci that contain DNA damage response (DDR) proteins. To understand how persistent damage foci differ from transient foci that mark repairable DNA lesions, we identify sequential events that differentiate transient foci from persistent foci, which we term 'DNA segments with chromatin alterations reinforcing senescence' (DNA-SCARS). Unlike transient foci, DNA-SCARS associate with PML nuclear bodies, lack the DNA repair proteins RPA and RAD51, lack single-stranded DNA and DNA synthesis and accumulate activated forms of the DDR mediators CHK2 and p53. DNA-SCARS form independently of p53, pRB and several other checkpoint and repair proteins but require p53 and pRb to trigger the senescence growth arrest. Importantly, depletion of the DNA-SCARS-stabilizing component histone H2AX did not deplete 53BP1 from DNA-SCARS but diminished the presence of MDC1 and activated CHK2. Furthermore, depletion of H2AX reduced both the p53-dependent senescence growth arrest and p53-independent cytokine secretion. DNA-SCARS were also observed following severe damage to multiple human cell types and mouse tissues, suggesting that they can be used in combination with other markers to identify senescent cells. Thus, DNA-SCARS are dynamically formed distinct structures that functionally regulate multiple aspects of the senescent phenotype.


Subject(s)
Cell Cycle/radiation effects , Cell Nucleus/radiation effects , Cellular Senescence/radiation effects , Chromatin/metabolism , Cytokines/metabolism , DNA Damage/radiation effects , Animals , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/genetics , Cytokines/genetics , Histones/metabolism , Humans , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , X-Rays
12.
Curr Protein Pept Sci ; 11(6): 451-8, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20491624

ABSTRACT

The androgen receptor (AR) plays a critical role in proliferation and viability of prostate cancer cells. Therefore, suppressing AR activity by androgen deprivation or anti-androgen treatment has been the frontline therapy for over six decades. However, these treatment strategies are not curative and patients succumb to castration-resistant disease. Although AR is evidently critical for proliferation of prostate cancer cells, very little is known about its mechanism of action in this process. Over the years, the role of AR in prostate cancer cell proliferation and viability has been studied by focusing primarily on its role as a transcription factor. However, recent observations indicate that besides its role as a transcription factor, AR interacts physically with components of the pre-replication complex (pre-RC) and DNA replication machinery (replitase). These interactions may enable AR to exert control over the process of DNA synthesis. In addition, alterations in the proteins that interact with AR in complexes required for DNA synthesis could lead to the development of hormone-refractory prostate cancer. These observations suggest a paradigm shift for the role of AR in proliferation of prostate cancer cells from its role as a transcription factor to a non-transcriptional role as a component of the replication machinery, interacting with cell cycle regulatory proteins and enzymes of DNA synthesis. We propose that a detailed understanding of the structural interactions between AR and the components of pre-RC and replitase may lead to the development of new strategies for the treatment of prostate cancer.


Subject(s)
Cell Cycle/physiology , DNA/biosynthesis , Prostatic Neoplasms/physiopathology , Receptors, Androgen/physiology , Cell Cycle Proteins/metabolism , DNA/genetics , DNA Replication , Humans , Male , Multienzyme Complexes/metabolism , Prostatic Neoplasms/metabolism , Protein Binding , Receptors, Androgen/metabolism
13.
PLoS Genet ; 6(4): e1000920, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20421929

ABSTRACT

The mechanisms governing telomere replication in humans are still poorly understood. To fill this gap, we investigated the timing of replication of single telomeres in human cells. Using in situ hybridization techniques, we have found that specific telomeres have preferential time windows for replication during the S-phase and that these intervals do not depend upon telomere length and are largely conserved between homologous chromosomes and between individuals, even in the presence of large subtelomeric segmental polymorphisms. Importantly, we show that one copy of the 3.3 kb macrosatellite repeat D4Z4, present in the subtelomeric region of the late replicating 4q35 telomere, is sufficient to confer both a more peripheral localization and a later-replicating property to a de novo formed telomere. Also, the presence of beta-satellite repeats next to a newly created telomere is sufficient to delay its replication timing. Remarkably, several native, non-D4Z4-associated, late-replicating telomeres show a preferential localization toward the nuclear periphery, while several early-replicating telomeres are associated with the inner nuclear volume. We propose that, in humans, chromosome arm-specific subtelomeric sequences may influence both the spatial distribution of telomeres in the nucleus and their replication timing.


Subject(s)
Cell Nucleus/metabolism , DNA Replication , Telomere/chemistry , Cell Line , Chromosomes/metabolism , Humans , S Phase , Telomerase/metabolism , Telomere/metabolism , Telomere-Binding Proteins/metabolism
14.
J Biol Chem ; 285(14): 10472-6, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20110352

ABSTRACT

The telomeric complex, shelterin, plays a critical role in protecting chromosome ends from erosion, and disruption of these complexes can lead to chromosomal instability culminating in cell death or malignant transformation. We reported previously that dominant-negative mutants of one of the telomeric proteins called TIN2 cause death of androgen receptor (AR)-negative but not AR-positive prostate cancer cells, raising the question of a possible role of AR in the structural stability of telomeric complexes. Consistent with this possibility, in the present study, we observed that the AR antagonist Casodex (bicalutamide) disrupted telomeric complexes in AR-positive LNCaP cells but not in AR-negative PC-3 cells. Immunofluorescent studies revealed colocalization of TIN2 and AR. Reciprocal immunoprecipitation studies showed association of AR with telomeric proteins. Furthermore, telomeric proteins were overexpressed in prostate cancer cells compared with normal prostate epithelial cells, and sucrose density gradient analysis showed co-sedimentation of AR with telomeric proteins in a shelterin-like mega complex. Together, these observations suggest an allosteric role of AR in telomere complex stability in prostate cancer cells and suggest that AR-antagonist Casodex-mediated cell death may be due to telomere complex disruption.


Subject(s)
Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Androgen Antagonists/pharmacology , Anilides/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Nitriles/pharmacology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Shelterin Complex , Telomere/genetics , Telomere-Binding Proteins/genetics , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism , Tosyl Compounds/pharmacology , Tumor Cells, Cultured , Tumor Suppressor p53-Binding Protein 1
15.
Int J Oncol ; 36(2): 443-50, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20043080

ABSTRACT

Identification of sensitive and specific biomarkers for early detection and prognosis of prostate cancer is essential for timely and appropriate treatment of the disease in individual patients. We identified an RNA transcript with sequence homology to TRPM8 (melastatin-related transient receptor potential member 8) that was overexpressed in tumor vs. patient-matched non-tumor prostate tissues by RT-PCR differential display (DD). Semi-quantitative RT-PCR analysis revealed that TRPM8 levels were higher in tumor than in non-tumor tissue from 31 of 40 (>75%) patients examined. Overexpression of TRPM8 was independent of changes in androgen receptor (AR) mRNA levels in tumor tissue. However, in studies with established cell lines, TRPM8 expression was detectable only in AR-positive, but not in AR-negative cells, and it was suppressed by steroid deprivation or anti-androgen bicalutamide (Casodex) treatment, suggesting the requirement of AR activity for TRPM8 expression in prostate cancer cells. TRPM8 mRNA was also detected in body fluids of men. Most importantly, its levels were significantly higher (p<0.001, n=18) in urine and blood of patients with metastatic disease than in those of healthy men. However, there was no significant difference (p>0.05, n=10) in its levels between prostate cancer patients with localized disease and healthy men. Together, these studies demonstrate that TRPM8 expression is androgen regulated in prostate cancer cells and that, while tissue TRPM8 mRNA levels can be used for detection of prostate cancer, urine and blood TRPM8 mRNA levels may prove to be useful for distinguishing metastatic disease from clinically localized prostate cancer at the time of diagnosis.


Subject(s)
Androgens/metabolism , Biomarkers, Tumor/analysis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , TRPM Cation Channels/metabolism , Blotting, Western , Humans , Male , RNA, Messenger/analysis , Receptors, Androgen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , TRPM Cation Channels/analysis
16.
Cell Cycle ; 8(6): 931-9, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19229133

ABSTRACT

Telomeres are specialized heterochromatin at the ends of linear chromosomes. Telomeres are crucial for maintaining genome stability and play important roles in cellular senescence and tumor biology. Six core proteins-TRF1, TRF2, TIN2, POT1, TPP1 and Rap1 (termed the telosome or shelterin complex)-regulate telomere structure and function. One of these proteins, TIN2, regulates telomere length and structure indirectly by interacting with TRF1, TRF2 and TPP1, but no direct function has been attributed to TIN2. Here we present evidence for a TIN2 isoform (TIN2L) that differs from the originally described TIN2 isoform (TIN2S) in two ways: TIN2L contains an additional 97 amino acids, and TIN2L associates strongly with the nuclear matrix. Stringent salt and detergent conditions failed to extract TIN2L from the nuclear matrix, despite removing other telomere components, including TIN2S. In human mammary epithelial cells, each isoform showed a distinct nuclear distribution both as a function of cell cycle position and telomere length. Our results suggest a dual role for TIN2 in mediating the function of the shelterin complex and tethering telomeres to the nuclear matrix.


Subject(s)
Nuclear Matrix/metabolism , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Animals , Cell Line, Tumor , Humans , Mice , Nuclear Matrix/ultrastructure , Protein Isoforms/metabolism , RNA Interference , Shelterin Complex , Telomere/ultrastructure , Telomere-Binding Proteins/genetics
17.
J Cell Biol ; 181(3): 447-60, 2008 May 05.
Article in English | MEDLINE | ID: mdl-18443218

ABSTRACT

Telomeres are maintained by three DNA-binding proteins (telomeric repeat binding factor 1 [TRF1], TRF2, and protector of telomeres 1 [POT1]) and several associated factors. One factor, TRF1-interacting protein 2 (TIN2), binds TRF1 and TRF2 directly and POT1 indirectly. Along with two other proteins, TPP1 and hRap1, these form a soluble complex that may be the core telomere maintenance complex. It is not clear whether subcomplexes also exist in vivo. We provide evidence for two TIN2 subcomplexes with distinct functions in human cells. We isolated these two TIN2 subcomplexes from nuclear lysates of unperturbed cells and cells expressing TIN2 mutants TIN2-13 and TIN2-15C, which cannot bind TRF2 or TRF1, respectively. In cells with wild-type p53 function, TIN2-15C was more potent than TIN2-13 in causing telomere uncapping and eventual growth arrest. In cells lacking p53 function, TIN2-15C was more potent than TIN2-13 in causing telomere dysfunction and cell death. Our findings suggest that distinct TIN2 complexes exist and that TIN2-15C-sensitive subcomplexes are particularly important for cell survival in the absence of functional p53.


Subject(s)
Cell Survival/physiology , Macromolecular Substances/metabolism , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 1/metabolism , Animals , Cell Line , Cellular Senescence/physiology , Chromosome Aberrations , Humans , Mice , Mutation , Shelterin Complex , Telomerase/metabolism , Telomere-Binding Proteins/genetics , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
18.
J Cell Sci ; 118(Pt 6): 1321-30, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15741234

ABSTRACT

Nuclear organization, such as the formation of specific nuclear subdomains, is generally thought to be involved in the control of cellular phenotype; however, there are relatively few specific examples of how mammalian nuclei organize during radical changes in phenotype, such as those occurring during differentiation and growth arrest. Using human mammary epithelial cells in which growth arrest is essential for morphological differentiation, we show that the arrest of cell proliferation is accompanied by a reorganization of the telomere-associated protein, TIN2, into one to three large nuclear subdomains. The large TIN2 domains do not contain telomeres and occur concomitant with the continued presence of TIN2 at telomeres. The TIN2 domains were sensitive to DNase, but not RNase, occurred frequently, but not exclusively near nucleoli, and overlapped often with dense domains containing heterochromatin protein 1gamma. Expression of truncated forms of TIN2 simultaneously prevented the formation of TIN2 domains and relaxed the stringent morphogenesis-induced growth arrest in human mammary epithelial cells. Here we show that a novel extra-telomeric organization of TIN2 is associated with the control of cell proliferation and identify TIN2 as an important regulator of mammary epithelial differentiation.


Subject(s)
Breast/cytology , Cell Nucleus/metabolism , Epithelial Cells/cytology , Telomere-Binding Proteins/metabolism , Blotting, Western , Bromodeoxyuridine/pharmacology , Cell Culture Techniques/methods , Cell Differentiation , Cell Proliferation , Cells, Cultured , Deoxyribonuclease I/metabolism , Deoxyribonucleases/metabolism , Epithelial Cells/metabolism , Heterochromatin/chemistry , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Ki-67 Antigen/biosynthesis , Microscopy, Fluorescence , Phenotype , Protein Structure, Tertiary , Retroviridae/genetics , Ribonuclease, Pancreatic/metabolism , Ribonucleases/metabolism , Telomere/metabolism , Telomere/ultrastructure
19.
Int J Biochem Cell Biol ; 37(5): 977-90, 2005 May.
Article in English | MEDLINE | ID: mdl-15743672

ABSTRACT

Telomeres are the specialized DNA-protein structures that cap the ends of linear chromosomes, thereby protecting them from degradation and fusion by cellular DNA repair processes. In vertebrate cells, telomeres consist of several kilobase pairs of DNA having the sequence TTAGGG, a few hundred base pairs of single-stranded DNA at the 3' end of the telomeric DNA tract, and a host of proteins that organize the telomeric double and single-stranded DNA into a protective structure. Functional telomeres are essential for maintaining the integrity and stability of genomes. When combined with loss of cell cycle checkpoint controls, telomere dysfunction can lead to genomic instability, a common cause and hallmark of cancer. Consequently, normal mammalian cells respond to dysfunctional telomeres by undergoing apoptosis (programmed cell death) or cellular senescence (permanent cell cycle arrest), two cellular tumor suppressor mechanisms. These tumor suppressor mechanisms are potent suppressors of cancer, but recent evidence suggests that they can antagonistically also contribute to aging phenotypes. Here, we review what is known about the structure and function of telomeres in mammalian cells, particularly human cells, and how telomere dysfunction may arise and contribute to cancer and aging phenotypes.


Subject(s)
Aging , Genomic Instability , Neoplasms/genetics , Telomere/physiology , Animals , DNA-Binding Proteins/physiology , Humans , Telomere/chemistry
20.
J Biol Chem ; 279(42): 43799-804, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15292264

ABSTRACT

Telomeres are protective structures at chromosome ends and are crucial for genomic stability. Mammalian TRF1 and TRF2 bind the double-stranded telomeric repeat sequence and in turn are bound by TIN2, TANK1, TANK2, and hRAP1. TRF1 is a negative regulator of telomere length in telomerase-positive cells, whereas TRF2 is important for telomere capping. TIN2 was identified as a TRF1-interacting protein that mediates TRF1 function. We show here that TIN2 also interacts with TRF2 in vitro and in yeast and mammalian cells. TIN2 mutants defective in binding of TRF1 or TRF2 induce a DNA damage response and destabilize TRF1 and TRF2 at telomeres in human cells. Our findings suggest that the functions of TRF1 and TRF2 are linked by TIN2.


Subject(s)
Telomere-Binding Proteins/metabolism , Telomere/physiology , Telomeric Repeat Binding Protein 2/metabolism , Cell Line , Humans , Protein Binding , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Telomerase/metabolism , Telomere-Binding Proteins/isolation & purification , Telomeric Repeat Binding Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...