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










Publication year range
1.
Cancer Res ; 80(21): 4805-4814, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32943441

ABSTRACT

NKX3.1 is the most commonly deleted gene in prostate cancer and is a gatekeeper suppressor. NKX3.1 is haploinsufficient, and pathogenic reduction in protein levels may result from genetic loss, decreased transcription, and increased protein degradation caused by inflammation or PTEN loss. NKX3.1 acts by retarding proliferation, activating antioxidants, and enhancing DNA repair. DYRK1B-mediated phosphorylation at serine 185 of NKX3.1 leads to its polyubiquitination and proteasomal degradation. Because NKX3.1 protein levels are reduced, but never entirely lost, in prostate adenocarcinoma, enhancement of NKX3.1 protein levels represents a potential therapeutic strategy. As a proof of principle, we used CRISPR/Cas9-mediated editing to engineer in vivo a point mutation in murine Nkx3.1 to code for a serine to alanine missense at amino acid 186, the target for Dyrk1b phosphorylation. Nkx3.1S186A/-, Nkx3.1+/- , and Nkx3.1+/+ mice were analyzed over one year to determine the levels of Nkx3.1 expression and effects of the mutant protein on the prostate. Allelic loss of Nkx3.1 caused reduced levels of Nkx3.1 protein, increased proliferation, and prostate hyperplasia and dysplasia, whereas Nkx3.1S186A/- mouse prostates had increased levels of Nkx3.1 protein, reduced prostate size, normal histology, reduced proliferation, and increased DNA end labeling. At 2 months of age, when all mice had normal prostate histology, Nkx3.1+/- mice demonstrated indices of metabolic activation, DNA damage response, and stress response. These data suggest that modulation of Nkx3.1 levels alone can exert long-term control over premalignant changes and susceptibility to DNA damage in the prostate. SIGNIFICANCE: These findings show that prolonging the half-life of Nkx3.1 reduces proliferation, enhances DNA end-labeling, and protects from DNA damage, ultimately blocking the proneoplastic effects of Nkx3.1 allelic loss.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , Homeodomain Proteins/genetics , Prostatic Neoplasms/genetics , Transcription Factors/genetics , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Point Mutation , Prostatic Neoplasms/pathology
2.
Cancer Res ; 79(16): 4124-4134, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31213464

ABSTRACT

NKX3.1 is the most commonly deleted gene in prostate cancer and a gatekeeper suppressor. NKX3.1 is a growth suppressor, mediator of apoptosis, inducer of antioxidants, and enhancer of DNA repair. PTEN is a ubiquitous tumor suppressor that is often decreased in prostate cancer during tumor progression. Steady-state turnover of NKX3.1 is mediated by DYRK1B phosphorylation at NKX3.1 serine 185 that leads to polyubiquitination and proteasomal degradation. In this study, we show PTEN is an NKX3.1 phosphatase that protects NKX3.1 from degradation. PTEN specifically opposed phosphorylation at NKX3.1(S185) and prolonged NKX3.1 half-life. PTEN and NKX3.1 interacted primarily in the nucleus as loss of PTEN nuclear localization abrogated its ability to bind to and protect NKX3.1 from degradation. The effect of PTEN on NKX3.1 was mediated via rapid enzyme-substrate interaction. An effect of PTEN on Nkx3.1 gene transcription was seen in vitro, but not in vivo. In gene-targeted mice, Nkx3.1 expression significantly diminished shortly after loss of Pten expression in the prostate. Nkx3.1 loss primarily increased prostate epithelial cell proliferation in vivo. In these mice, Nkx3.1 mRNA was not affected by Pten expression. Thus, the prostate cancer suppressors PTEN and NKX3.1 interact and loss of PTEN is responsible, at least in part, for progressive loss of NKX3.1 that occurs during tumor progression. SIGNIFICANCE: PTEN functions as a phosphatase of NKX3.1, a gatekeeper suppressor of prostate cancer.


Subject(s)
Homeodomain Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/pathology , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , Male , Mice, Knockout , PTEN Phosphohydrolase/genetics , Phosphorylation , Prostate/pathology , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Transcription Factors/genetics , Dyrk Kinases
3.
Cancer Res ; 75(13): 2686-98, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25977336

ABSTRACT

TMPRSS2 gene rearrangements occur at DNA breaks formed during androgen receptor-mediated transcription and activate expression of ETS transcription factors at the early stages of more than half of prostate cancers. NKX3.1, a prostate tumor suppressor that accelerates the DNA repair response, binds to androgen receptor at the ERG gene breakpoint and inhibits both the juxtaposition of the TMPRSS2 and ERG gene loci and also their recombination. NKX3.1 acts by accelerating DNA repair after androgen-induced transcriptional activation. NKX3.1 influences the recruitment of proteins that promote homology-directed DNA repair. Loss of NKX3.1 favors recruitment to the ERG gene breakpoint of proteins that promote error-prone nonhomologous end-joining. Analysis of prostate cancer tissues showed that the presence of a TMPRSS2-ERG rearrangement was highly correlated with lower levels of NKX3.1 expression consistent with the role of NKX3.1 as a suppressor of the pathogenic gene rearrangement.


Subject(s)
DNA Damage , DNA Repair , Homeodomain Proteins/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Serine Endopeptidases/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Cell Line, Tumor , Gene Rearrangement , Homeodomain Proteins/biosynthesis , Humans , Male , Prostatic Neoplasms/metabolism , Transcription Factors/biosynthesis , Transcriptional Activation , Transcriptional Regulator ERG
4.
Cell Rep ; 4(3): 516-29, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23890999

ABSTRACT

The prostate tumor suppressor NKX3.1 augments response to DNA damage and enhances survival after DNA damage. Within minutes of DNA damage, NKX3.1 undergoes phosphorylation at tyrosine 222, which is required for a functional interaction with ataxia telangiectasia mutated (ATM) kinase. NKX3.1 binds to the N-terminal region of ATM, accelerates ATM activation, and hastens the formation of γhistone2AX. NKX3.1 enhances DNA-dependent ATM kinase activation by both the MRN complex and H2O2 in a DNA-damage-independent manner. ATM, bound to the NKX3.1 homeodomain, phosphorylates NKX3.1, leading to ubiquitination and degradation. Thus, NKX3.1 and ATM have a functional interaction leading to ATM activation and then NKX3.1 degradation in a tightly regulated DNA damage response specific to prostate epithelial cells. These findings demonstrate a mechanism for the tumor-suppressor properties of NKX3.1, demonstrate how NKX3.1 may enhance DNA integrity in prostate stem cells and may help to explain how cells differ in their sensitivity to DNA damage.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Damage , Homeodomain Proteins/metabolism , Prostatic Neoplasms/metabolism , Transcription Factors/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Line, Tumor , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Humans , Male , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transcription Factors/genetics
5.
Biochem J ; 453(1): 125-36, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23557481

ABSTRACT

NKX3.1 (NK3 homeobox 1) is a prostate tumour suppressor protein with a number of activities that are critical for its role in tumour suppression. NKX3.1 mediates the cellular response to DNA damage by interacting with ATM (ataxia telangiectasia mutated) and by activation of topoisomerase I. In the present study we characterized the interaction between NKX3.1 and topoisomerase I. The NKX3.1 homeodomain binds to a region of topoisomerase I spanning the junction between the core and linker domains. Loss of the topoisomerase I N-terminal domain, a region for frequent protein interactions, did not affect binding to NKX3.1 as was shown by the activation of Topo70 (N-terminal truncated topoisomerase I) in vitro. In contrast, NKX3.1 interacts with the enzyme reconstituted from peptide fragments of the core and linker active site domains, but inhibits the DNA-resolving activity of the reconstituted enzyme in vitro. The effect of NKX3.1 on both Topo70 and the reconstituted enzyme was seen in the presence and absence of camptothecin. Neither NKX3.1 nor CPT (camptothecin) had an effect on the interaction of the other with topoisomerase I. Therefore the interactions of NKX3.1 and CPT with the linker domain of topoisomerase I are mutually exclusive. However, in cells the effect of NKX3.1 on topoisomerase binding to DNA sensitized the cells to cellular toxicity and the induction of apoptosis by low doses of CPT. Lastly, topoisomerase I is important for the effect of NKX3.1 on cell survival after DNA damage as topoisomerase knockdown blocked the effect of NKX3.1 on clonogenicity after DNA damage. Therefore NKX3.1 and topoisomerase I interact in vitro and in cells to affect the CPT sensitivity and DNA-repair functions of NKX3.1.


Subject(s)
DNA Topoisomerases, Type I/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Apoptosis/drug effects , Camptothecin/pharmacology , Cell Line, Tumor , DNA Damage/drug effects , DNA Repair/drug effects , Humans , Male , Prostatic Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism
6.
Cancer Res ; 70(8): 3089-97, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20395202

ABSTRACT

The prostate-specific tumor suppressor homeodomain protein NKX3.1 is inactivated by a variety of mechanisms in the earliest phases of prostate carcinogenesis and in premalignant regions of the prostate gland. The mechanisms by which NKX3.1 exercises tumor suppression have not been well elucidated. Here, we show that NKX3.1 affects DNA damage response and cell survival after DNA damage. NKX3.1 expression in PC-3 prostate cancer cells enhances colony formation after DNA damage but has minimal effect on apoptosis. NKX3.1 also diminishes and regulates total cellular accumulation of gammaH2AX. Endogenous NKX3.1 in LNCaP cells localizes to sites of DNA damage where it affects the recruitment of phosphorylated ATM and the phosphorylation of H2AX. Knockdown of NKX3.1 in LNCaP cells attenuates the acute responses of both ATM and H2AX phosphorylation to DNA damage and their subnuclear localization to DNA damage sites. NKX3.1 expression enhances activation of ATM as assayed by autophosphorylation at serine 1981 and activation of ATR as assayed by phosphorylation of CHK1. An inherited mutation of NKX3.1 that predisposes to early prostate cancer and attenuates in vitro DNA binding was devoid of the ability to activate ATM and to colocalize with gammaH2AX at foci of DNA damage. These data show a novel mechanism by which a homeoprotein can affect DNA damage repair and act as a tumor suppressor.


Subject(s)
Apoptosis , DNA Damage , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Prostatic Neoplasms/metabolism , Transcription Factors/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Checkpoint Kinase 1 , DNA Repair , Humans , Male , Mutation , Phosphorylation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
7.
Cancer Res ; 68(17): 6896-901, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18757402

ABSTRACT

Inflammation of the prostate is a risk factor for the development of prostate cancer. In the aging prostate, regions of inflammatory atrophy are foci for prostate epithelial cell transformation. Expression of the suppressor protein NKX3.1 is reduced in regions of inflammatory atrophy and in preinvasive prostate cancer. Inflammatory cytokines tumor necrosis factor (TNF)-alpha and interleukin-1beta accelerate NKX3.1 protein loss by inducing rapid ubiquitination and proteasomal degradation. The effect of TNF-alpha is mediated via the COOH-terminal domain of NKX3.1 where phosphorylation of serine 196 is critical for cytokine-induced degradation. Mutation of serine 196 to alanine abrogates phosphorylation at that site and the effect of TNF-alpha on NKX3.1 ubiquitination and protein loss. This is in contrast to control of steady-state NKX3.1 turnover, which is mediated by serine 185. Mutation of serine 185 to alanine increases NKX3.1 protein stability by inhibiting ubiquitination and doubling the protein half-life. A third COOH-terminal serine at position 195 has a modulating effect on both steady-state protein turnover and on ubiquitination induced by TNF-alpha. Thus, cellular levels of the NKX3.1 tumor suppressor are affected by inflammatory cytokines that target COOH-terminal serine residues to activate ubiquitination and protein degradation. Our data suggest that strategies to inhibit inflammation or to inhibit effector kinases may be useful approaches to prostate cancer prevention.


Subject(s)
Cytokines/physiology , Homeodomain Proteins/metabolism , Inflammation Mediators/physiology , Prostate/metabolism , Transcription Factors/metabolism , Ubiquitin/metabolism , Alanine/genetics , Cell Line, Tumor , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , Male , Mutagenesis, Site-Directed , Phosphorylation , Serine/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
8.
Neurol India ; 56(2): 186-8, 2008.
Article in English | MEDLINE | ID: mdl-18688146

ABSTRACT

Aspergillus sellar abscess is a very rare form of fungal infections of the central nervous system (CNS). In this report, we describe the successful treatment of a patient with aspergillus sellar abscess. A 65-year-old woman presented with headache, nasal discharge and decreased visual acuity. The diagnosis of sellar mass was made on the basis of magnetic resonance imaging (MRI) examination. The computed tomography (CT) scan revealed sellar enlargement and sellar floor bony destruction. After hospitalization the patient underwent transsphenoidal surgery. Histopathological examination of the sellar mass revealed aspergillosis. Postoperatively, amphotericine-B and itraconazole therapy was started. During a six-month follow-up, the patient's headache and inertia disappeared, visual acuity improved. Aspergillus sellar abscess must be considered in the differential diagnosis of a sellar mass. The correct diagnosis of pituitary aspergillosis can only be achieved by histopathological examination. Surgical intervention and antifungal therapy should be considered the optimal treatment.


Subject(s)
Abscess/complications , Aspergillosis/complications , Sella Turcica/pathology , Abscess/therapy , Aged , Antifungal Agents/therapeutic use , Aspergillosis/therapy , Female , Humans , Magnetic Resonance Imaging , Sella Turcica/drug effects , Sella Turcica/surgery , Tomography, X-Ray Computed
9.
Cancer Res ; 67(2): 455-64, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17234752

ABSTRACT

The prostate-specific homeodomain protein NKX3.1 is a tumor suppressor that is commonly down-regulated in human prostate cancer. Using an NKX3.1 affinity column, we isolated topoisomerase I (Topo I) from a PC-3 prostate cancer cell extract. Topo I is a class 1B DNA-resolving enzyme that is ubiquitously expressed in higher organisms and many prokaryotes. NKX3.1 interacts with Topo I to enhance formation of the Topo I-DNA complex and to increase Topo I cleavage of DNA. The two proteins interacted in affinity pull-down experiments in the presence of either DNase or RNase. The NKX3.1 homeodomain was essential, but not sufficient, for the interaction with Topo I. NKX3.1 binding to Topo I occurred independently of the Topo I NH2-terminal domain. The binding of equimolar amounts of Topo I to NKX3.1 caused displacement of NKX3.1 from its cognate DNA recognition sequence. Topo I activity in prostates of Nkx3.1+/- and Nkx3.1-/- mice was reduced compared with wild-type mice, whereas Topo I activity in livers, where no NKX3.1 is expressed, was independent of Nkx3.1 genotype. Endogenous Topo I and NKX3.1 could be coimmunoprecipitated from LNCaP cells, where NKX3.1 and Topo I were found to colocalize in the nucleus and comigrate within the nucleus in response to either gamma-irradiation or mitomycin C exposure, two DNA-damaging agents. This is the first report that a homeodomain protein can modify the activity of Topo I and may have implications for organ-specific DNA replication, transcription, or DNA repair.


Subject(s)
DNA Topoisomerases, Type I/metabolism , Homeodomain Proteins/metabolism , Prostatic Neoplasms/metabolism , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Chromatography, Affinity/methods , DNA Topoisomerases, Type I/isolation & purification , DNA, Neoplasm/metabolism , Enzyme Activation , Humans , Kinetics , Male , Mice , Prostatic Neoplasms/enzymology , Protein Binding
10.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-585425

ABSTRACT

Objective To study the effects and techniques of microsurgical resection of giant hypervascular meningiomas. Methods A retrospective analysis was performed on clinical data of 32 cases of giant hypervascular meningiomas in this hospital from June 1999 to June 2002. Results The Simpson Grade 1 resection was achieved in 15 cases, Grade 2 in 9 cases, Grade 3 in 6 cases, and Grade 4, 2 cases. There were 2 fatal cases. Complications included 4 cases of intracranial hematoma, 6 cases of cerebral edema and infarction (re-operation of decompression was required in 4 cases), 1 case of mutism, 3 cases of cerebrospinal fluid leakage, and 1 case of intracranial infection. The mental dysfunctions or symptoms became worse than before the operation in 7 cases. Follow-up checkups in 30 patients found no recurrence in Simpson Grade 1 resection, 4 cases of recurrence in Simpson Grade 2 resection, and 5 cases of recurrence in Grade 3 and 4. A re-operation was performed to remove the tumor in 6 cases. The size of tumor was unchanged during follow-up period in 7 cases undergoing radiotherapy. Postoperatively, assessments with activity of daily living (ADL) associated with mental dysfunctions revealed grade Ⅰ in 25 cases, grade Ⅱ in 5 cases, and grade Ⅲ in 2. No significant differences were seen in ADL assessments before and after operation (P=0.696). Conclusions Every effort should be made to complete the total resection of intracranial meningiomas. Sufficient preoperative preparation, adequate surgical exposure, efficient management of operative bleeding, and fractionated resection of tumor using microsurgical techniques are important factors to improve clinical outcomes.

11.
Prostate ; 55(2): 111-7, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12661036

ABSTRACT

BACKGROUND: NKX3.1, a member of the NK-class of homeodomain proteins, is expressed primarily in the adult prostate and has growth suppression and differentiating effects in prostate epithelial cells. METHODS: We performed immunohistochemical analysis for NKX3.1 and PSA expression in 4,061 samples included in a tissue microarray of a broad spectrum of human cancers and normal tissues. RESULTS: NKX3.1 expression was seen in prostate epithelial cells, prostate cancer, normal testis, 9% of primary and 5% of metastatic infiltrating ductal breast carcinoma, and 27% of primary and 26% of metastatic infiltrating lobular breast carcinoma. In a cohort of 474 primary breast cancers with median follow-up over 62.5 month survival, we found no effect of NKX3.1 expression on prognosis. NKX3.1 expression was more restricted than the spectrum of prostate specific antigen expression. CONCLUSIONS: Expression of NKX3.1 is highly restricted and is found primarily in benign and malignant prostatic epithelial cells and also in normal testis and lobular carcinoma of the breast.


Subject(s)
Homeodomain Proteins/metabolism , Neoplasms/metabolism , Transcription Factors/metabolism , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Lobular/metabolism , Epithelial Cells/metabolism , Female , Follow-Up Studies , Humans , Immunohistochemistry , Male , Prognosis , Prostate/metabolism , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/metabolism , Reference Values , Testis/metabolism
12.
J Biol Chem ; 277(47): 44969-79, 2002 Nov 22.
Article in English | MEDLINE | ID: mdl-12297496

ABSTRACT

Restoration of expression of the retinoblastoma gene to DU-145 prostate-cancer cells sensitizes them to apoptosis induced by gamma-irradiation. In contrast, RB expression-protected cells from UV-induced cell death. RB, a caspase substrate, remained intact during apoptosis in gamma-irradiated DU-145 cells because serine proteases, but not caspases, were activated. In DU-145 cells, RB-mediated apoptosis involved biphasic activation of ABL kinase. ABL kinase was activated within minutes of irradiation, but in the presence of RB expression ABL kinase activation was enhanced 48 h after irradiation, coincident with the onset of cell death. Apoptosis was inhibited by RB mutants with constitutive ABL binding, but ABL overexpression overcame the effect of the RB mutant constructs. Expression of kinase-dead ABL had a dominant-negative effect on RB-mediated cell death. Activation of JUN N-terminal kinase depended on the presence of RB and occurred within 8 h of irradiation. Mutant JUN proteins that lacked the N-terminal transactivation domain and serine substrates for JUN N-terminal kinase inhibited cell death in a dominant-negative manner. Irradiation of DU-145 cells caused activation of p38 MAPK independent of the expression of RB. Inhibitors of p38 MAPK blocked apoptosis after irradiation of RB-expressing cells. The data show that after gamma-irradiation, intact RB mediates transcriptional activation that leads to activation of JNK and late activation of ABL kinase. In addition, p38 MAPK activation occurred independent of RB. ABL kinase, JUN N-terminal kinase, and p38 MAPK activity were all required for RB-mediated DU-145 cell death after gamma-irradiation.


Subject(s)
Apoptosis/radiation effects , Gamma Rays , Protein Serine-Threonine Kinases , Retinoblastoma Protein/metabolism , Tumor Cells, Cultured/radiation effects , Apoptosis/physiology , Caspases/metabolism , DNA Fragmentation , Enzyme Activation , Genes, Reporter , Humans , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , JNK Mitogen-Activated Protein Kinases , Male , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Prostatic Neoplasms , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-akt , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoblastoma Protein/genetics , Signal Transduction/physiology , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...