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










Publication year range
1.
Cells ; 10(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34943932

ABSTRACT

The extracellular protozoan parasite Giardia duodenalis is a well-known and important causative agent of diarrhea on a global scale. Macrophage pyroptosis has been recognized as an important innate immune effector mechanism against intracellular pathogens. Yet, the effects of noninvasive Giardia infection on macrophage pyroptosis and the associated molecular triggers and regulators remain poorly defined. Here we initially observed that NLRP3 inflammasome-mediated pyroptosis was activated in Giardia-treated macrophages, and inhibition of ROS, NLRP3, or caspase-1 could block GSDMD cleavage, IL-1ß, IL-18 and LDH release, and the cell viability reduction. We also confirmed that Giardia-induced NLRP3 inflammasome activation was involved in its K63 deubiquitination. Thus, six candidate deubiquitinases were screened, among which A20 was identified as an effective regulator. We then screened TLRs on macrophage membranes and found that upon stimulation TLR4 was tightly correlated to ROS enhancement, A20-mediated NLRP3 deubiquitination, and pyroptotic signaling. In addition, several Giardia-secreted proteins were predicted as trigger factors via secretome analysis, of which peptidyl-prolyl cis-trans isomerase B (PPIB) independently induced macrophage pyroptosis. This was similar to the findings from the trophozoite treatment, and also led to the TLR4-mediated activation of NLRP3 through K63 deubiquitination by A20. Collectively, the results of this study have significant implications for expanding our understanding of host defense mechanisms after infection with G. duodenalis.


Subject(s)
Diarrhea/genetics , Giardia lamblia/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Animals , Cell Survival/drug effects , Deubiquitinating Enzymes/genetics , Diarrhea/immunology , Diarrhea/parasitology , Disease Models, Animal , Giardia lamblia/immunology , Giardia lamblia/pathogenicity , Host-Parasite Interactions/drug effects , Host-Parasite Interactions/immunology , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Inflammasomes/drug effects , Inflammasomes/immunology , Interleukin-18/genetics , Intracellular Signaling Peptides and Proteins/genetics , Isoenzymes/pharmacology , Macrophages/drug effects , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Peptidylprolyl Isomerase/pharmacology , Phosphate-Binding Proteins/genetics , Pyroptosis/drug effects , Pyroptosis/immunology , Reactive Oxygen Species/metabolism , Toll-Like Receptor 4/immunology , Trophozoites/drug effects , Trophozoites/pathogenicity , Tumor Necrosis Factor alpha-Induced Protein 3/immunology , Ubiquitination/genetics
2.
Sci Rep ; 7: 43322, 2017 02 27.
Article in English | MEDLINE | ID: mdl-28240301

ABSTRACT

Our previous studies revealed that many proteins in addition to the known allergens of D. farinae have not been fully characterized. We observed that Pplase did not respond to serum collected from patients sensitized to D. farinae. In a mouse model, Pplase significantly enhanced airway hyperresponsiveness (AHR) and Th2 responses induced by ovalbumin (OVA) compared with mice treated with OVA alone. Moreover, exposure to Pplase significantly increased the expression of IRF4, CD80, CD83, MHCII and TNF-α in DC2.4 cells, which was abolished in the presence of a TLR4 inhibitor. In vitro T cell polarization experiments revealed that Pplase alone could not induce T cell polarization but enhanced T cell polarization together with OVA. In addition, transfer of Pplase-primed bone marrow-derived DCs (BMDCs) to naïve mice enhanced AHR and Th2 immune responses in mice sensitized to OVA. In conclusion, Pplase is not an allergen of D. farinae but can activate DC cells to facilitate OVA-induced allergic responses.


Subject(s)
Arthropod Proteins/pharmacology , Dendritic Cells/immunology , Dermatophagoides farinae/immunology , Peptidylprolyl Isomerase/pharmacology , Respiratory Hypersensitivity/immunology , Adult , Allergens/administration & dosage , Animals , Antigens, CD/genetics , Antigens, CD/immunology , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Cell Line , Dendritic Cells/drug effects , Dendritic Cells/pathology , Dendritic Cells/transplantation , Dermatophagoides farinae/chemistry , Disease Models, Animal , Female , Gene Expression Regulation , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Immunoglobulins/genetics , Immunoglobulins/immunology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Macrophage Activation/drug effects , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Middle Aged , Ovalbumin/administration & dosage , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/immunology , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/pathology , Signal Transduction , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , CD83 Antigen
3.
J Biomol Screen ; 21(7): 701-12, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27185744

ABSTRACT

In this work, a sensitive and convenient protease-based fluorimetric high-throughput screening (HTS) assay for determining peptidyl-prolyl cis-trans isomerase activity was developed. The assay was based on a new intramolecularly quenched substrate, whose fluorescence and structural properties were examined together with kinetic constants and the effects of solvents on its isomerization process. Pilot screens performed using the Library of Pharmacologically Active Compounds (LOPAC) and cyclophilin A (CypA), as isomerase model enzyme, indicated that the assay was robust for HTS, and that comparable results were obtained with a CypA inhibitor tested both manually and automatically. Moreover, a new compound that inhibits CypA activity with an IC50 in the low micromolar range was identified. Molecular docking studies revealed that the molecule shows a notable shape complementarity with the catalytic pocket confirming the experimental observations. Due to its simplicity and precision in the determination of extent of inhibition and reaction rates required for kinetic analysis, this assay offers many advantages over other commonly used assays.


Subject(s)
Cyclophilin A/antagonists & inhibitors , Enzyme Inhibitors/isolation & purification , High-Throughput Screening Assays/methods , Peptidylprolyl Isomerase/chemistry , Catalytic Domain , Cyclophilin A/chemistry , Enzyme Inhibitors/chemistry , Fluorescence Resonance Energy Transfer/methods , Humans , Kinetics , Magnetic Resonance Spectroscopy/methods , Molecular Docking Simulation/methods , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/pharmacology , Solvents/chemistry , Substrate Specificity
4.
Biosci Biotechnol Biochem ; 74(10): 2067-70, 2010.
Article in English | MEDLINE | ID: mdl-20944414

ABSTRACT

The phenotypes of mice lacking peptidyl prolyl cis/trans isomerase Pin1 (Pin1(-/-)) indicated that deficient Pin1 might be related to a variety of diseases. We created TAT-Pin1, a fusion protein of human immunodeficiency virus 1 trans-activator of transcription factor with Pin1. Treatment of HeLa cells with TAT-Pin1 increased the ratio of the S phase. Moreover, TAT-Pin1 restored the proliferating function of Pin1(-/-) mouse embryonic fibroblasts which cannot restart proliferation after G0 arrest. These results indicate that TAT-Pin1 is useful in studying the functions of Pin1 and can be developed as a macromolecular drug for diseases related to Pin1 loss.


Subject(s)
Peptidylprolyl Isomerase/chemistry , Peptidylprolyl Isomerase/metabolism , Protein Engineering/methods , Amino Acid Sequence , Animals , HeLa Cells , Humans , Mice , Models, Molecular , NIMA-Interacting Peptidylprolyl Isomerase , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/pharmacology , Protein Transport , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , S Phase/drug effects , tat Gene Products, Human Immunodeficiency Virus/chemistry
5.
J Proteome Res ; 9(11): 6060-70, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20804218

ABSTRACT

We present the first study of protein regulation by ligands in Caenorhabditis elegans. The ligands were peptidyl-prolyl isomerase inhibitors of cyclophilins. Up-regulation is observed for several heat shock proteins and one ligand in particular caused a greater than 2-fold enhancement of cyclophilin CYN-5. Additionally, several metabolic enzymes display elevated levels. This approach, using label-free relative quantification, provides an extremely attractive way of measuring the effect of ligands on an entire proteome, with minimal sample pretreatment, which could be applicable to large-scale studies. In this initial study, which compares the effect of three ligands, 54 unique proteins have been identified that are up- (51) or down- (3) regulated in the presence of a given ligand. A total of 431 C. elegans proteins were identified. Our methodology provides an intriguing new direction for in vivo screening of the effects of novel and untested ligands at the whole organism level.


Subject(s)
Caenorhabditis elegans/chemistry , Cyclophilins/drug effects , Gene Expression Regulation/drug effects , Proteome/drug effects , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cyclophilins/genetics , Ligands , Peptidylprolyl Isomerase/pharmacology
6.
Exp Biol Med (Maywood) ; 234(10): 1155-65, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19596832

ABSTRACT

Autosomal dominant polycystic kidney (ADPKD) is highly prevalent genetic disease. Liver cyst disease is the most common extrarenal manifestation in ADPKD and accounts for up to 10% of ADPKD morbidity and mortality. The clinical features of ADPKD liver disease arise from dramatic increases in liver cyst volumes. To identify mechanisms that promote liver cyst growth, the present study characterized the degree of vascularization of liver cyst walls and determined that cyst-specific cytokines and growth factors can drive endothelial cell proliferation and development. Microscopic techniques demonstrated liver cyst walls are well vascularized. A comparative analysis found the vascular density in free liver cyst walls was greater in mice than in humans. Treatment of human micro-vascular endothelial cells (HMEC-1) with human liver cyst fluid (huLCF) induced a rapid increase in vascular endothelium growth factor receptor 2 (VEGFR2) phosphorylation that persisted for 45-60 min and was blocked by 20 microM SU5416, a VEGFR tyrosine kinase inhibitor. Similarly, huLCF treatment of HMEC-1 cells induced an increase in the cell proliferation rate (131 +/- 6% of control levels; P > 0.05) and the degree of vascular development ('tube' diameter assay: 92 +/- 14 microm for huLCF vs. 12 +/- 7 microm for vehicle); P > 0.05). Both cell proliferation and vascular development were sensitive to SU5416. These studies indicate that factors secreted by liver cyst epithelia can activate VEGF signaling pathways and induce endothelial cell proliferation and differentiation. The present studies suggest that targeting VEGFR2-dependent angiogenesis may be an effective therapeutic strategy in blocking ADPKD liver cyst vascularization and growth.


Subject(s)
Cell Proliferation/drug effects , Cysts/metabolism , Cytokines/pharmacology , Endothelial Cells/physiology , Liver Diseases/metabolism , Animals , Cells, Cultured , Cyst Fluid/metabolism , Cysts/blood supply , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Humans , Indoles/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Neovascularization, Pathologic/metabolism , Peptidylprolyl Isomerase/pharmacology , Phosphorylation , Polycystic Kidney, Autosomal Dominant/metabolism , Pyrroles/pharmacology , TRPP Cation Channels/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Exp Cell Res ; 314(3): 616-28, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18054915

ABSTRACT

Initially identified as a cyclosporin-A binding protein, cyclophilin B (CyPB) is an inflammatory mediator that induces adhesion of T lymphocytes to fibronectin, by a mechanism dependent on CD147 and alpha 4 beta 1 integrins. Recent findings have suggested that another cell membrane protein, CD98, may cooperate with CD147 to regulate beta1 integrin functions. Based on these functional relationships, we examined the contribution of CD98 in the pro-adhesive activity of CyPB, by utilizing the responsive promonocyte cell line THP-1. We demonstrated that cross-linking CD98 with CD98-AHN-18 antibody mimicked the responses induced by CyPB, i.e. homotypic aggregation, integrin-mediated adhesion to fibronectin and activation of p44/42 MAPK. Consistent with previous data, immunoprecipitation confirmed the existence of a heterocomplex wherein CD147, CD98 and beta1 integrins were associated. We then demonstrated that CyPB-induced cell adhesion and p44/42 MAPK activation were dependent on the participation of phosphoinositide 3-kinase and subsequent activation of protein kinase C-delta. Finally, silencing the expression of CD98 by RNA interference potently reduced CyPB-induced cell responses, thus confirming the role of CD98 in the pro-adhesive activity of CyPB. Altogether, our results support a model whereby CyPB induces integrin-mediated adhesion via interaction with a multimolecular unit formed by the association between CD147, CD98 and beta1 integrins.


Subject(s)
Cell Membrane/metabolism , Cyclophilins/physiology , Fusion Regulatory Protein-1/metabolism , Integrins/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Peptidylprolyl Isomerase/physiology , Protein Kinase C-delta/metabolism , Antibodies/pharmacology , Basigin/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line, Tumor , Cell Membrane/drug effects , Cyclophilins/genetics , Cyclophilins/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Fibronectins/metabolism , Fusion Regulatory Protein-1/antagonists & inhibitors , Fusion Regulatory Protein-1/genetics , Humans , Integrin beta1/metabolism , Integrins/drug effects , Integrins/genetics , Macromolecular Substances/metabolism , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C-delta/drug effects , RNA Interference/physiology
8.
FASEB J ; 20(3): 524-6, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16410343

ABSTRACT

Aggregation of alpha-synuclein (alpha-SYN) plays a key role in Parkinson's disease (PD). We have used fluorescence correlation spectroscopy (FCS) to study alpha-SYN aggregation in vitro and discovered that this process is clearly accelerated by addition of FK506 binding proteins (FKBPs). This effect was observed both with E. coli SlyD FKBP and with human FKBP12 and was counteracted by FK506, a specific inhibitor of FKBP. The alpha-SYN aggregates formed in the presence of FKBP12 showed fibrillar morphology. The rotamase activity of FKBP apparently accelerates the folding and subsequent aggregation of alpha-SYN. Since FK506 and other non-immunosuppressive FKBP inhibitors are known to display neuroregenerative and neuroprotective properties in disease models, the observed inhibition of rotamase activity and alpha-SYN aggregation, may explain their mode of action. Our results open perspectives for the treatment of PD with immunophilin ligands that inhibit a specific member of the FKBP family.


Subject(s)
Escherichia coli Proteins/pharmacology , Peptidylprolyl Isomerase/pharmacology , Tacrolimus Binding Protein 1A/pharmacology , alpha-Synuclein/chemistry , Escherichia coli Proteins/isolation & purification , Escherichia coli Proteins/physiology , Humans , Microscopy, Electron , Nephelometry and Turbidimetry , Peptidylprolyl Isomerase/isolation & purification , Peptidylprolyl Isomerase/physiology , Protein Folding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/drug effects , Spectrometry, Fluorescence , Tacrolimus/pharmacology , Tacrolimus Binding Protein 1A/physiology , alpha-Synuclein/drug effects , alpha-Synuclein/genetics , alpha-Synuclein/ultrastructure
9.
Med Sci (Paris) ; 19(12): 1251-8, 2003 Dec.
Article in French | MEDLINE | ID: mdl-14691750

ABSTRACT

Peptidyl-prolyl isomerases (PPIases) are chaperone enzymes which alter the peptide bond between a given amino acid and a proline, changing it from the cis to the trans conformation and vice versa. This modification can cause dramatic structural modifications which can affect the properties of targeted proteins. The ubiquitous PPIase Pin1, conserved from yeast to human, has been shown to be necessary for entry into mitosis. The yeast homologue, Ess1, is essential for cell survival. Pin1 possesses a WW domain which specifically recognizes pSer-Pro and pThr-Pro motifs in which the first amino acid is phosphorylated. Pin1 binds to many proteins implicated in cell cycle regulation (e.g. p53, Myt1, Wee1, and Cdc25C). Pin1 also targets tau, a protein forming part of hte neuronal cytoskeleton which is hyper-phosphorylated in patients suffering from Alzheimer's disease (AD). Pin1 could, therefore, be involved in the pathogenesis of Ad. Furthermore, Pin1 also binds two proteins involved in transcription: Rpb1, the largest subunit of RNA polymerase II and Spt5, a regulator of the elongation of transcription. Both theses proteins possess domains rich in S/T-P motifs which can be targeted by Pin1 when phosphorylated. Recent studies show that Pin1 modulates the dephosphorylation of some proteins by allowing trans-specific phosphatases to recognize their target after isomerization. This unexpected role might allow protein regulation via peptidyl-prolyl isomerase activity.


Subject(s)
Alzheimer Disease/physiopathology , Cell Survival , Peptidylprolyl Isomerase/pharmacology , Cell Cycle/physiology , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/metabolism , Phosphorylation , Protein Binding , Transcription, Genetic , tau Proteins/metabolism
10.
Mol Cell Biol ; 22(15): 5281-95, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12101225

ABSTRACT

Oncogenes Neu/HER2/ErbB2 and Ras can induce mammary tumorigenesis via upregulation of cyclin D1. One major regulatory mechanism in these oncogenic signaling pathways is phosphorylation of serines or threonines preceding proline (pSer/Thr-Pro). Interestingly, the pSer/Thr-Pro motifs in proteins exist in two completely distinct cis and trans conformations, whose conversion is catalyzed specifically by the essential prolyl isomerase Pin1. By isomerizing pSer/Thr-Pro bonds, Pin1 can regulate the conformation and function of certain phosphorylated proteins. We have previously shown that Pin1 is overexpressed in breast tumors and positively regulates cyclin D1 by transcriptional activation and posttranslational stabilization. Moreover, in Pin1 knockout mice, mammary epithelial cells fail to undergo massive proliferation during pregnancy, as is the case in cyclin D1 null mice. These results indicate that Pin1 is upregulated in breast cancer and may be involved in mammary tumors. However, the mechanism of Pin1 overexpression in cancer and its significance in cell transformation remain largely unknown. Here we demonstrate that PIN1 expression is mediated by the transcription factor E2F and enhanced by c-Neu and Ha-Ras via E2F. Furthermore, overexpression of Pin1 not only confers transforming properties on mammary epithelial cells but also enhances the transformed phenotypes of Neu/Ras-transformed mammary epithelial cells. In contrast, inhibition of Pin1 suppresses Neu- and Ras-induced transformed phenotypes, which can be fully rescued by overexpression of a constitutively active cyclin D1 mutant that is refractory to the Pin1 inhibition. Thus, Pin1 is an E2F target gene that is essential for the Neu/Ras-induced transformation of mammary epithelial cells through activation of cyclin D1.


Subject(s)
Cell Cycle Proteins , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins , Epithelial Cells/metabolism , Peptidylprolyl Isomerase/genetics , Receptor, ErbB-2/metabolism , Transcription Factors/metabolism , ras Proteins/metabolism , Base Sequence , Breast/cytology , Breast/metabolism , Cell Division/physiology , Cell Line , Cell Line, Transformed , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cyclin D1/metabolism , E2F Transcription Factors , Epithelial Cells/cytology , Female , Gene Expression Regulation, Neoplastic/physiology , Genes, Dominant , Humans , Molecular Sequence Data , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/metabolism , Peptidylprolyl Isomerase/pharmacology , Phenotype , Promoter Regions, Genetic/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection
11.
Mol Cell ; 7(5): 1071-83, 2001 May.
Article in English | MEDLINE | ID: mdl-11389853

ABSTRACT

Pin1 is an essential protein that can peptidyl-prolyl-isomerize small phosphopeptides. It has been suggested that Pin1 regulates entry into mitosis by catalyzing the cis/trans-isomerization of prolines on critical protein substrates in response to phosphorylation. We show that Pin1 catalytically generates a conformational change on the mitotic phosphatase Cdc25, as assayed by limited protease digestion, differential reactivity to a phosphoserine-proline-directed monoclonal antibody (MPM-2), and by changes in Cdc25 enzymatic activity. Pin1 catalytically modifies the conformation of Cdc25 at stoichiometries less than 0.0005, and mutants of Pin1 in the prolyl isomerase domain are not active. We suggest that, although difficult to detect, phosphorylation-dependent conformational changes mediated by prolyl isomerization may play an important regulatory role in the cell cycle.


Subject(s)
Peptidylprolyl Isomerase/pharmacology , cdc25 Phosphatases/drug effects , Animals , CDC2 Protein Kinase/drug effects , CDC2 Protein Kinase/metabolism , CDC2 Protein Kinase/pharmacology , Catalytic Domain/genetics , Dose-Response Relationship, Drug , Humans , Kinetics , Mutation , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics , Phosphorylation , Proline/drug effects , Protein Conformation , Xenopus , cdc25 Phosphatases/chemistry , cdc25 Phosphatases/metabolism
12.
J Biol Chem ; 276(10): 7027-32, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11096119

ABSTRACT

Targets of rapamycin (TORs) are conserved phosphatidylinositol kinase-related kinases that are involved in the coordination between nutritional or mitogenic signals and cell growth. Here we report the initial characterization of two Schizosaccharomyces pombe TOR homologs, tor1(+) and tor2(+). tor2(+) is an essential gene, whereas tor1(+) is required only under starvation and other stress conditions. Specifically, Deltator1 cells fail to enter stationary phase or undergo sexual development and are sensitive to cold, osmotic stress, and oxidative stress. In complex with the prolyl isomerase FKBP12, the drug rapamycin binds a conserved domain in TORs, FRB, thus inhibiting some of the functions of TORs. Mutations at a conserved serine within the FRB domain of Saccharomyces cerevisiae TOR proteins led to rapamycin resistance but did not otherwise affect the functions of the proteins. The S. pombe tor1(+) exhibits different features; substitution of the conserved serine residue, Ser(1834), with arginine compromises its functions and has no effect on the inhibition that rapamycin exerts on sexual development in S. pombe.


Subject(s)
Fungal Proteins/chemistry , Fungal Proteins/physiology , Phosphatidylinositol 3-Kinases , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/physiology , Protein Kinases/chemistry , Protein Kinases/physiology , Saccharomyces cerevisiae Proteins , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/physiology , Serine/chemistry , Amino Acid Sequence , Antifungal Agents/pharmacology , Arginine/chemistry , Blotting, Western , Cell Cycle Proteins , Cloning, Molecular , Cold Temperature , Conserved Sequence , Drug Resistance , Epitopes/chemistry , Flow Cytometry , Fungal Proteins/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Osmosis , Oxidative Stress , Peptidylprolyl Isomerase/pharmacology , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Binding , Protein Kinases/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Sirolimus/pharmacology , Tacrolimus Binding Protein 1A/pharmacology , Time Factors
13.
Science ; 287(5458): 1644-7, 2000 Mar 03.
Article in English | MEDLINE | ID: mdl-10698738

ABSTRACT

The peptidyl-prolyl isomerase Pin1 has been implicated in regulating cell cycle progression. Pin1 was found to be required for the DNA replication checkpoint in Xenopus laevis. Egg extracts depleted of Pin1 inappropriately transited from the G2 to the M phase of the cell cycle in the presence of the DNA replication inhibitor aphidicolin. This defect in replication checkpoint function was reversed after the addition of recombinant wild-type Pin1, but not an isomerase-inactive mutant, to the depleted extract. Premature mitotic entry in the absence of Pin1 was accompanied by hyperphosphorylation of Cdc25, activation of Cdc2/cyclin B, and generation of epitopes recognized by the mitotic phosphoprotein antibody, MPM-2. Therefore, Pin1 appears to be required for the checkpoint delaying the onset of mitosis in response to incomplete replication.


Subject(s)
Cell Cycle Proteins , DNA Replication , Mitosis , Nuclear Proteins , Peptidylprolyl Isomerase/metabolism , Xenopus Proteins , Animals , Aphidicolin/pharmacology , Cell Cycle , Cyclin B/metabolism , Enzyme Inhibitors/pharmacology , G2 Phase , NIMA-Interacting Peptidylprolyl Isomerase , Nucleic Acid Synthesis Inhibitors , Oocytes , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/pharmacology , Point Mutation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Xenopus laevis , cdc25 Phosphatases/metabolism
14.
Biophys J ; 78(3): 1293-305, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10692317

ABSTRACT

The R domain of cystic fibrosis transmembrane conductance regulator (CFTR), when phosphorylated, undergoes conformational change, and the chloride channel opens. We investigated the contribution of R domain conformation, apart from the changes induced by phosphorylation, to channel opening, by testing the effect of the peptidyl-prolyl isomerase, cyclophilin A, on the CFTR channel. When it was applied after the channel had been opened by PKA phosphorylation, cyclophilin A increased the open probability of wild-type CFTR (from P(o) = 0.197 +/- 0.010 to P(o) = 0.436 +/- 0. 029) by increasing the number of channel openings, not open time. Three highly conserved proline residues in the R domain, at positions 740, 750, and 759, were considered as candidate targets for cyclophilin A. Mutations of these prolines to alanines (P3A mutant) resulted in a channel unresponsive to cyclophilin A but with pore properties similar to the wild type, under strict control of PKA and ATP, but with significantly increased open probability (P(o) = 0.577 +/- 0.090) compared to wild-type CFTR, again due to an increase in the number of channel openings and not open time. Mutation of each of the proline residues separately and in pairs demonstrated that all three proline mutations are required for maximal P(o). When P3A was expressed in 293 HEK cells and tested by SPQ assay, chloride efflux was significantly increased compared to cells transfected with wild-type CFTR. Thus, treatments favoring the trans-peptidyl conformation about conserved proline residues in the R domain of CFTR affect openings of CFTR, above and beyond the effect of PKA phosphorylation.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Ion Channel Gating/physiology , Alanine , Amino Acid Substitution , Cell Line , Conserved Sequence , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Humans , Ion Channel Gating/drug effects , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mutagenesis, Site-Directed , Peptidylprolyl Isomerase/pharmacology , Phosphorylation , Probability , Proline , Protein Conformation/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection
15.
Biochemistry ; 39(2): 453-62, 2000 Jan 18.
Article in English | MEDLINE | ID: mdl-10631007

ABSTRACT

The in vitro protein folding activity of an FKBP (FK506 binding protein, abbreviated to MTFK) from a thermophilic archaeon, Methanococcus thermolithotrophicus, was investigated. MTFK exhibited FK506 sensitive PPIase (peptidyl prolyl cis-trans isomerase) activity which accelerated the speed of ribonuclease T1 refolding, which is rate-limited by isomerization of two prolyl peptide bonds. In addition, MTFK suppressed the aggregation of folding intermediates and elevated the final yield of rhodanese refolding. We called this activity of MTFK the chaperone activity. The chaperone activity of MTFK was also inhibited by FK506. Alignment of the amino acid sequences of MTFK with human FKBP12 showed that MTFK has two insertion sequences, consisting of 13 and 44 amino acids, at the N- and C-termini, respectively [Furutani, M., Iida, T., Yamano, S., Kamino, K., and Maruyama, T. (1998) J. Bacteriol. 180, 388-394]. To study the relationship between chaperone and PPIase activities of MTFK, mutant MTFKs with deletions of these insertion sequences or with amino acid substitutions were created. Their PPIase and chaperone activities were measured using a synthetic oligopeptide and denatured rhodanese as the substrates, respectively. The far-UV circular dichroism spectra of the wild type and the mutants were also analyzed. The results suggested that (1) the PPIase activity did not correlate with chaperone activity, (2) both insertion sequences were required for MTFK to take a proper conformation, and (3) the insertion sequence (44 amino acids) in the C-terminus was important for the chaperone activity.


Subject(s)
Immunophilins/chemistry , Methanococcus/chemistry , Molecular Chaperones/chemistry , Binding Sites , Escherichia coli/metabolism , Gene Expression , Immunophilins/genetics , Immunophilins/isolation & purification , Methanococcus/genetics , Molecular Structure , Mutation , Peptidylprolyl Isomerase/chemistry , Peptidylprolyl Isomerase/pharmacology , Protein Folding , Ribonuclease T1/chemistry , Tacrolimus/chemistry , Tacrolimus Binding Proteins , Thiosulfate Sulfurtransferase/chemistry
16.
J Chromatogr A ; 853(1-2): 469-77, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10486755

ABSTRACT

The affinity capillary electrophoretic separation of the complex of the enzyme cyclophilin (Cyp) with the immunosuppressive drug cyclosporin A (CsA) from uncomplexed Cyp and CsA in phosphate buffer (pH 8) under non-denaturing conditions by equilibrium-mixture analysis is reported. Using a new approach combining mobility-shift analysis and electrophoretically mediated microanalysis the binding constant of rhCyp18 to CsA and derivatives was estimated.


Subject(s)
Cyclosporins/isolation & purification , Electrophoresis, Capillary/methods , Peptidylprolyl Isomerase/isolation & purification , Cyclosporins/pharmacology , Drug Interactions , Drug Monitoring/methods , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Immunosuppressive Agents/isolation & purification , Immunosuppressive Agents/pharmacology , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/pharmacology
17.
Eur J Biochem ; 263(2): 534-42, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10406964

ABSTRACT

Transforming growth factor-beta (TGFbeta) induces gene expression of the glycoprotein clusterin in a variety of cell types via a consensus AP-1 binding site. Here, we demonstrate, by supershift analysis, that JunB, JunD, Fra1, Fra2, and c-Fos bound to AP-1 but that prior treatment of the cells with TGFbeta reduced dramatically c-Fos binding, suggesting that c-Fos might be playing a negative regulatory role in clusterin gene expression. Transient cotransfection assays in mink lung epithelial (CCL64) cells, using a human c-Fos expressing plasmid together with a clusterin promoter/reporter construct or the artificial TGFbeta-inducible reporter construct 3TPLux, revealed that c-Fos was indeed repressive for TGFbeta-induced promoter transactivation. Further, we demonstrate that in stable c-Fos-overexpressing cell lines, TGFbeta induction of endogenous clusterin mRNA, as well as clusterin promoter transactivation are blocked. Co-transfection with c-Fos deletion constructs revealed that the C-terminal region, including the homologue box 2 motif and the extreme C-terminal serine phosphorylation sites (Ser362 and Ser374) are required for repression of clusterin and 3TPLux transactivation. TGFbeta treatment of CCL64 cells resulted in the induction of c-Fos mRNA but caused no alternation in total c-Fos protein levels. The results suggest that the c-Fos represses clusterin gene expression, maintaining a low basal level in the absence of TGFbeta, and that TGFbeta, presumably through its effects on c-Fos protein synthesis and/or stability, abrogates the repression of c-Fos, thereby resulting in gene expression.


Subject(s)
Gene Expression Regulation , Glycoproteins/metabolism , Molecular Chaperones , Proto-Oncogene Proteins c-fos/metabolism , Transforming Growth Factor beta/physiology , Animals , Base Sequence , Cell Line , Clusterin , Dose-Response Relationship, Drug , Epithelial Cells , Humans , Mink/metabolism , Models, Biological , Molecular Sequence Data , Peptidylprolyl Isomerase/pharmacology , Promoter Regions, Genetic , Time Factors , Transfection , Transforming Growth Factor beta/pharmacology
18.
Biochem J ; 341 ( Pt 1): 127-32, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10377253

ABSTRACT

Cyclophilin-A is the cytosolic isoform of a family of peptidylproline cis-trans-isomerases that bind cyclosporin A. This study investigates the role of cyclophilin-A in necrotic cell death, induced by 'chemical ischaemia' and by t-butylhydroperoxide. An 18-mer antisense phosphorothioate oligodeoxynucleotide was used to target a translated region of cyclophilin-A mRNA in rat neonatal cardiomyocytes. After a 24 h exposure to the oligonucleotide, the amount of cyclophilin-A in the cells was decreased by at least 93% as judged by immunological and enzymic criteria. For the enzyme assays, peptidyl proline cis-trans-isomerase activity was measured fluorimetrically in small (10 microl) volumes of cell extract. Immunoblots were developed with a polyclonal anti-cyclophilin-A antibody after sample isoelectric focusing and SDS/PAGE. Cyclophilin-A suppression had no effect on cyanide-plus-2-deoxyglucose-induced cell death. However, cyclophilin-A-suppressed cells were markedly more sensitive to t-butylhydroperoxide. Cyclosporin A conferred some resistance to the peroxide in both types of cell, but protection was greater in cyclophilin-A-suppressed cells, where cyclosporin A increased the survival time 2-fold. It is concluded that two cyclophilin isoforms are involved, in quite different ways, in peroxide-induced cell death. Cyclophilin-A has a protective role. Another isoform, possibly mitochondrial cyclophilin-D, has a deleterious role, such that blockade by cyclosporin A leads to protection.


Subject(s)
Cyclophilins , Heart/drug effects , Oxidative Stress/physiology , Peptidylprolyl Isomerase/drug effects , Peptidylprolyl Isomerase/pharmacology , Animals , Cell Death/drug effects , Cells, Cultured , Cyanides/toxicity , Peptidyl-Prolyl Isomerase F , Gene Expression/drug effects , Immunophilins/drug effects , Myocardium/cytology , Oligodeoxyribonucleotides, Antisense/pharmacology , Rats , Rats, Sprague-Dawley , tert-Butylhydroperoxide/toxicity
19.
Mol Biochem Parasitol ; 99(2): 167-81, 1999 Apr 30.
Article in English | MEDLINE | ID: mdl-10340482

ABSTRACT

Two major protein phosphatase (PP) activities were purified from cytosolic extracts of the erythrocytic stage of the malaria parasite, Plasmodium falciparum. Both enzymes were specific for phosphoserine and phosphothreonine residues with very little activity against phosphotyrosine residues. The biochemical properties of the enzymes suggested their strong similarity with eukaryotic PP2A and PP2B protein phosphatases. Both enzymes preferentially dephosphorylated the alpha subunit of phosphorylase kinase, and were resistant to inhibitor-1. The PP2A-like enzyme required Mn2+ for activity and was inhibited by nanomolar concentrations of okadaic acid (OA). The cDNA sequence of the PP2A-like enzyme was identified through a match of its predicted amino acid sequence with the N-terminal sequence of the catalytic subunit. The PP2B-like (calcineurin) enzyme was stimulated by calmodulin and Ca2+ or Ni2+, but was resistant to OA. Malarial calcineurin was strongly and specifically inhibited by cyclosporin A (CsA) only in the presence of wild type P. falciparum cyclophilin but not a mutant cyclophilin. The inhibition was noncompetitive, and provides a potential explanation for the cyclosporin-sensitivity of the parasite. There was no significant quantitative difference in the total protein Ser/Thr phosphatase activity among the ring, trophozoite, and schizont stages.


Subject(s)
Calcineurin Inhibitors , Cyclosporine/pharmacology , Peptidylprolyl Isomerase/pharmacology , Phosphoric Monoester Hydrolases/metabolism , Plasmodium falciparum/enzymology , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , DNA, Complementary/analysis , Humans , Molecular Sequence Data , Mutation , Okadaic Acid/pharmacology , Peptidylprolyl Isomerase/genetics , Phosphoric Monoester Hydrolases/isolation & purification , Phosphoserine/metabolism , Phosphothreonine/metabolism , Protozoan Proteins/isolation & purification , Sequence Homology, Amino Acid
20.
J Cell Biochem ; 72(2): 251-61, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-10022507

ABSTRACT

Osteoprotegerin (OPG) and its ligand (OPGL) negatively and positively regulate osteoclastogenesis in the mouse. OPG inhibits osteoclastogenesis by sequestering its ligand, OPGL, the osteoclast differentiation and activation factor. This study demonstrates the effects of soluble muOPGL and huOPG on the developing human osteoclast phenotype, on bone slices, using peripheral blood mononuclear cells (PBMCs), cultured for 2 weeks, without stromal cells. OPGL (2-50 ng/ml), in combination with CSF-1, hydrocortisone (HC), and 1,25(OH)2D3, increases the size of osteoclast-like cells on bone, as defined by the acquisition of osteoclast markers: vitronectin receptor (VR), tartrate-resistant acid phosphatase (TRAP), multinuclearity, and bone resorption. By 14 days, with 20 ng/ml OPGL, the largest cells/10x field have achieved an average diameter of 163+/-38 microm, but only approximately 10-20 microm in its absence and the number of osteoclast-like cells/mm2 bone surface is about 128. By scanning electron microscopy, OPGL-treated (20-ng/ml) cultures contain small osteoclast-like cells on bone with ruffled "apical" surfaces by day 7; by day 15, large osteoclast-like cells are spread over resorption lacunae. At 15 ng/ml OPGL, about 37% of the bone slice area is covered by resorption lacunae. OPG (5-250 ng/ml) antagonizes the effects of OPGL on the morphology of the osteoclast-like cells that form, as well as bone erosion. For cells grown on plastic, Cathepsin K mRNA levels, which are barely detectable at plating, are elevated 7-fold, by 5 days, in the presence, not the absence, of OPGL (20 ng/ml) + CSF-1 (25 ng/ml). Similar findings are observed in experiments performed in the absence of HC and 1,25(OH)2D3, indicating that HC and 1,25(OH)2D3 are not needed for OPGL-induced osteoclast differentiation. In conclusion, this study confirms a pivotal role for OPGL and OPG in the modulation of human osteoclast differentiation and function, suggesting a use for OPG for treating osteoclast-mediated bone disease in humans.


Subject(s)
Carrier Proteins , Cytokines/pharmacology , Glycoproteins/pharmacology , Hematopoietic Stem Cells/physiology , Membrane Glycoproteins , Monocytes/physiology , Osteoclasts/drug effects , Receptors, Cytoplasmic and Nuclear , Animals , Bone Development/physiology , Bone and Bones/anatomy & histology , Bone and Bones/physiology , Bone and Bones/ultrastructure , Cathepsin K , Cathepsins/pharmacology , Cell Differentiation , Cytokines/blood , Dose-Response Relationship, Drug , Glycoproteins/blood , Humans , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Osteoprotegerin , Peptidylprolyl Isomerase/pharmacology , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptors, Tumor Necrosis Factor , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...