Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
2.
Oncogene ; 36(36): 5177-5188, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28481868

ABSTRACT

BRD4 has emerged as an important factor in tumorigenesis by promoting the transcription of genes involved in cancer development. However, how BRD4 is regulated in cancer cells remains largely unknown. Here, we report that the stability and functions of BRD4 are positively regulated by prolyl isomerase PIN1 in gastric cancer cells. PIN1 directly binds to phosphorylated threonine (T) 204 of BRD4 as revealed by peptide binding and crystallographic studies and enhances BRD4's stability by inhibiting its ubiquitination. PIN1 also catalyses the isomerization of proline 205 of BRD4 and induces its conformational change, which promotes its interaction with CDK9 and increases BRD4's transcriptional activity. Substitution of BRD4 with PIN1-binding-defective BRD4-T204A mutant in gastric cancer cells reduces BRD4's stability, attenuates BRD4-mediated gene expression by impairing its interaction with CDK9 and suppresses gastric cancer cell proliferation, migration and invasion, and tumor formation. Our results identify BRD4 as a new target of PIN1 and suggest that interfering with their interaction could be a potential therapeutic approach for cancer treatment.


Subject(s)
Carcinogenesis , Gene Expression Regulation, Neoplastic , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Stomach Neoplasms/pathology , Transcription Factors/chemistry , Transcription Factors/genetics , Cell Cycle Proteins , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Humans , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Nuclear Proteins/metabolism , Phosphorylation , Point Mutation , Protein Conformation , Protein Stability , Stomach Neoplasms/metabolism , Transcription Factors/metabolism , Tumor Cells, Cultured , Ubiquitination
3.
Curr Mol Med ; 16(9): 793-802, 2016.
Article in English | MEDLINE | ID: mdl-27889995

ABSTRACT

Infection is the process of pathogen invasion, as well as the host reaction to the foreign agents. Proline-directed phosphorylation is a major regulatory mechanism that regulates the function of fundamental proteins involved in infection and infection-induced cancer. Recently, the identification of the phosphorylation-dependent prolyl isomerase Pin1 has uncovered a unique regulatory signaling mechanism controlling protein conformation and function after phosphorylation. Pin1 is the only proline isomerase that specifically recognizes certain Pro-directed Ser/Thr phosphorylation motifs. Pin1 has emerged as a major regulator of cancerrelated viral and bacterial infections notably via activating Toll-like receptor signaling and NF-κB pathways. This paper will specifically review recent findings on the role of Pin1 in cancer-related viral and bacterial infections and also discuss newly discovered Pin1 inhibitors as promising drugs for the prevention and treatment of viral and bacterial infections and associated tumorigenesis.


Subject(s)
Bacterial Infections/metabolism , Neoplasms/metabolism , Peptidylprolyl Isomerase/metabolism , Virus Diseases/metabolism , Animals , Carcinogenesis/metabolism , Humans , Phosphorylation/physiology , Protein Conformation
4.
Nature ; 520(7547): 378-82, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25624101

ABSTRACT

Infectious agents develop intricate mechanisms to interact with host cell pathways and hijack their genetic and epigenetic machinery to change host cell phenotypic states. Among the Apicomplexa phylum of obligate intracellular parasites, which cause veterinary and human diseases, Theileria is the only genus that transforms its mammalian host cells. Theileria infection of bovine leukocytes induces proliferative and invasive phenotypes associated with activated signalling pathways, notably JNK and AP-1 (ref. 2). The transformed phenotypes are reversed by treatment with the theilericidal drug buparvaquone. We used comparative genomics to identify a homologue of the peptidyl-prolyl isomerase PIN1 in T. annulata (TaPIN1) that is secreted into the host cell and modulates oncogenic signalling pathways. Here we show that TaPIN1 is a bona fide prolyl isomerase and that it interacts with the host ubiquitin ligase FBW7, leading to its degradation and subsequent stabilization of c-JUN, which promotes transformation. We performed in vitro and in silico analysis and in vivo zebrafish xenograft experiments to demonstrate that TaPIN1 is directly inhibited by the anti-parasite drug buparvaquone (and other known PIN1 inhibitors) and is mutated in a drug-resistant strain. Prolyl isomerization is thus a conserved mechanism that is important in cancer and is used by Theileria parasites to manipulate host oncogenic signalling.


Subject(s)
Cell Transformation, Neoplastic , Host-Parasite Interactions , Leukocytes/pathology , Peptidylprolyl Isomerase/metabolism , Theileria/enzymology , Theileria/pathogenicity , Animals , Cattle , Cell Line , Cell Transformation, Neoplastic/drug effects , Drug Resistance/genetics , Humans , Leukocytes/drug effects , Leukocytes/parasitology , NIMA-Interacting Peptidylprolyl Isomerase , Naphthoquinones/pharmacology , Parasites/drug effects , Parasites/enzymology , Parasites/pathogenicity , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/genetics , Protein Stability , Proto-Oncogene Proteins c-jun/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction/drug effects , Theileria/drug effects , Theileria/genetics , Transcription Factor AP-1/metabolism , Ubiquitination , Xenograft Model Antitumor Assays , Zebrafish/embryology
5.
Genet Mol Res ; 13(3): 6126-34, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25117371

ABSTRACT

We examined the influence of the promoter polymorphisms -250G/A (rs2070895) and -514C/T (rs1800588) in the human hepatic lipase (LIPC) gene on dyslipidemia and hypertensive disorders complicating pregnancy (HDCP) in a Chinese population. Clinically defined HDCP patients (N = 321) and healthy pregnant women (N = 331) were recruited and genotyped using polymerase chain reaction-restriction fragment length polymorphism for the two LIPC single nucleotide polymorphisms (SNPs). The results showed significant relationships between HDCP and triglycerides, apolipoprotein A1, and high-density lipoprotein cholesterol (P < 0.05), which confirmed that HDCP was accompanied by dyslipidemia. The results also demonstrated that in gestational hypertension (GH) patients, both total cholesterol (TC) and systolic blood pressure (SBP) levels were related to the two SNPs (P ≤ 0.004), although no significant association was found between HDCP and LIPC genotypes or alleles. Significant linkage disequilibrium of the two SNPs was found in both HDCP patients (R(2) = 0.867) and controls (R(2) = 0.91). Body mass index (BMI) was associated with -250G/A in women with mild preeclampsia (MPE) (P = 0.01). Carriers of the mutant homozygote -250AA genotype presented higher BMI in the MPE group. In conclusion, the LIPC -250G/A and -514C/T variants influenced TC and SBP levels in GH patients and the BMI level in the MPE group, although there was no evidence to validate an association between HDCP and LIPC allele, genotype, or haplotype frequencies.


Subject(s)
Asian People/genetics , Genetic Association Studies , Hypertension, Pregnancy-Induced/genetics , Lipase/genetics , Polymorphism, Single Nucleotide , Adult , Alleles , Blood Pressure , Case-Control Studies , China , Female , Gene Frequency , Genetic Predisposition to Disease , Genotype , Humans , Hypertension, Pregnancy-Induced/diagnosis , Lipids/blood , Lipoproteins/blood , Pregnancy , Risk
6.
Curr Mol Med ; 13(7): 1098-109, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23157676

ABSTRACT

Proline-directed protein phosphorylation (pSer/Thr-Pro), a central signaling mechanism in diverse cellular processes in physiology and disease, has been proposed to be subject to further cis-trans conformational regulation by the unique prolyl isomerase Pin1. Until recently, no tool is available to directly detect the cis-trans conformation of Pin1-catalyzed cis-trans conformational changes in vivo. We have developed novel peptide chemistry that enables to generate the first antibodies that can distinguish cis from trans pThr231-Pro conformation in tau (p-tau). Using these conformation-specific antibodies, we have discovered that cis, but not trans, p-tau appears early in mild cognitive impairment (MCI) neurons and further accumulates in neurofibrillary degenerated neurons as Alzheimer's disease (AD) progresses, localizing to the dystrophic neurites, an early hallmark change that correlates with synaptic and cognitive deficits. Unlike trans p-tau, the cis not only cannot promote microtubule assembly, but also is more resistant to dephosphorylation and degradation, and prone to aggregation. Pin1 accelerates cis to trans conversion to prevent the accumulation of the pathogenic cis p-tau conformation in AD, providing the first structural evidence for how Pin1 protects against AD. These findings develop the first tool to directly detect cis-trans prolyl isomerization, especially after phosphorylation and uncover cis p-tau as the very early pathogenic conformation leading to tau pathology and memory loss in AD. These results also suggest novel conformation-specific diagnoses and therapies for AD and likely others.


Subject(s)
Alzheimer Disease/metabolism , Peptidylprolyl Isomerase/metabolism , Proline/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , Humans , Molecular Targeted Therapy , NIMA-Interacting Peptidylprolyl Isomerase , Neurites/metabolism , Neurites/pathology , Neurons/metabolism , Neurons/pathology , Phosphorylation , Protein Binding , Signal Transduction
7.
Oncogene ; 31(45): 4798-802, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22249250

ABSTRACT

Human basal-like breast cancer (BLBC) is an enigmatic and aggressive malignancy with a poor prognosis. There is an urgent need to identify therapeutic targets for BLBC, because current treatment modalities are limited and not effective. The forkhead box transcription factor FOXC1 has recently been identified as a critical functional biomarker for BLBC. However, how it orchestrates BLBC cells was not clear. Here we show that FOXC1 activates the transcription factor nuclear factor-κB (NF-κB) in BLBC cells by increasing p65/RelA protein stability. High NF-κB activity has been associated with estrogen receptor-negative breast cancer, particularly BLBC. The effect of FOXC1 on p65/RelA protein stability is mediated by increased expression of Pin1, a peptidyl-prolyl isomerase. FOXC1 requires NF-κB for its regulation of cell proliferation, migration and invasion. Notably, FOXC1 overexpression renders breast cancer cells more susceptible to pharmacological inhibition of NF-κB. These results suggest that BLBC cells may rely on FOXC1-driven NF-κB signaling. Interventions of this pathway may provide modalities for the treatment of BLBC.


Subject(s)
Breast Neoplasms/metabolism , Forkhead Transcription Factors/metabolism , NF-kappa B/metabolism , Neoplasms, Basal Cell/metabolism , Signal Transduction , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Enzyme Activation , Female , Gene Expression Regulation, Neoplastic , Humans , NF-kappa B/genetics , NIMA-Interacting Peptidylprolyl Isomerase , Neoplasms, Basal Cell/genetics , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/metabolism , Protein Stability , Transcription Factor RelA/metabolism
8.
J Mol Cell Cardiol ; 38(6): 983-96, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15910882

ABSTRACT

To study the complex interaction between oxidative injury and the pathogenesis of vascular disease, vascular gene expression was examined in male Sprague-Dawley rats given 35 or 70 mg/kg allylamine, a synthetic amine converted to acrolein and hydrogen peroxide within the vascular wall. Vascular lesions and extensive vascular remodeling, coupled to increased production of 8-epi-PGF2alpha, nuclear localization of NFkappaB, and alterations in glutathione homeostasis, were observed in animals treated with allylamine for up to 20 days. Transcriptional profiling, immunohistochemistry, and in situ hybridization showed that genes involved in adhesion and extracellular matrix (ECM) (alpha(1) integrin, collagen), cytoskeletal rearrangements (alpha-smooth muscle actin, alpha-tropomyosin), and signal transduction (NFkappaB, osteopontin, and LINE) were altered by oxidant treatment. To evaluate mechanisms of gene dysregulation, cultured aortic smooth muscle cells were challenged with allylamine or its metabolites and processed for molecular analysis. These agents increased formation of reactive oxygen species and elicited changes in gene expression similar to those observed in vivo. Oxidative stress and changes in gene expression were inhibited by N-acetyl cysteine, a precursor of glutathione. These results indicate that genes along the ECM-integrin-cytoskeletal axis, in addition to LINE, are molecular targets in oxidant-induced vascular injury.


Subject(s)
Oxidants/pharmacology , Acetylcysteine/metabolism , Acrolein/metabolism , Acrolein/pharmacology , Allylamine/metabolism , Allylamine/pharmacology , Animals , Aorta/metabolism , Blotting, Western , Cluster Analysis , Cytoskeleton/metabolism , Dinoprost/analogs & derivatives , Dinoprost/biosynthesis , Dose-Response Relationship, Drug , Gene Expression Regulation , Genome , Glutathione/metabolism , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Image Processing, Computer-Assisted , In Situ Hybridization, Fluorescence , Integrin alpha1/metabolism , Integrins/metabolism , Male , Microscopy, Fluorescence , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Osteopontin , Oxidants/metabolism , Oxidative Stress , Oxygen/metabolism , RNA/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Reverse Transcriptase Polymerase Chain Reaction , Sialoglycoproteins/metabolism , Tropomyosin/chemistry , Tropomyosin/metabolism
9.
Physiol Genomics ; 13(3): 263-75, 2003 May 13.
Article in English | MEDLINE | ID: mdl-12657712

ABSTRACT

Atherogenic stimuli trigger complex responses in vascular smooth muscle cells (VSMCs) that culminate in activation/repression of overlapping signal transduction cascades involving oxidative stress. In the case of benzo[a]pyrene (BaP), a polycyclic aromatic hydrocarbon present in tobacco smoke, the atherogenic response involves interference with redox homeostasis by oxidative intermediates of BaP metabolism. The present studies were conducted to define genomic profiles and predictive gene biological networks associated with the atherogenic response of murine (aortic) VSMCs to BaP. A combined oxidant-antioxidant treatment regimen was used to identify redox-sensitive targets during the early course of the atherogenic response. Gene expression profiles were defined using cDNA microarrays coupled to analysis of variance and several clustering methodologies. A predictor algorithm was then applied to gain insight into critical gene-gene interactions during atherogenesis. Supervised and nonsupervised analyses identified clones highly regulated by BaP, unaffected by antioxidant, and neutralized by combined chemical treatments. Lymphocyte antigen-6 complex, histocompatibility class I component factors, secreted phosphoprotein, and several interferon-inducible proteins were identified as novel redox-regulated targets of BaP. Predictor analysis confirmed these relationships and identified immune-related genes as critical molecular targets of BaP. Redox-dependent patterns of gene deregulation indicate that oxidative stress plays a prominent role during the early stages of BaP-induced atherogenesis.


Subject(s)
Arteriosclerosis/chemically induced , Arteriosclerosis/genetics , Benzo(a)pyrene/toxicity , Gene Expression Profiling/methods , Genomics/methods , Oligonucleotide Array Sequence Analysis/methods , Oxidants/toxicity , Algorithms , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Arteriosclerosis/metabolism , Benzo(a)pyrene/metabolism , Cells, Cultured , Cluster Analysis , Computational Biology/methods , Gene Expression Profiling/statistics & numerical data , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Oxidants/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , Predictive Value of Tests
10.
FEBS Lett ; 514(2-3): 193-8, 2002 Mar 13.
Article in English | MEDLINE | ID: mdl-11943150

ABSTRACT

Pin2/TRF1 was independently identified as a telomeric DNA-binding protein (TRF1) that regulates telomere length, and as a protein (Pin2) that can bind the mitotic kinase NIMA and suppress its lethal phenotype. We have previously demonstrated that Pin2/TRF1 levels are cell cycle-regulated and its overexpression induces mitotic arrest and then apoptosis. This Pin2/TRF1 activity can be potentiated by microtubule-disrupting agents, but suppressed by phosphorylation of Pin2/TRF1 by ATM; this negative regulation is critical in mediating for many, but not all, ATM-dependent phenotypes. Interestingly, Pin2/TRF1 specifically localizes to mitotic spindles in mitotic cells and affects the microtubule polymerization in vitro. These results suggest a role of Pin2/TRF1 in mitosis. However, nothing is known about whether Pin2/TRF1 affects the spindle function in mitotic progression. Here we characterized a new Pin2/TRF1-interacting protein, EB1, that was originally identified in our yeast two-hybrid screen. Pin2/TRF1 bound EB1 both in vitro and in vivo and they also co-localize at the mitotic spindle in cells. Furthermore, EB1 inhibits the ability of Pin2/TRF1 to promote microtubule polymerization in vitro. Given that EB1 is a microtubule plus end-binding protein, these results further confirm a specific interaction between Pin2/TRF1 and the mitotic spindle. More importantly, we have shown that inhibition of Pin2/TRF1 in ataxia-telangiectasia cells is able to fully restore their mitotic spindle defect in response to microtubule disruption, demonstrating for the first time a functional involvement of Pin2/TRF1 in mitotic spindle regulation.


Subject(s)
DNA-Binding Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Spindle Apparatus/metabolism , Telomere/metabolism , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Cell Line , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/pharmacology , Glutathione Transferase/genetics , HeLa Cells/cytology , HeLa Cells/drug effects , HeLa Cells/metabolism , Humans , Luminescent Proteins/genetics , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/pharmacology , Microtubules/chemistry , Microtubules/metabolism , Mitosis/drug effects , Protein Binding/physiology , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spindle Apparatus/drug effects , Telomeric Repeat Binding Protein 1 , Transfection , Tumor Suppressor Proteins
11.
Atherosclerosis ; 160(2): 273-80, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11849648

ABSTRACT

Treatment of cultured vascular smooth muscle cells (vSMCs) with benzo(a)pyrene (BaP), a prooxidant present in the particulate phase of tobacco smoke, induces highly proliferative (i.e. atherogenic) phenotypes. Critical early target genes in vSMCs have been identified, but patterns of gene expression following repeated cycles of carcinogen treatment in vivo have yet to be evaluated. In the present study, male Sprague-Dawley rats (175-200 g) were given weekly injections of BaP (10 mg/kg) for 8 weeks to induce atherogenic phenotypes. At the end of this atherogenic regimen, vSMCs were established in serial culture and monitored for patterns of proliferative activity and gene expression. vSMCs isolated from BaP-treated animals (hence forth referred to as BaP cells) exhibited constitutively increased growth rates, and marked enhancement of proliferation in response to serum mitogens. Differential display polymerase chain reaction (DD-PCR) and Northern blot analyses revealed that mRNAs for ribosomal protein L31 and Zis genes were suppressed, while gas-5 and mitochondrial mRNAs were overexpressed in BaP cells relative to control mRNA populations. In situ hybridization experiments in vascular tissue confirmed these alterations in vivo. This is the first report linking expression of these genes to proliferative dysregulation during the course of experimentally-induced atherogenesis.


Subject(s)
Arteriosclerosis/metabolism , Gene Expression , Mitochondria, Muscle/metabolism , Muscle, Smooth, Vascular/metabolism , Oxidants/pharmacology , RNA, Messenger/metabolism , RNA, Small Nucleolar/metabolism , RNA-Binding Proteins/metabolism , Ribosomal Proteins/metabolism , Animals , Arteriosclerosis/chemically induced , Arteriosclerosis/genetics , Benzo(a)pyrene/pharmacology , Blotting, Northern , Carcinogens/pharmacology , Cell Division/drug effects , Cell Division/genetics , Cells, Cultured , In Situ Hybridization , Male , Muscle, Smooth, Vascular/cytology , Phenotype , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Mitochondrial , RNA, Small Nucleolar/genetics , RNA-Binding Proteins/genetics , Rats , Rats, Sprague-Dawley , Ribosomal Proteins/genetics
12.
Cell ; 107(3): 347-59, 2001 Nov 02.
Article in English | MEDLINE | ID: mdl-11701125

ABSTRACT

Telomerase activity is critical for normal and transformed human cells to escape from crisis and is implicated in oncogenesis. Here we describe a novel Pin2/TRF1 binding protein, PinX1 that inhibits telomerase activity and affects tumorigenicity. PinX1 and its small TID domain bind the telomerase catalytic subunit hTERT and potently inhibit its activity. Overexpression of PinX1 or its TID domain inhibits telomerase activity, shortens telomeres, and induces crisis, whereas depletion of endogenous PinX1 increases telomerase activity and elongates telomeres. Depletion of PinX1 also increases tumorigenicity in nude mice, consistent with its chromosome localization at 8p23, a region with frequent loss of heterozygosity in a number of human cancers. Thus, PinX1 is a potent telomerase inhibitor and a putative tumor suppressor.


Subject(s)
DNA-Binding Proteins/metabolism , Telomerase/antagonists & inhibitors , Tumor Suppressor Proteins/physiology , Amino Acid Sequence , Animals , Cell Cycle Proteins , Gene Expression , HeLa Cells , Humans , Mice , Mice, Nude , Molecular Sequence Data , Neoplasms, Experimental , Telomerase/metabolism , Telomeric Repeat Binding Protein 1 , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
13.
Curr Biol ; 11(19): 1512-6, 2001 Oct 02.
Article in English | MEDLINE | ID: mdl-11591318

ABSTRACT

Pin2/TRF1 was independently identified as a telomeric DNA binding protein (TRF1) [1] and as a protein (Pin2) that can bind the mitotic kinase NIMA and suppress its ability to induce mitotic catastrophe [2, 3]. Pin2/TRF1 has been shown to bind telomeric DNA as a dimer [3-7] and to negatively regulate telomere length [8-11]. Interestingly, Pin2/TRF1 levels are regulated during the cell cycle, being increased in late G2 and mitosis and degraded as cells exit from mitosis [3]. Furthermore, overexpression of Pin2/TRF1 induces mitotic entry and then apoptosis [12]. This Pin2/TRF1 activity can be significantly potentiated by the microtubule-disrupting agent nocodazole [12] but is suppressed by phosphorylation of Pin2/TRF1 by ATM; this negative regulation is important for preventing apoptosis upon DNA damage [13]. These results suggest a role for Pin2/TRF1 in mitosis. However, nothing is known about how Pin2/TRF1 is involved in mitotic progression. Here, we describe a surprising physical interaction between Pin2/TRF1 and microtubules in a cell cycle-specific manner. Both expressed and endogenous Pin2/TRF1 proteins were localized to the mitotic spindle during mitosis. Furthermore, Pin2/TRF1 directly bound microtubules via its C-terminal domain. Moreover, Pin2/TRF1 also promoted microtubule polymerization in vitro. These results demonstrate for the first time a specific interaction between Pin2/TRF1 and microtubules in a mitosis-specific manner, and they suggest a new role for Pin2/TRF1 in modulating the function of microtubules during mitosis.


Subject(s)
DNA-Binding Proteins/metabolism , Spindle Apparatus/metabolism , Binding Sites , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Microtubules/metabolism , Polymers , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Telomeric Repeat Binding Protein 1
14.
Nat Cell Biol ; 3(9): 793-801, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11533658

ABSTRACT

Phosphorylation on a serine or threonine residue preceding proline (Ser/Thr-Pro) is a key regulatory mechanism, and the conformation of certain phosphorylated Ser/Thr-Pro bonds is regulated specifically by the prolyl isomerase Pin1. Whereas the inhibition of Pin1 induces apoptosis, Pin1 is strikingly overexpressed in a subset of human tumours. Here we show that Pin1 regulates beta-catenin turnover and subcellular localization by interfering with its interaction with adenomatous polyposis coli protein (APC). A differential-display screen reveals that Pin1 increases the transcription of several beta-catenin target genes, including those encoding cyclin D1 and c-Myc. Manipulation of Pin1 levels affects the stability of beta-catenin in vitro. Furthermore, beta-catenin levels are decreased in Pin1-deficient mice but are increased and correlated with Pin1 overexpression in human breast cancer. Pin1 directly binds a phosphorylated Ser-Pro motif next to the APC-binding site in beta-catenin, inhibits its interaction with APC and increases its translocation into the nucleus. Thus, Pin1 is a novel regulator of beta-catenin signalling and its overexpression might contribute to the upregulation of beta-catenin in tumours such as breast cancer, in which APC or beta-catenin mutations are not common.


Subject(s)
Adenomatous Polyposis Coli/genetics , Cytoskeletal Proteins/metabolism , Gene Expression Regulation , Peptidylprolyl Isomerase/metabolism , Trans-Activators , Adenomatous Polyposis Coli/enzymology , Adenomatous Polyposis Coli/pathology , Amino Acid Sequence , Amino Acid Substitution , Cadherins/metabolism , Cell Nucleus/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Genes, Reporter , HeLa Cells , Humans , Kinetics , Mutagenesis, Site-Directed , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/chemistry , Peptidylprolyl Isomerase/genetics , Phosphorylation , Phosphothreonine/metabolism , Protein Transport , Recombinant Fusion Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection , beta Catenin
15.
Curr Biol ; 11(13): 1062-7, 2001 Jul 10.
Article in English | MEDLINE | ID: mdl-11470413

ABSTRACT

Human EB1 was originally cloned as a protein that interacts with the COOH terminus of adenomatous polyposis coli (APC). Interestingly, this interaction is often disrupted in colon cancer, due to mutations in APC. EB1 also interacts with the plus-ends of microtubules and targets APC to microtubule tips. Since APC is detected on the kinetochores of chromosomes, it has been hypothesized that the EB1-APC interaction connects microtubule spindles to the kinetochores and regulates microtubule stability. In yeast, EB1 regulates microtubule dynamics, and its binding domain in APC may be conserved in Kar9, an EB1 binding protein involved in the microtubule-capturing mechanism. These results suggest that the interaction of EB1 and APC is important and may be conserved. However, it is largely unknown whether the EB1-APC interaction affects microtubule dynamics. Here, we show that EB1 potently promotes microtubule polymerization in vitro and in permeabilized cells, but, surprisingly, only in the presence of the COOH-terminal EB1 binding domain of APC (C-APC). Significantly, this C-APC activity is abolished by phosphorylation, which also disrupts its ability to bind to EB1. Furthermore, yeast EB1 protein effectively substitutes for the human protein but also requires C-APC in promoting microtubule polymerization. Finally, C-APC is able to promote microtubule polymerization when stably expressed in APC mutant cells, demonstrating the ability of C-APC to promote microtubule assembly in vivo. Thus, the interaction between EB1 and APC plays an essential role in the regulation of microtubule polymerization, and a similar mechanism may be conserved in yeast.


Subject(s)
Cytoskeletal Proteins/physiology , Microtubule-Associated Proteins/physiology , Microtubules/metabolism , 3T3 Cells , Adenomatous Polyposis Coli Protein , Amino Acid Sequence , Animals , Biological Evolution , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Fungal Proteins/physiology , Humans , Mice , Molecular Sequence Data , Mutation , Phosphorylation , Polymers/metabolism , Protein Structure, Tertiary
16.
EMBO J ; 20(13): 3459-72, 2001 Jul 02.
Article in English | MEDLINE | ID: mdl-11432833

ABSTRACT

Phosphorylation on serines or threonines preceding proline (Ser/Thr-Pro) is a major signaling mechanism. The conformation of a subset of phosphorylated Ser/Thr-Pro motifs is regulated by the prolyl isomerase Pin1. Inhibition of Pin1 induces apoptosis and may also contribute to neuronal death in Alzheimer's disease. However, little is known about the role of Pin1 in cancer or in modulating transcription factor activity. Here we report that Pin1 is strikingly overexpressed in human breast cancers, and that its levels correlate with cyclin D1 levels in tumors. Overexpression of Pin1 increases cellular cyclin D1 protein and activates its promoter. Furthermore, Pin1 binds c-Jun that is phosphorylated on Ser63/73-Pro motifs by activated JNK or oncogenic Ras. Moreover, Pin1 cooperates with either activated Ras or JNK to increase transcriptional activity of c-Jun towards the cyclin D1 promoter. Thus, Pin1 is up-regulated in human tumors and cooperates with Ras signaling in increasing c-Jun transcriptional activity towards cyclin D1. Given the crucial roles of Ras signaling and cyclin D1 overexpression in oncogenesis, our results suggest that overexpression of Pin1 may promote tumor growth.


Subject(s)
Breast Neoplasms/metabolism , Breast/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic/physiology , Peptidylprolyl Isomerase/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/physiology , Transcription, Genetic , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Breast/cytology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma in Situ/genetics , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Cells, Cultured , Female , Gene Expression Regulation/physiology , Humans , JNK Mitogen-Activated Protein Kinases , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics , Phosphorylation , Promoter Regions, Genetic , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Tumor Cells, Cultured , ras Proteins/metabolism
17.
FEBS Lett ; 496(2-3): 105-8, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11356192

ABSTRACT

Nuclear factor of activated T cells (NFAT) plays a key role in T cell activation. The activation of NFAT involves calcium- and calcineurin-dependent dephosphorylation and nuclear translocation from the cytoplasm, a process that is opposed by protein kinases. We show here that the peptidyl prolyl cis-trans isomerase Pin1 interacts specifically with the phosphorylated form of NFAT. The NFAT-Pin1 interaction is mediated through the WW domain of Pin1 and the serine-proline-rich domains of NFAT. Furthermore, binding of Pin1 to NFAT inhibits the calcineurin-mediated dephosphorylation of NFAT in vitro, and overexpression of Pin1 in T cells inhibits calcium-dependent activation of NFAT in vivo. These results suggest a possible role for Pin1 in the regulation of NFAT in T cells.


Subject(s)
Calcium/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Nuclear Proteins , Peptidylprolyl Isomerase/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , cis-trans-Isomerases/chemistry , Amino Acid Motifs , Blotting, Western , Calcineurin/metabolism , Doxycycline/pharmacology , Glutathione Transferase/metabolism , Humans , Ionomycin/pharmacology , Jurkat Cells , Lymphocyte Activation , NFATC Transcription Factors , NIMA-Interacting Peptidylprolyl Isomerase , Phosphorylation , Plasmids/metabolism , Proline/chemistry , Protein Binding , Protein Biosynthesis , Protein Structure, Tertiary , Serine/chemistry , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic , Transfection
18.
J Biol Chem ; 276(31): 29282-91, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11375976

ABSTRACT

ATM mutations are responsible for the genetic disease ataxia-telangiectasia (A-T). ATM encodes a protein kinase that is activated by ionizing radiation-induced double strand DNA breaks. Cells derived from A-T patients show many abnormalities, including accelerated telomere loss and hypersensitivity to ionizing radiation; they enter into mitosis and apoptosis after DNA damage. Pin2 was originally identified as a protein involved in G(2)/M regulation and is almost identical to TRF1, a telomeric protein that negatively regulates telomere elongation. Pin2 and TRF1, probably encoded by the same gene, PIN2/TRF1, are regulated during the cell cycle. Furthermore, up-regulation of Pin2 or TRF1 induces mitotic entry and apoptosis, a phenotype similar to that of A-T cells after DNA damage. These results suggest that ATM may regulate the function of Pin2/TRF1, but their exact relationship remains unknown. Here we show that Pin2/TRF1 coimmunoprecipitated with ATM, and its phosphorylation was increased in an ATM-dependent manner by ionizing DNA damage. Furthermore, activated ATM directly phosphorylated Pin2/TRF1 preferentially on the conserved Ser(219)-Gln site in vitro and in vivo. The biological significance of this phosphorylation is substantiated by functional analyses of the phosphorylation site mutants. Although expression of Pin2 and its mutants has no detectable effect on telomere length in transient transfection, a Pin2 mutant refractory to ATM phosphorylation on Ser(219) potently induces mitotic entry and apoptosis and increases radiation hypersensitivity of A-T cells. In contrast, Pin2 mutants mimicking ATM phosphorylation on Ser(219) completely fail to induce apoptosis and also reduce radiation hypersensitivity of A-T cells. Interestingly, the phenotype of the phosphorylation-mimicking mutants is the same as that which resulted from inhibition of endogenous Pin2/TRF1 in A-T cells by its dominant-negative mutants. These results demonstrate for the first time that ATM interacts with and phosphorylates Pin2/TRF1 and suggest that Pin2/TRF1 may be involved in the cellular response to double strand DNA breaks.


Subject(s)
DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Telomere/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Cell Line , Cricetinae , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , HeLa Cells , Humans , Kinetics , Mice , Mutagenesis, Site-Directed , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Serine , T-Lymphocytes , Telomere/genetics , Telomeric Repeat Binding Protein 1 , Transfection , Tumor Suppressor Proteins
19.
Oncogene ; 20(12): 1497-508, 2001 Mar 22.
Article in English | MEDLINE | ID: mdl-11313893

ABSTRACT

Telomeres are essential for cell survival and have been implicated in the mitotic control. The telomeric protein Pin2/TRF1 controls telomere elongation and its expression is tightly regulated during cell cycle. We previously reported that overexpression of Pin2/TRF1 affects mitotic progression. However, the role of Pin2/TRF1 at the interface between cell division and cell survival remains to be determined. Here we show that overexpression of Pin2 induced apoptosis in cells containing short telomeres, but not in cells with long telomeres. Furthermore, before entering apoptosis, Pin2-expressing cells first accumulated in mitosis and strongly stained with the mitosis-specific MPM2 antibody. Moreover, Pin2-induced apoptosis is potentiated by arresting cells in mitosis, but suppressed by accumulating cells in G1. In addition, overexpression of Pin2 also resulted in activation of caspase-3, and its proapoptotic activity was significantly reduced by inhibition of caspase-3. These results indicate that up-regulation of Pin2/TRF1 can specifically induce entry into mitosis and apoptosis, likely via a mechanism related to activation of caspase-3. Significantly, we also found that, out of 51 human breast cancer tissues and 10 normal controls examined, protein levels of Pin2/TRF1 in tumors were significantly lower than in normal tissues, as detected by immunoblotting analysis and immunocytochemistry. Since down-regulation of Pin2/TRF1 allows cells to maintain long telomeres, these results suggest that down-regulation of Pin2/TRF1 may be important for cancer cells to extend their proliferative potential.


Subject(s)
Apoptosis , Breast Neoplasms/genetics , DNA-Binding Proteins/metabolism , Mitosis , Telomere , Adaptor Proteins, Signal Transducing , Breast Neoplasms/pathology , Caspase 3 , Caspases/metabolism , DNA-Binding Proteins/genetics , Down-Regulation , Enzyme Activation , Female , HeLa Cells , Humans , LIM Domain Proteins , Membrane Proteins , Telomeric Repeat Binding Protein 1
20.
J Biol Chem ; 276(17): 13517-23, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11118438

ABSTRACT

The phosphorylation-specific peptidyl prolyl cis/trans isomerase (PPIase) Pin1 in humans and its homologues in yeast and animal species play an important role in cell cycle regulation. These PPIases consist of an NH(2)-terminal WW domain that binds to specific phosphoserine- or phosphothreonine-proline motifs present in a subset of phosphoproteins and a COOH-terminal PPIase domain that specifically isomerizes the phosphorylated serine/threonine-proline peptide bonds. Here, we describe the isolation of MdPin1, a Pin1 homologue from the plant species apple (Malus domestica) and show that it has the same phosphorylation-specific substrate specificity and can be inhibited by juglone in vitro, as is the case for Pin1. A search in the plant expressed sequence tag data bases reveals that the Pin1-type PPIases are present in various plants, and there are multiple genes in one organism, such as soybean (Glycine max) and tomato (Lycopersicon esculentum). Furthermore, all these plant Pin1-type PPIases, including AtPin1 in Arabidopsis thaliana, do not have a WW domain, but all contain a four-amino acid insertion next to the phospho-specific recognition site of the active site. Interestingly, like Pin1, both MdPin1 and AtPin1 are able to rescue the lethal mitotic phenotype of a temperature-sensitive mutation in the Pin1 homologue ESS1/PTF1 gene in Saccharomyces cerevisiae. However, deleting the extra four amino acid residues abolished the ability of AtPin1 to rescue the yeast mutation under non-overexpression conditions, indicating that these extra amino acids may be important for mediating the substrate interaction of plant enzymes. Finally, expression of MdPin1 is tightly associated with cell division both during apple fruit development in vivo and during cell cultures in vitro. These results have demonstrated that phosphorylation-specific PPIases are highly conserved functionally in yeast, animal, and plant species. Furthermore, the experiments suggest that although plant Pin1-type enzymes do not have a WW domain, they may fulfill the same functions as Pin1 and its homologues do in other organisms.


Subject(s)
Peptidylprolyl Isomerase/chemistry , Peptidylprolyl Isomerase/metabolism , Plant Proteins/chemistry , Plants/enzymology , Amino Acid Sequence , Arabidopsis Proteins , Base Sequence , Binding Sites , Blotting, Northern , Cloning, Molecular , Conserved Sequence , DNA, Complementary/metabolism , Databases, Factual , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Kinetics , Mitosis , Molecular Sequence Data , Mutation , NIMA-Interacting Peptidylprolyl Isomerase , Naphthoquinones/pharmacology , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/physiology , Phenotype , Phosphorylation , Protein Binding , Protein Structure, Tertiary , RNA/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins , Sequence Homology, Amino Acid , Substrate Specificity , Temperature , Time Factors , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...