Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 130: 104498, 2019 10.
Article in English | MEDLINE | ID: mdl-31181281

ABSTRACT

Prion diseases typically involve brain deposition of abnormally folded prion protein, which is associated with activated glia and increased cytokine production. Cyclophilin A (CypA) is a ubiquitous protein with peptidyl prolyl cis-trans isomerase activity, which regulates protein folding, and can be secreted by cells in response to inflammatory stimuli. On the basis of in vitro studies, CypA was proposed to mediate glial activation during prion infection. To investigate the role of CypA in vivo, we inoculated CypA+/+, CypA+/- and CypA-/- mice with the RML prion strain, and recorded the time to onset of neurological signs and to terminal disease, and the astrocyte and microglia response at presymptomatic and symptomatic stages. Time to onset of disease and survival were significantly shorter in CypA-deficient mice than CypA-expressing controls. CypA-deficient mice had significantly greater microglial activation in the presymptomatic stage, and analysis of anti- and pro-inflammatory microglial markers indicated a shift towards a pro-inflammatory phenotype. There was no difference in astrocyte activation. This suggests that CypA contributes to dampening the pro-inflammatory microglial response during the early stage of RML-induced prion disease.


Subject(s)
Brain/pathology , Microglia/metabolism , Peptidylprolyl Isomerase/deficiency , Prion Diseases/metabolism , Prion Diseases/pathology , Animals , Brain/metabolism , Mice , Mice, Knockout
2.
Biochem Biophys Res Commun ; 499(4): 967-972, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29626483

ABSTRACT

Binding of native bacterial protein SlyD to metal affinity matrices remains a major problem in affinity purification of His-tagged recombinant proteins from Escherichia coli cells. In this study, four novel E. coli strains that lack the expression of SlyD/SlyX, were engineered using λ-red mediated chromosomal deletion. The resultant mutant E. coli strains allow us to obtain SlyD-free proteins immediately after metal affinity chromatography, and eliminate additional purification processes. As a model protein, bispecific antibodies composed of anti-F4/80 VHH module and anti-TNF VHH module (MYSTI-2) were used. Using this protein we have shown that the SlyD/SlyX-deficient E. coli strains allow us to obtain a fully functional protein.


Subject(s)
Escherichia coli/metabolism , Peptidylprolyl Isomerase/deficiency , Recombinant Proteins/isolation & purification , Escherichia coli Proteins/metabolism , Flow Cytometry , Humans , Peptidylprolyl Isomerase/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
3.
Biochem Biophys Res Commun ; 452(3): 468-72, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25159840

ABSTRACT

Peptidylprolyl cis/trans isomerase, NIMA-interacting 1 (PIN1) modulates phospho-signaling by catalyzing rotation of the bond between a phosphorylated serine or threonine before proline in proteins. As depletion of PIN1 increased inflammatory protein expression in cultured endothelial cells treated with bacterial endotoxin (lipopolysaccharide, LPS) and interferon-γ, we hypothesized that PIN1 knockout would increase sensitivity to LPS-induced lung inflammation in mice. Mortality due to a high dose of LPS (30mg/kg) was greater in knockout than wildtype mice. Lung myeloperoxidase activity, reflecting neutrophils, was increased to a 35% higher level in PIN1 knockout mouse lung, as compared with wildtype, after treatment with a sublethal dose of 3mgLPS/kg, ip. Unexpectedly, plasma tumor necrosis factor-α (TNF) was approximately 50% less than in wildtype mice. Knockout mice, however, were more sensitive than wildtype to TNF-induced neutrophil accumulation. The neutrophil adhesion molecule, E-selectin, was also elevated in lungs of knockout mice treated with TNF, suggesting that PIN1 depletion increases endothelial sensitivity to TNF. Indeed, TNF induced more reactive oxygen species in cultured endothelial cells depleted of PIN1 with short hairpin RNA than in control cells. Collectively, the results indicate that while PIN1 normally facilitates TNF production in LPS-treated mice, it suppresses pulmonary and endothelial reactions to the cytokine. Tissue or cell-specific effects of PIN1 may affect the overall inflammatory response to LPS and other stimuli.


Subject(s)
Lung/enzymology , Peptidylprolyl Isomerase/genetics , Pneumonia/enzymology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , E-Selectin/genetics , E-Selectin/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/pathology , Mice , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Peptidylprolyl Isomerase/deficiency , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/pharmacology
4.
Cell Death Dis ; 5: e1237, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24853415

ABSTRACT

The presence of tangles composed of phosphorylated tau is one of the neuropathological hallmarks of Alzheimer's disease (AD). Tau, a microtubule (MT)-associated protein, accumulates in AD potentially as a result of posttranslational modifications, such as hyperphosphorylation and conformational changes. However, it has not been fully understood how tau accumulation and phosphorylation are deregulated. In the present study, we identified a novel role of death-associated protein kinase 1 (DAPK1) in the regulation of the tau protein. We found that hippocampal DAPK1 expression is markedly increased in the brains of AD patients compared with age-matched normal subjects. DAPK1 overexpression increased tau protein stability and phosphorylation at multiple AD-related sites. In contrast, inhibition of DAPK1 by overexpression of a DAPK1 kinase-deficient mutant or by genetic knockout significantly decreased tau protein stability and abolished its phosphorylation in cell cultures and in mice. Mechanistically, DAPK1-enhanced tau protein stability was mediated by Ser71 phosphorylation of Pin1, a prolyl isomerase known to regulate tau protein stability, phosphorylation, and tau-related pathologies. In addition, inhibition of DAPK1 kinase activity significantly increased the assembly of MTs and accelerated nerve growth factor-mediated neurite outgrowth. Given that DAPK1 has been genetically linked to late onset AD, these results suggest that DAPK1 is a novel regulator of tau protein abundance, and that DAPK1 upregulation might contribute to tau-related pathologies in AD. Therefore, we offer that DAPK1 might be a novel therapeutic target for treating human AD and other tau-related pathologies.


Subject(s)
Alzheimer Disease/enzymology , Brain/enzymology , Death-Associated Protein Kinases/metabolism , Neurons/enzymology , tau Proteins/metabolism , Age Factors , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Brain/pathology , Case-Control Studies , Death-Associated Protein Kinases/deficiency , Death-Associated Protein Kinases/genetics , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Knockout , Mutation , NIH 3T3 Cells , NIMA-Interacting Peptidylprolyl Isomerase , Neurites/enzymology , Neurites/pathology , Neurons/pathology , PC12 Cells , Peptidylprolyl Isomerase/deficiency , Peptidylprolyl Isomerase/genetics , Phosphorylation , Protein Stability , RNA Interference , Rats , Time Factors , Transfection , tau Proteins/genetics
5.
J Cell Mol Med ; 17(8): 989-1005, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23750710

ABSTRACT

Type 2 diabetes (T2D) is associated with accelerated restenosis rates after angioplasty. We have previously proved that Pin1 played an important role in vascular smooth muscle cell (VSMC) cycle and apoptosis. But neither the role of Pin1 in restenosis by T2D, nor the molecular mechanism of Pin1 in these processes has been elucidated. A mouse model of T2D was generated by the combination of high-fat diet (HFD) and streptozotocin (STZ) injections. Both Immunohistochemistry and Western blot revealed that Pin1 expression was up-regulated in the arterial wall in T2D mice and in VSMCs in culture conditions mimicking T2D. Next, increased activity of Pin1 was observed in neointimal hyperplasia after arterial injury in T2D mice. Further analysis confirmed that 10% serum of T2D mice and Pin1-forced expression stimulated proliferation, inhibited apoptosis, enhanced cell cycle progression and migration of VSMCs, whereas Pin1 knockdown resulted in the converse effects. We demonstrated that STAT3 signalling and mitochondria-dependent pathways played critical roles in the involvement of Pin1 in cell cycle regulation and apoptosis of VSMCs in T2D. In addition, VEGF expression was stimulated by Pin1, which unveiled part of the mechanism of Pin1 in regulating VSMC migration in T2D. Finally, the administration of juglone via pluronic gel onto injured common femoral artery resulted in a significant inhibition of the neointima/media ratio. Our findings demonstrated the vital effect of Pin1 on the VSMC proliferation, cell cycle progression, apoptosis and migration that underlie neointima formation in T2D and implicated Pin1 as a potential therapeutic target to prevent restenosis in T2D.


Subject(s)
Coronary Restenosis/metabolism , Diabetes Mellitus, Type 2/metabolism , Mitochondria/metabolism , Peptidylprolyl Isomerase/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Animals , Apoptosis , Caspases/metabolism , Cell Cycle , Cell Movement , Cell Proliferation , Coronary Restenosis/blood , Coronary Restenosis/complications , Cytochromes c/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Enzyme Activation , Femoral Artery/injuries , Femoral Artery/metabolism , Femoral Artery/pathology , Gene Knockdown Techniques , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , NIMA-Interacting Peptidylprolyl Isomerase , Neointima/metabolism , Neointima/pathology , Peptidylprolyl Isomerase/deficiency , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism , bcl-2-Associated X Protein/metabolism
6.
Circ Res ; 112(9): 1244-52, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23487407

ABSTRACT

RATIONALE: Cardiac hypertrophy results from the complex interplay of differentially regulated cascades based on the phosphorylation status of involved signaling molecules. Although numerous critical regulatory kinases and phosphatases have been identified in the myocardium, the intracellular mechanism for temporal regulation of signaling duration and intensity remains obscure. In the nonmyocyte context, control of folding, activity, and stability of proteins is mediated by the prolyl isomerase Pin1, but the role of Pin1 in the heart is unknown. OBJECTIVE: To establish the role of Pin1 in the heart. METHODS AND RESULTS: Here, we show that either genetic deletion or cardiac overexpression of Pin1 blunts hypertrophic responses induced by transaortic constriction and consequent cardiac failure in vivo. Mechanistically, we find that Pin1 directly binds to Akt, mitogen activated protein kinase (MEK), and Raf-1 in cultured cardiomyocytes after hypertrophic stimulation. Furthermore, loss of Pin1 leads to diminished hypertrophic signaling of Akt and MEK, whereas overexpression of Pin1 increases Raf-1 phosphorylation on the autoinhibitory site Ser259, leading to reduced MEK activation. CONCLUSIONS: Collectively, these data support a role for Pin1 as a central modulator of the intensity and duration of 2 major hypertrophic signaling pathways, thereby providing a novel target for regulation and control of cardiac hypertrophy.


Subject(s)
Cardiomegaly/enzymology , Myocytes, Cardiac/enzymology , Peptidylprolyl Isomerase/metabolism , Signal Transduction , Animals , Cardiomegaly/diagnostic imaging , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiomegaly/prevention & control , Dependovirus/genetics , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/metabolism , Myocytes, Cardiac/pathology , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/deficiency , Peptidylprolyl Isomerase/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Rats , Time Factors , Transduction, Genetic , Transfection , Ultrasonography , raf Kinases/metabolism
7.
PLoS One ; 7(3): e31823, 2012.
Article in English | MEDLINE | ID: mdl-22412843

ABSTRACT

BACKGROUND: A peptidyl prolyl cis/trans isomerase, Pin1, regulates insulin signal transduction. Pin1 reduces responses to insulin stimulation by binding CRTC2 (CREB-regulated transcriptional co-activator 2) and PPARγ (peroxisome prolifereator- activated receptor γ), but conversely enhances insulin signaling by binding IRS-1 (insulin receptor substrate-1), Akt kinase, and Smad3. Therefore, it is still unclear whether Pin1 inhibits or enhances adipose cell differentiation. METHODOLOGY/PRINCIPAL FINDINGS: Pin1(-/-) and wild-type mice were fed with high fat diets and adipose tissue weight was measured. Compared to wild-type mice, Pin1(-/-) mice had lower adipose tissue weight, while the weight of other tissues was similar. Mouse embryo fibroblasts (MEFs), prepared from both groups of mice, were induced to differentiate into adipose cells by stimulation with insulin. However, the rate of differentiation of MEFs from Pin1(-/-) mice was less than that of MEFs from wild-type mice. The rate of insulin-induced MEF cell differentiation in Pin1(-/-) mice was restored by increasing expression of Pin1. We found that Pin1 binds to phosphoThr172- and phosphoSer271-Pro sites in CREB suppress the activity in COS-7 cells. CONCLUSION AND SIGNIFICANCE: Pin1 enhanced the uptake of triglycerides and the differentiation of MEF cells into adipose cells in response to insulin stimulation. Results of this study suggest that Pin1 down-regulation could be a potential approach in obesity-related dysfunctions, such as high blood pressure, diabetes, non-alcoholic steatohepatitis.


Subject(s)
Adipocytes/cytology , Adipocytes/enzymology , Cell Differentiation , Fibroblasts/cytology , Fibroblasts/enzymology , Peptidylprolyl Isomerase/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Cell Differentiation/genetics , Cell Line , Cell Size , Chlorocebus aethiops , Cyclic AMP Response Element-Binding Protein/metabolism , Diet, High-Fat , Embryo, Mammalian/cytology , Humans , Lipid Metabolism , Male , Mice , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/deficiency , Peptidylprolyl Isomerase/genetics , Protein Binding , Tomography, X-Ray Computed , Transcriptional Activation
8.
Hypertension ; 58(3): 431-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21810655

ABSTRACT

Pin1 is a peptidyl prolyl cis-trans isomerase that only binds to and isomerizes phosphorylated serine/threonine-proline motifs, inducing conformational changes that alter target protein function and phosphorylation. We have shown previously that deficiency of another peptidyl prolyl isomerase, FK506 binding protein 12/12.6, alters endothelial NO synthase phosphorylation and causes endothelial dysfunction and hypertension. Endothelial NO synthase contains the Pin1 binding sequence at (p)serine 116-proline 117 and phosphorylation of endothelial NO synthase serine 116 inhibits NO production; however, whether Pin1 deficiency alters vascular function and blood pressure is unknown. We hypothesized that Pin1 isomerizes p-endothelial NO synthase serine 116, which enables dephosphorylation and stimulates NO production. Immunoprecipitation of endothelial NO synthase and probing for Pin1 in rat aortic endothelial cells confirmed the interaction between the two. Pin1 knockdown via small interfering RNA or inhibition by juglone increased endothelial NO synthase serine 116 phosphorylation and prevented vascular endothelial growth factor-induced serine 116 dephosphorylation in endothelial cells. Acute treatment of isolated mouse aortas with juglone increased endothelial NO synthase serine 116 phosphorylation and decreased NO production and relaxation responses. Mice treated with juglone for 2 weeks, as well as Pin1 knockout mice, exhibited increased aortic endothelial NO synthase serine 116 phosphorylation, endothelial dysfunction, and hypertension. These data demonstrate that Pin1 binds endothelial NO synthase and enables dephosphorylation of serine 116, which increases NO production and endothelium-dependent dilation, leading to blood pressure maintenance.


Subject(s)
Endothelium, Vascular/metabolism , Hypertension/genetics , Nitric Oxide Synthase Type III/genetics , Peptidylprolyl Isomerase/genetics , Amino Acid Substitution , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/physiopathology , Binding Sites/genetics , Blood Pressure/drug effects , Cells, Cultured , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Enzyme Inhibitors/pharmacology , Hypertension/metabolism , Hypertension/physiopathology , Immunoblotting , Immunoprecipitation , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , NIMA-Interacting Peptidylprolyl Isomerase , Naphthoquinones/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/deficiency , Phosphorylation , Protein Binding , RNA Interference , Rats , Serine/genetics , Serine/metabolism , Vasodilation/drug effects
9.
J Biol Chem ; 285(3): 1754-64, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19920136

ABSTRACT

Transforming growth factor-beta (TGF-beta) regulates a wide variety of biological activities. It induces potent growth-inhibitory responses in normal cells but promotes migration and invasion of cancer cells. Smads mediate the TGF-beta responses. TGF-beta binding to the cell surface receptors leads to the phosphorylation of Smad2/3 in their C terminus as well as in the proline-rich linker region. The serine/threonine phosphorylation sites in the linker region are followed by the proline residue. Pin1, a peptidyl-prolyl cis/trans isomerase, recognizes phosphorylated serine/threonine-proline motifs. Here we show that Smad2/3 interacts with Pin1 in a TGF-beta-dependent manner. We further show that the phosphorylated threonine 179-proline motif in the Smad3 linker region is the major binding site for Pin1. Although epidermal growth factor also induces phosphorylation of threonine 179 and other residues in the Smad3 linker region the same as TGF-beta, Pin1 is unable to bind to the epidermal growth factor-stimulated Smad3. Further analysis suggests that phosphorylation of Smad3 in the C terminus is necessary for the interaction with Pin1. Depletion of Pin1 by small hairpin RNA does not significantly affect TGF-beta-induced growth-inhibitory responses and a number of TGF-beta/Smad target genes analyzed. In contrast, knockdown of Pin1 in human PC3 prostate cancer cells strongly inhibited TGF-beta-mediated migration and invasion. Accordingly, TGF-beta induction of N-cadherin, which plays an important role in migration and invasion, is markedly reduced when Pin1 is depleted in PC3 cells. Because Pin1 is overexpressed in many cancers, our findings highlight the importance of Pin1 in TGF-beta-induced migration and invasion of cancer cells.


Subject(s)
Cell Movement/drug effects , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Peptidylprolyl Isomerase/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Biocatalysis , Cell Line, Tumor , Cell Movement/genetics , Cell Nucleus/metabolism , Gene Knockdown Techniques , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/deficiency , Peptidylprolyl Isomerase/genetics , Phosphorylation , Smad2 Protein/metabolism , Smad3 Protein/chemistry , Smad3 Protein/metabolism , Substrate Specificity , Threonine/metabolism , Transforming Growth Factor beta/metabolism
10.
Oncogene ; 28(42): 3735-45, 2009 Oct 22.
Article in English | MEDLINE | ID: mdl-19668231

ABSTRACT

The brain tumor glioblastoma (GBM) remains one of the most aggressive and devastating tumors despite decades of effort to find more effective treatments. A hallmark of GBM is the constitutive activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kappaB) signaling pathway, which regulates cell proliferation, inflammation, migration and apoptosis. The prolyl isomerase, Pin1, has been found to bind directly to the NF-kappaB protein, p65, and cause increases in NF-kappaB promoter activity in a breast cancer model. We now present evidence that this interaction occurs in GBM and that it has important consequences on NF-kappaB signaling. We demonstrate that Pin1 levels are enhanced in primary GBM tissues compared with controls, and that this difference in Pin1 expression affects the migratory capacity of GBM-derived cells. Pin1 knockdown decreases the amount of activated, phosphorylated p65 in the nucleus, resulting in inhibition of the transcriptional program of the IL-8 gene. Through the use of microarray, we also observed changes in the expression levels of other NF-kappaB regulated genes due to Pin1 knockdown. Taken together, these data suggest that Pin1 is an important regulator of NF-kappaB in GBM, and support the notion of using Pin1 as a therapeutic target in the future.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Interleukin-8/genetics , NF-kappa B/metabolism , Peptidylprolyl Isomerase/metabolism , Signal Transduction , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Inverted Repeat Sequences , Mice , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/deficiency , Peptidylprolyl Isomerase/genetics , Phosphorylation , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tetracycline/pharmacology , Transcription Factor RelA/metabolism
11.
J Hepatol ; 51(2): 296-306, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19515451

ABSTRACT

BACKGROUND/AIMS: Our previous work suggested an important role for the peptidyl-prolyl isomerase, Pin1, in hepatic NF-kappaB activation and liver injury during ischemia/reperfusion (I/R). In this study, we sought to determine the function of Pin1 in the injury response to hepatic I/R. METHODS: Wild-type and Pin1(-/-) mice were subjected to partial hepatic I/R. In addition, hepatocytes and Kupffer cells were isolated from these mice. RESULTS: Pin1(-/-) mice had reduced hepatic NF-kappaB activation and more liver injury after I/R than wild-type mice. The increased injury was not a result of enhanced inflammation as Pin1(-/-) mice had the same level of proinflammatory cytokine production and less neutrophil accumulation in the liver. The reduced NF-kappaB activation was not a result of a defect in nuclear translocation of NF-kappaB. In fact, hepatic nuclear p65 protein expression was higher in Pin1(-/-) mice than wild-type mice. This suggests that Pin1 is important for NF-kappaB-DNA binding. This effect was specific to hepatocytes as isolated Kupffer cells from wild-type and Pin1(-/-) mice were identical in their activation of NF-kappaB and production of cytokines after stimulation. In contrast, hepatocytes stimulated with TNFalpha had greatly reduced NF-kappaB activation, reduced production of the CXC chemokine, MIP-2, and increased cell death. CONCLUSIONS: These data suggest that Pin1 is a critical regulator of NF-kappaB activation in hepatocytes and its role in these cells appears to confer direct protective effects.


Subject(s)
Hepatocytes/metabolism , Liver/enzymology , Liver/injuries , NF-kappa B/metabolism , Peptidylprolyl Isomerase/physiology , Reperfusion Injury/prevention & control , Animals , Cell Death/drug effects , Chemokine CXCL2/biosynthesis , Cytokines/biosynthesis , DNA/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , I-kappa B Proteins/metabolism , In Vitro Techniques , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Liver/blood supply , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , NF-KappaB Inhibitor alpha , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/deficiency , Peptidylprolyl Isomerase/genetics , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology
12.
J Biol Chem ; 284(25): 16840-16847, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19401603

ABSTRACT

Neurodegenerative tauopathies, including Alzheimer disease, are characterized by abnormal hyperphosphorylation of the microtubule-associated protein Tau. One group of tauopathies, known as frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17), is directly associated with mutations of the gene tau. However, it is unknown why mutant Tau is highly phosphorylated in the patient brain. In contrast to in vivo high phosphorylation, FTDP-17 Tau is phosphorylated less than wild-type Tau in vitro. Because phosphorylation is a balance between kinase and phosphatase activities, we investigated dephosphorylation of mutant Tau proteins, P301L and R406W. Tau phosphorylated by Cdk5-p25 was dephosphorylated by protein phosphatases in rat brain extracts. Compared with wild-type Tau, R406W was dephosphorylated faster and P301L slower. The two-dimensional phosphopeptide map analysis suggested that faster dephosphorylation of R406W was due to a lack of phosphorylation at Ser-404, which is relatively resistant to dephosphorylation. We studied the effect of the peptidyl-prolyl isomerase Pin1 or microtubule binding on dephosphorylation of wild-type Tau, P301L, and R406W in vitro. Pin1 catalyzes the cis/trans isomerization of phospho-Ser/Thr-Pro sequences in a subset of proteins. Dephosphorylation of wild-type Tau was reduced in brain extracts of Pin1-knockout mice, and this reduction was not observed with P301L and R406W. On the other hand, binding to microtubules almost abolished dephosphorylation of wild-type and mutant Tau proteins. These results demonstrate that mutation of Tau and its association with microtubules may change the conformation of Tau, thereby suppressing dephosphorylation and potentially contributing to the etiology of tauopathies.


Subject(s)
Microtubules/metabolism , Mutation , Peptidylprolyl Isomerase/metabolism , tau Proteins/genetics , tau Proteins/metabolism , Amino Acid Substitution , Animals , Base Sequence , Brain/metabolism , DNA Primers/genetics , Humans , In Vitro Techniques , Kinetics , Mice , Mice, Knockout , Models, Neurological , NIMA-Interacting Peptidylprolyl Isomerase , Peptide Mapping , Peptidylprolyl Isomerase/deficiency , Peptidylprolyl Isomerase/genetics , Phosphorylation , Protein Conformation , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tauopathies/etiology , Tauopathies/genetics , Tauopathies/metabolism , tau Proteins/chemistry
13.
Mol Cancer ; 7: 91, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19077306

ABSTRACT

UNLABELLED: Overexpression of HER-2/Neu occurs in about 25-30% of breast cancer patients and is indicative of poor prognosis. While Her2/Neu overexpression is primarily a result of erbB2 amplification, it has recently been recognized that erbB2 levels are also regulated on the protein level. However, factors that regulate Her2/Neu protein stability are less well understood. The prolyl isomerase Pin1 catalyzes the isomerization of specific pSer/Thr-Pro motifs that have been phosphorylated in response to mitogenic signaling. We have previously reported that Pin1-catalyzed post-phosphorylational modification of signal transduction modulates the oncogenic pathways downstream from c-neu. The goal of this study was to examine the expression of prolyl isomerase Pin1 in human Her2+ breast cancer, and to study if Pin1 affects the expression of Her2/Neu itself. METHODS: Immunohistochemistry for Her2 and Pin1 were performed on two hundred twenty-three human breast cancers, with 59% of the specimen from primary cancers and 41% from metastatic sites. Pin1 inhibition was achieved using siRNA in Her2+ breast cancer cell lines, and its effects were studied using cell viability assays, immunoblotting and immunofluorescence. RESULTS: Sixty-four samples (28.7%) stained positive for Her2 (IHC 3+), and 54% (122/223) of all breast cancers stained positive for Pin1. Of the Her2-positive cancers 40 (62.5%) were also Pin1-positive, based on strong nuclear or nuclear and cytoplasmic staining. Inhibition of Pin1 via RNAi resulted in significant suppression of Her2-positive tumor cell growth in BT474, SKBR3 and AU565 cells. Pin1 inhibition greatly increased the sensitivity of Her2-positive breast cancer cells to the mTOR inhibitor Rapamycin, while it did not increase their sensitivity to Trastuzumab, suggesting that Pin1 might act on Her2 signaling. We found that Pin1 interacted with the protein complex that contains ubiquitinated erbB2 and that Pin1 inhibition accelerated erbB2 degradation, which could be prevented by treatments with the proteasome inhibitor ALLnL. CONCLUSION: Pin1 is a novel regulator of erbB2 that modulates the ubiquitin-mediated degradation of erbB2. The overexpression of Pin1 in a majority of Her2-overexpressing breast cancer may contribute to maintain erbB2 levels. Pin1 inhibition alone and in conjunction with mTOR inhibition suppresses the growth of Her2+ breast cancer cells.


Subject(s)
Breast Neoplasms/enzymology , Peptidylprolyl Isomerase/metabolism , Receptor, ErbB-2/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Humans , Immunohistochemistry , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/deficiency , Peptidylprolyl Isomerase/genetics , Proteasome Inhibitors , Protein Processing, Post-Translational/drug effects , Protein Stability/drug effects , Sirolimus/pharmacology , Transcription, Genetic/drug effects , Trastuzumab
14.
J Neurosci ; 27(31): 8395-404, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17670986

ABSTRACT

Although oligodendrocytes undergo apoptosis after spinal cord injury, molecular mechanisms responsible for their death have been unknown. We report that oligodendrocyte apoptosis is regulated oppositely by c-Jun N-terminal kinase 3 (JNK3) and protein interacting with the mitotic kinase, never in mitosis A I (Pin1), the actions of which converge on myeloid cell leukemia sequence-1 (Mcl-1). Activated after injury, JNK3 induces cytochrome c release by facilitating the degradation of Mcl-1, the stability of which is maintained in part by Pin1. Pin1 binds Mcl-1 at its constitutively phosphorylated site, Thr163Pro, and stabilizes it by inhibiting ubiquitination. After injury JNK3 phosphorylates Mcl-1 at Ser121Pro, facilitating the dissociation of Pin1 from Mcl-1. JNK3 thus induces Mcl-1 degradation by counteracting the protective binding of Pin1. These results are confirmed by the opposing phenotypes observed between JNK3-/- and Pin1-/- mice: oligodendrocyte apoptosis and cytochrome c release are reduced in JNK3-/- but elevated in Pin1-/- mice. This report thus unveils a mechanism by which cytochrome c release is under the opposite control of JNK3 and Pin1, regulators for which the activities are intricately coupled.


Subject(s)
Apoptosis/physiology , Mitogen-Activated Protein Kinase 10/physiology , Oligodendroglia/enzymology , Peptidylprolyl Isomerase/physiology , Spinal Cord Injuries/enzymology , Animals , Apoptosis/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase 10/deficiency , Mitogen-Activated Protein Kinase 10/genetics , NIMA-Interacting Peptidylprolyl Isomerase , Oligodendroglia/cytology , Oligodendroglia/pathology , Peptidylprolyl Isomerase/deficiency , Peptidylprolyl Isomerase/genetics , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Ubiquitin/antagonists & inhibitors , Ubiquitin/metabolism
15.
EMBO J ; 26(7): 1761-71, 2007 Apr 04.
Article in English | MEDLINE | ID: mdl-17347650

ABSTRACT

The microtubule binding protein gephyrin plays a prominent role in establishing and maintaining a high concentration of inhibitory glycine receptors juxtaposed to presynaptic releasing sites. Here, we show that endogenous gephyrin undergoes proline-directed phosphorylation, which is followed by the recruitment of the peptidyl-prolyl isomerase Pin1. The interaction between gephyrin and Pin1 is strictly dependent on gephyrin phosphorylation and requires serine-proline consensus sites encompassing the gephyrin proline-rich domain. Upon binding, Pin1 triggers conformational changes in the gephyrin molecule, thus enhancing its ability to bind the beta subunit of GlyRs. Consistently, a downregulation of GlyR clusters was detected in hippocampal neurons derived from Pin1 knockout mice, which was paralleled by a reduction in the amplitude of glycine-evoked currents. Our results suggest that phosphorylation-dependent prolyl isomerisation of gephyrin represents a mechanism for regulating GlyRs function.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , Peptidylprolyl Isomerase/metabolism , Receptors, Glycine/metabolism , Animals , Brain/metabolism , Carrier Proteins/chemistry , Cytoplasmic Structures/metabolism , Epitopes , Evoked Potentials , Hippocampus/cytology , Hippocampus/enzymology , Humans , Membrane Proteins/chemistry , Mice , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Neurons/cytology , Neurons/enzymology , Peptidylprolyl Isomerase/deficiency , Phosphorylation , Proline/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Subunits/metabolism , Recombinant Proteins/metabolism , Serine/metabolism
16.
J Biol Chem ; 282(17): 12813-21, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17339317

ABSTRACT

The FK506-binding proteins (FKBs) represent ubiquitous enzymes that catalyze the rate-limiting peptidyl prolyl cis-trans isomerization step in protein folding. The nematode Caenorhabditis elegans has eight FKBs, three of which (FKB-3, -4, and -5) have dual peptidyl prolyl cis-trans isomerase (PPIase) domains, signal peptides and ER retention signals. PPIase activity has been detected for recombinant FKB-3. Both FKB-3 and -5 are expressed in the exoskeleton-synthesizing hypodermis with transcript peaks that correspond to the molting and collagen synthesis cycles. FKB-4 is expressed at a low level throughout development. No phenotypes were observed in deletion mutants in each of the secretory pathway FKBs. Combined triple and fkb-4, -5 double deletion mutants were however found to arrest at 12 degrees C, but developed normally at 15-25 degrees C. This cold-sensitive larval lethal effect was not maternally derived, occurred during embryogenesis, and could be rescued following the transgenic introduction of a wild type copy of either fkb-4 or fkb-5. The temperature-sensitive defects also affected molting, cuticle collagen expression, hypodermal seam cell morphology, and the structural integrity of the cuticular extracellular matrix. This study establishes that the secretory pathway FK506-binding PPIase enzymes are essential for normal nematode development, collagen biogenesis, and the formation of an intact exoskeleton under adverse physiological conditions.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/metabolism , Collagen/metabolism , Extracellular Matrix/metabolism , Peptidylprolyl Isomerase/deficiency , Tacrolimus Binding Proteins/deficiency , Animals , Animals, Genetically Modified , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Cold Temperature , Extracellular Matrix/pathology , Gene Deletion , Molting/genetics
17.
PLoS One ; 2(2): e226, 2007 Feb 21.
Article in English | MEDLINE | ID: mdl-17311089

ABSTRACT

UNLABELLED: BACKGROUND/ABSTRACT: Immune responses initiated by T cell receptor (TCR) and costimulatory molecule mediated signaling culminate in maximal cytokine mRNA production and stability. The transcriptional responses to co-stimulatory T cell signalling involve calcineurin and NF-AT, which can be antagonized by interference with the cis-trans peptidyl-prolyl isomerases (PPIase), cyclophilin A and FKBP. Signalling molecules downstream of CD28 which are essential for the stabilization of cytokine mRNAs are largely unknown. METHODOLOGY/PRINCIPAL FINDINGS: We now show that Pin1, a third member of the PPIase family mediates the post-transcriptional regulation of Th1 cytokines by activated T cells. Blockade of Pin1 by pharmacologic or genetic means greatly attenuated IFN-gamma, IL-2 and CXCL-10 mRNA stability, accumulation and protein expression after cell activation. In vivo, Pin1 blockade prevented both the acute and chronic rejection of MHC mismatched, orthotopic rat lung transplants by reducing the expression of IFN-gamma and CXCL-10. Combined transcriptional and post-transcriptional blockade with cyclosporine A and the Pin1 inhibitor, juglone, was synergistic. CONCLUSIONS/SIGNIFICANCE: These data suggest Pin1 inhibitors should be explored for use as immunosuppressants and employed with available calcineurin inhibitors to reduce toxicity and enhance effectiveness.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Peptidylprolyl Isomerase/physiology , Th1 Cells/immunology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/genetics , Cyclosporine/pharmacology , Graft Rejection/immunology , Graft Rejection/prevention & control , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Interleukin-2/genetics , Lung Transplantation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Naphthoquinones/pharmacology , Peptidylprolyl Isomerase/deficiency , Peptidylprolyl Isomerase/genetics , RNA Processing, Post-Transcriptional/drug effects , RNA Stability , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Inbred F344 , Rats, Inbred WKY , Transcription, Genetic/drug effects , Transplantation, Homologous/immunology
18.
Oncogene ; 26(26): 3835-45, 2007 May 31.
Article in English | MEDLINE | ID: mdl-17160015

ABSTRACT

Tumor suppressor p53 is essential for checkpoint control in response to a variety of genotoxic stresses. DNA damage leads to phosphorylation on the Ser/Thr-Pro motifs of p53, which facilitates interaction with Pin1, a pSer/pThr-Pro-specific peptidyl prolyl isomerase. Pin1 is required for the timely activation of p53, resulting in apoptosis or cell cycle arrest. To investigate the physiological relationship between Pin1 and p53, we created Pin1-/-p53-/- mice. These p53-deficient mice spontaneously developed lymphomas, mainly of thymic origin, as well as generalized lymphoma infiltration into other organs, including the liver, kidneys and lungs. Ablation of Pin1, in addition to p53, accelerated the thymic hyperplasia, but the thymocytes in these Pin1-/-p53-/- mice did not infiltrate other organs. The thymocytes in 12-week-old Pin1-/-p53-/- mice were CD4(-)CD8(-) (double negative) and had significantly higher levels of the intracellular form of Notch1 (NIC) than the thymocytes of p53-/- or wild-type mice. Presenilin-1, a cleavage enzyme for NIC generation from full-length Notch1 was increased in the thymocytes of Pin1-/-p53-/- mice. Pin1 depletion also inhibited the degradation of NIC by proteasomes. These results suggest that both Pin1 and p53 control the normal proliferation and differentiation of thymocytes by regulating the NIC level.


Subject(s)
Peptidylprolyl Isomerase/deficiency , Receptor, Notch1/metabolism , T-Lymphocytes/metabolism , Thymus Hyperplasia/metabolism , Tumor Suppressor Protein p53/deficiency , Animals , Blotting, Western , Female , Flow Cytometry , Intracellular Fluid/chemistry , Male , Mice , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Presenilin-1/metabolism , T-Lymphocytes/immunology , Thymus Hyperplasia/genetics , Thymus Hyperplasia/pathology , Tumor Suppressor Protein p53/genetics
20.
Mol Cell Biol ; 26(4): 1463-79, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16449657

ABSTRACT

Phosphorylation on Ser/Thr-Pro motifs is a major mechanism regulating many events involved in cell proliferation and transformation, including centrosome duplication, whose defects have been implicated in oncogenesis. Certain phosphorylated Ser/Thr-Pro motifs can exist in two distinct conformations whose conversion in certain proteins is catalyzed specifically by the prolyl isomerase Pin1. Pin1 is prevalently overexpressed in human cancers and is important for the activation of multiple oncogenic pathways, and its deletion suppresses the ability of certain oncogenes to induce cancer in mice. However, little is known about the role of Pin1 in centrosome duplication and the significance of Pin1 overexpression in cancer development in vivo. Here we show that Pin1 overexpression correlates with centrosome amplification in human breast cancer tissues. Furthermore, Pin1 localizes to and copurifies with centrosomes in interphase but not mitotic cells. Moreover, Pin1 ablation in mouse embryonic fibroblasts drastically delays centrosome duplication without affecting DNA synthesis and Pin1 inhibition also suppresses centrosome amplification in S-arrested CHO cells. In contrast, overexpression of Pin1 drives centrosome duplication and accumulation, resulting in chromosome missegregation, aneuploidy, and transformation in nontransformed NIH 3T3 cells. More importantly, transgenic overexpression of Pin1 in mouse mammary glands also potently induces centrosome amplification, eventually leading to mammary hyperplasia and malignant mammary tumors with overamplified centrosomes. These results demonstrate for the first time that the phosphorylation-specific isomerase Pin1 regulates centrosome duplication and its deregulation can induce centrosome amplification, chromosome instability, and oncogenesis.


Subject(s)
Centrosome/metabolism , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/metabolism , Amino Acid Motifs , Aneuploidy , Animals , Breast Neoplasms/etiology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CHO Cells , Cell Transformation, Neoplastic , Chromosomal Instability , Cricetinae , Female , Gene Expression , Humans , Interphase , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Mice, Transgenic , Mitosis , NIH 3T3 Cells , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/deficiency , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...