Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Bone ; 111: 101-108, 2018 06.
Article in English | MEDLINE | ID: mdl-29551750

ABSTRACT

Various substitution mutations in ALK2, a transmembrane serine/threonine kinase receptor for bone morphogenetic proteins (BMPs), have been identified in patients with genetic disorders such as fibrodysplasia ossificans progressiva (FOP), diffuse intrinsic pontine glioma (DIPG) and heart defects. In this study, we characterized the ALK2 mutants R258G, G328V and F246Y, which were identified in patients with severe FOP, DIPG and unusual hereditary skeletal dysplasia, respectively. Both R258G and G328V were gain-of-function mutations, but F246Y was equivalent to wild-type ALK2. We also examined the effect of the suppressor FKBP12 on the signal transduction of a further 14 ALK2 mutations associated with FOP and/or DIPG. To varying extents FKBP12 over-expression suppressed the basal signaling induced by thirteen of the ALK2 mutants, whereas PF197-8L was uniquely resistant. In the PF197-8L mutant, the modelled ALK2 residue L197 induced a steric clash with the D36 residue in FKBP12 and dissociated their interaction. The co-expression of BMP type II receptors or stimulation with ligands relieved the suppression by FKBP12 by disrupting the interaction between mutant ALK2 and FKBP12. Taken together, FKBP12 binds to and suppresses mutant ALK2 proteins associated with FOP and DIPG, except for PF197-8L.


Subject(s)
Activin Receptors, Type I/genetics , Bone Diseases, Developmental/genetics , Bone Morphogenetic Protein Receptors, Type II/physiology , Brain Stem Neoplasms/genetics , Glioma/genetics , Myositis Ossificans/genetics , Tacrolimus Binding Protein 1A/physiology , Animals , Bone Morphogenetic Proteins/metabolism , Cell Line , Humans , Mice , Myositis Ossificans/pathology , Ossification, Heterotopic/genetics , Signal Transduction
2.
Cancer Cell ; 31(3): 424-435, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28292440

ABSTRACT

Although signaling from phosphatidylinositol 3-kinase (PI3K) and AKT to mechanistic target of rapamycin (mTOR) is prominently dysregulated in high-grade glial brain tumors, blockade of PI3K or AKT minimally affects downstream mTOR activity in glioma. Allosteric mTOR inhibitors, such as rapamycin, incompletely block mTORC1 compared with mTOR kinase inhibitors (TORKi). Here, we compared RapaLink-1, a TORKi linked to rapamycin, with earlier-generation mTOR inhibitors. Compared with rapamycin and Rapalink-1, TORKi showed poor durability. RapaLink-1 associated with FKBP12, an abundant mTOR-interacting protein, enabling accumulation of RapaLink-1. RapaLink-1 showed better efficacy than rapamycin or TORKi, potently blocking cancer-derived, activating mutants of mTOR. Our study re-establishes mTOR as a central target in glioma and traces the failure of existing drugs to incomplete/nondurable inhibition of mTORC1.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Multiprotein Complexes/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Female , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred BALB C , Sirolimus/therapeutic use , Tacrolimus Binding Protein 1A/physiology
3.
J Am Soc Nephrol ; 27(5): 1456-64, 2016 05.
Article in English | MEDLINE | ID: mdl-26432904

ABSTRACT

Tacrolimus is a widely used immunosuppressive drug that inhibits the phosphatase calcineurin when bound to the 12 kDa FK506-binding protein (FKBP12). When this binding occurs in T cells, it leads to immunosuppression. Tacrolimus also causes side effects, however, such as hypertension and hyperkalemia. Previously, we reported that tacrolimus stimulates the renal thiazide-sensitive sodium chloride cotransporter (NCC), which is necessary for the development of hypertension. However, it was unclear if tacrolimus-induced hypertension resulted from tacrolimus effects in renal epithelial cells directly or in extrarenal tissues, and whether inhibition of calcineurin was required. To address these questions, we developed a mouse model in which FKBP12 could be deleted along the nephron. FKBP12 disruption alone did not cause phenotypic effects. When treated with tacrolimus, however, BP and the renal abundance of phosphorylated NCC were lower in mice lacking FKBP12 along the nephron than in control mice. Mice lacking FKBP12 along the nephron also maintained a normal relationship between plasma potassium levels and the abundance of phosphorylated NCC with tacrolimus treatment. In cultured cells, tacrolimus inhibited dephosphorylation of NCC. Together, these results suggest that tacrolimus causes hypertension predominantly by inhibiting calcineurin directly in cells expressing NCC, indicating thiazide diuretics may be particularly effective for lowering BP in tacrolimus-treated patients with hypertension.


Subject(s)
Hypertension/chemically induced , Hypertension/prevention & control , Immunosuppressive Agents/adverse effects , Tacrolimus Binding Protein 1A/physiology , Tacrolimus/adverse effects , Animals , Gene Deletion , Kidney , Male , Mice , Tacrolimus Binding Protein 1A/genetics
4.
Yeast ; 31(7): 253-64, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24733494

ABSTRACT

Genetic analysis of protein function requires a rapid means of inactivating the gene under study. Typically, this exploits temperature-sensitive mutations or promoter shut-off techniques. We report the adaptation to Schizosaccharomyces pombe of the anchor-away technique, originally designed in budding yeast by Laemmli lab. This method relies on a rapamycin-mediated interaction between the FRB- and FKBP12-binding domains to relocalize nuclear proteins of interest to the cytoplasm. We demonstrate a rapid nuclear depletion of abundant proteins as proof of principle.


Subject(s)
Minichromosome Maintenance Complex Component 4/physiology , Schizosaccharomyces pombe Proteins/physiology , Schizosaccharomyces/physiology , Sirolimus/pharmacology , Tacrolimus Binding Protein 1A/physiology , Crosses, Genetic , DNA, Fungal/chemistry , DNA, Fungal/genetics , Microscopy, Fluorescence , Plasmids , Polymerase Chain Reaction , Transformation, Genetic
5.
Mol Cell ; 41(2): 173-85, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21255728

ABSTRACT

A cycle of palmitoylation/depalmitoylation of H-Ras mediates bidirectional trafficking between the Golgi apparatus and the plasma membrane, but nothing is known about how this cycle is regulated. We show that the prolyl isomerase (PI) FKBP12 binds to H-Ras in a palmitoylation-dependent fashion and promotes depalmitoylation. A variety of inhibitors of the PI activity of FKBP12, including FK506, rapamycin, and cycloheximide, increase steady-state palmitoylation. FK506 inhibits retrograde trafficking of H-Ras from the plasma membrane to the Golgi in a proline 179-dependent fashion, augments early GTP loading of Ras in response to growth factors, and promotes H-Ras-dependent neurite outgrowth from PC12 cells. These data demonstrate that FKBP12 regulates H-Ras trafficking by promoting depalmitoylation through cis-trans isomerization of a peptidyl-prolyl bond in proximity to the palmitoylated cysteines.


Subject(s)
Proto-Oncogene Proteins p21(ras)/metabolism , Tacrolimus Binding Protein 1A/physiology , Acylation , Animals , Lipoylation , PC12 Cells , Protein Transport , Proto-Oncogene Proteins p21(ras)/chemistry , Rats , Signal Transduction , Tacrolimus Binding Protein 1A/metabolism
6.
Med Sci (Paris) ; 26(12): 1056-60, 2010 Dec.
Article in French | MEDLINE | ID: mdl-21187044

ABSTRACT

The discovery of rapamycin from a soil sample on Easter Island in the mid 60's marked the beginning of an exciting field of research in cell biology and medicine. While it was first used as an antifungal and as an immunosuppressive drug, more recent studies confirmed rapamycin's antiproliferative properties over a variety of solid tumors. Research aimed at identifying its mechanism of action uncovered mTOR (mammalian target of rapamycin), a protein kinase that regulates mRNA translation and protein synthesis, an essential step in cell division and proliferation. Recent evidence suggests a more complex role for mTOR in the regulation of several growth factor-stimulated protein kinases, including the proto-oncogene Akt. This article reviews mTOR function and regulation, and briefly details the future challenges for anti-cancer therapies based on mTOR inhibition.


Subject(s)
TOR Serine-Threonine Kinases/physiology , Animals , Antifungal Agents/pharmacology , Antineoplastic Agents/pharmacology , Autophagy/physiology , Humans , Immunosuppressive Agents/pharmacology , Intercellular Signaling Peptides and Proteins/physiology , Molecular Structure , Multiprotein Complexes/physiology , Phosphorylation , Protein Biosynthesis/physiology , Protein Kinases/physiology , Protein Processing, Post-Translational , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt/physiology , RNA, Messenger/genetics , Ribosomes/physiology , Sirolimus/chemistry , Sirolimus/isolation & purification , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tacrolimus Binding Protein 1A/physiology , Transcription Factors/physiology
7.
FASEB J ; 24(2): 357-63, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19805579

ABSTRACT

FK506 binding protein 12.6 kDa (FKBP12.6), a protein that regulates ryanodine Ca(2+) release channels, may act as an important regulator of insulin secretion. In this study, the role of FKBP12.6 in the control of insulin secretion and blood glucose is clarified using FKBP12.6(-/-) mice. FKBP12.6(-/-) mice showed significant fed hyperinsulinemia but exhibited normoglycemia, fasting normoinsulinemia, and normal body weight compared with wild-type (WT) littermate control mice. Deletion of FKBP12.6 resulted in enhanced glucose-stimulated insulin secretion (GSIS) both in vivo and in vitro, a result that is due to enhanced glucose-induced islet Ca(2+) elevation. After a high-fat dietary challenge (HF diet) for 3 mo, FKBP12.6(-/-) mice displayed higher body weight, hyperinsulinemia, and lower fed blood glucose concentrations compared with WT mice. FKBP12.6(-/-) mice displayed hyperinsulinemia, and resistance to HF diet-induced hyperglycemia, suggesting that FKBP12.6 plays an important role in insulin secretion and blood glucose control, and raising the possibility that it may be a potential therapeutic target for the treatment of type 2 diabetes.


Subject(s)
Blood Glucose/metabolism , Dietary Fats/adverse effects , Hyperglycemia/metabolism , Hyperinsulinism/etiology , Insulin/metabolism , Tacrolimus Binding Protein 1A/deficiency , Animals , Hyperglycemia/prevention & control , Insulin Resistance/physiology , Insulin Secretion , Mice , Mice, Knockout , Tacrolimus Binding Protein 1A/genetics , Tacrolimus Binding Protein 1A/physiology
8.
J Pharmacol Sci ; 109(2): 185-92, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19234362

ABSTRACT

The immunophilin 12-kDa FK506 binding protein (FKBP12) stabilizes intracellular Ca(2+) release channel (CRC) activity in different tissues. In this work, the presence of FKBP12 in rat vas deferens (RVD) and its possible contribution to RVD function was investigated. Treatment under appropriate pH, temperature, and ionic conditions was used to strip FKBP12 from CRC binding sites; Western blotting revealed FKBP12 in control but not in treated homogenates. Disruption of the FKBP12-CRC complex in RVD decreased the Ca(2+) content of sarcoplasmic reticulum (SR) by increasing Ca(2+) leakage through the ryanodine receptor (RyR3 isoform) but not through 1,4,5-inositol trisphosphate receptors (IP(3)R1 and IP(3)R3 isoforms). The decrease of SR Ca(2+) content was not related to inhibition of SERCA ATPase. It seems that dissociation of FKBP12-RyR leads to conformational changes in RyR that make it difficult for ryanodine to access its binding site. Rapamycin, which is commonly used as a pharmacological tool to disrupt the FKBP12-RyR complex, decreased phenylephrine-induced contractions in RVD epididymal halves. The data suggest that FKBP12 is expressed in RVD in a labile association with RyR3. Disruption of the FKBP12-RyR3 complex may lead to modifications of RVD physiology and in consequence may compromise male fertility.


Subject(s)
Sarcoplasmic Reticulum/metabolism , Tacrolimus Binding Protein 1A/physiology , Animals , Calcium/metabolism , Male , Phenylephrine/pharmacology , Rats , Ryanodine Receptor Calcium Release Channel/metabolism , Sirolimus/pharmacology , Tacrolimus Binding Protein 1A/metabolism , Vas Deferens/drug effects
9.
Fungal Genet Biol ; 46(4): 308-20, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19116175

ABSTRACT

The functional characterization of the FKBP12 encoding gene from the phytopathogenic fungus Botrytis cinerea was carried out. B. cinerea genome sequence owns a single ortholog, named BcFKBP12, encoding a FK506-binding protein of 12kDa. BcFKBP12 mediates rapamycin sensitivity both in B. cinerea and in Saccharomyces cerevisiae, a property unique to FKBP12 proteins, probably via the inhibition of the protein kinase TOR (target of rapamycin). The relative abundance of the prolyl isomerase appeared to be regulated and increased in response to the presence of extracellular nutrients. Surprisingly, the BcFKBP12 deletion did not affect the pathogenic development of the strain B05.10, while it was reported to cause a reduction of the virulence of the strain T4. We report for the first time the BcFKBP12 involvement in the sulfur repression of the synthesis of a secreted serine protease. Rapamycin treatment did not relieve the sulfur repression of the reporter system in the wild-type strain. Thus BcFKBP12 may participate in sulfur regulation and its contribution seems to be independent of TOR.


Subject(s)
Botrytis/physiology , Botrytis/pathogenicity , Fungal Proteins/physiology , Gene Expression Regulation, Fungal , Sulfur/metabolism , Tacrolimus Binding Protein 1A/physiology , Antifungal Agents/pharmacology , DNA, Fungal/genetics , Gene Deletion , Microbial Sensitivity Tests , Molecular Sequence Data , Saccharomyces cerevisiae/genetics , Sequence Analysis, DNA , Serine Endopeptidases/biosynthesis , Sirolimus/pharmacology , Tacrolimus Binding Protein 1A/genetics
10.
Biochemistry ; 45(51): 15776-84, 2006 Dec 26.
Article in English | MEDLINE | ID: mdl-17176100

ABSTRACT

Multiple intracellular receptors of the FK506 binding protein (FKBP) family of peptidylprolyl cis/trans-isomerases are potential targets for the immunosuppressive drug FK506. Inhibition of the protein phosphatase calcineurin (CaN), which has been implicated in the FK506-mediated blockade of T cell proliferation, was shown to involve a gain of function in the FKBP12/FK506 complex. We studied the potential of six human FKBPs to contribute to CaN inhibition by comparative examination of inhibition constants of the respective FK506/FKBP complexes. Interestingly, these FKBPs form tight complexes with FK506, exhibiting comparable dissociation constants, but the resulting FK506/FKBP complexes differ greatly in their affinity for CaN, with IC50 values in the range of 0.047-17 microM. The different capacities of FK506/FKBP complexes to affect CaN activity are partially caused by substitutions corresponding to the amino acid side chains K34 and I90 of FKBP12. Only the FK506 complexes of FKBP12, FKBP12.6, and FKBP51 showed high affinity to CaN; small interfering RNA against these FKBP allowed defining the contribution of individual FKBP in an NFAT reporter gene assay. Our results allow quantitative correlation between FK506-mediated CaN effects and the abundance of the different FKBPs in the cell.


Subject(s)
Calcineurin Inhibitors , Enzyme Inhibitors/metabolism , Immunosuppressive Agents/pharmacology , Multienzyme Complexes/metabolism , Tacrolimus Binding Proteins/physiology , Amino Acid Sequence , Animals , Calcineurin/metabolism , Calmodulin/physiology , Cattle , Enzyme Inhibitors/pharmacology , Humans , Immunosuppressive Agents/metabolism , Jurkat Cells , Molecular Sequence Data , Multienzyme Complexes/physiology , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Sirolimus/metabolism , Sirolimus/pharmacology , Substrate Specificity , Tacrolimus/metabolism , Tacrolimus/pharmacology , Tacrolimus Binding Protein 1A/physiology , Tacrolimus Binding Proteins/metabolism
11.
Handb Exp Pharmacol ; (172): 359-404, 2006.
Article in English | MEDLINE | ID: mdl-16610367

ABSTRACT

Peptide bond isomerases are involved in important physiological processes that can be targeted in order to treat neurodegenerative disease, cancer, diseases of the immune system, allergies, and many others. The folding helper enzyme class of Peptidyl-Prolyl-cis/trans Isomerases (PPIases) contains the three enzyme families of cyclophilins (Cyps), FK506 binding proteins (FKBPs), and parvulins (Pars). Although they are structurally unrelated, all PPIases catalyze the cis/trans isomerization of the peptide bond preceding the proline in a polypeptide chain. This process not only plays an important role in de novo protein folding, but also in isomerization of native proteins. The native state isomerization plays a role in physiological processes by influencing receptor ligand recognition or isomer-specific enzyme reaction or by regulating protein function by catalyzing the switch between native isomers differing in their activity, e.g., ion channel regulation. Therefore elucidating PPIase involvement in physiological processes and development of specific inhibitors will be a suitable attempt to design therapies for fatal and deadly diseases.


Subject(s)
Cyclophilins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Peptidylprolyl Isomerase/antagonists & inhibitors , Protein Folding , Tacrolimus Binding Protein 1A/antagonists & inhibitors , Animals , Cardiovascular Diseases/drug therapy , Catalysis , Cyclophilins/physiology , Cyclosporine/pharmacology , Drug Design , Enzyme Inhibitors/therapeutic use , Humans , Immunosuppressive Agents/pharmacology , Inflammation/drug therapy , NIMA-Interacting Peptidylprolyl Isomerase , Neoplasms/drug therapy , Peptidylprolyl Isomerase/physiology , Reperfusion Injury/drug therapy , Skin Diseases/drug therapy , Tacrolimus Binding Protein 1A/physiology
12.
J Am Chem Soc ; 128(17): 5718-27, 2006 May 03.
Article in English | MEDLINE | ID: mdl-16637639

ABSTRACT

Transverse relaxation dispersion NMR spectroscopy can provide atom-specific information about time scales, populations, and the extent of structural reorganization in proteins under equilibrium conditions. A method is described that uses side-chain methyl groups as local reporters for conformational transitions taking place in the microsecond regime. The experiment measures carbon nuclear spin relaxation rates in the presence of continuous wave off-resonance irradiation, in proteins uniformly enriched with 13C, and partially randomly labeled with 2H. The method was applied to human FK-506 binding protein (FKBP12), which uses a common surface for binding substrates in its dual role as both an immunophilin and folding assistant. Conformational dynamics on a time scale of approximately 130 micros were detected for methyl groups located in the substrate binding pocket, demonstrating its plasticity in the absence of substrate. The spatial arrangement of affected side-chain atoms suggests that substrate recognition involves the rapid relative movement of the subdomain comprising residues Ala81-Thr96 and that the observed dynamics play an important role in facilitating the interaction of this protein with its many partners, including calcineurin.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular/methods , Tacrolimus Binding Protein 1A/physiology , Carbon Isotopes , Humans , Tacrolimus Binding Protein 1A/chemistry , Thermodynamics
13.
J Mol Biol ; 358(2): 372-86, 2006 Apr 28.
Article in English | MEDLINE | ID: mdl-16497329

ABSTRACT

The ascomycete Botrytis cinerea is a broad-spectrum plant pathogen. Here, we describe the first macroarray transcriptomic study of the fungus in real-time infection conditions. Infection of Arabidopsis thaliana leaves by B.cinerea was monitored using macroarrays, containing 3032 genes. Variance analysis revealed that 7% of B.cinerea genes are differentially expressed during infection and allowed us to identify 27 genes significantly up-regulated in planta. Among them, two genes have already been associated with fungal pathogenicity, while eight genes have unidentified functions. The 27 genes were separated into three groups according to their expression profile. The first group showed maximal expression at the early stage following fungal penetration, the second one showed maximal expression at the outset of the colonization of plant leaves and the third group showed maximal expression when the colonization of plant leaves was completed. A gene of the last group (BcPIC5), which is homologous to FKBP12 proteins, was disrupted in order to determine its role in pathogenicity. At seven days post-inoculation, the lesions caused by the DeltaBcPIC5 mutant on bean leaves were reduced by 69% and did not further expand compared to the wild-type. These results confirm that transcriptomic analysis under infection conditions can be very valuable for the identification of fungal genes related to pathogenicity.


Subject(s)
Arabidopsis/microbiology , Botrytis/genetics , Gene Expression Profiling , Gene Expression Regulation, Fungal/genetics , Genes, Fungal/physiology , Plant Leaves/microbiology , Tacrolimus Binding Protein 1A/physiology , Arabidopsis/genetics , Arabidopsis/metabolism , Botrytis/pathogenicity , Gene Targeting , Molecular Sequence Data , Mutation , Phylogeny , Plant Leaves/genetics , Plant Leaves/metabolism , Tacrolimus Binding Protein 1A/genetics , Transcription, Genetic , Up-Regulation
14.
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
15.
J Biol Chem ; 281(10): 6349-57, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16380387

ABSTRACT

The protein phosphatase 2A (PP2A) phosphatase activator (PTPA) is an essential protein involved in the regulation of PP2A and the PP2A-like enzymes. In this study we demonstrate that PTPA and its yeast homologues Ypa1 and Ypa2 can induce a conformational change in some model substrates. Using these model substrates in different assays with and without helper proteases, this isomerase activity is similar to the isomerase activity of FKBP12, the human cyclophilin A, and one of its yeast homologs Cpr7 but dissimilar to the isomerase activity of Pin1. However, neither FKBP12 nor Cpr7 can reactivate the inactive form of PP2A. Therefore, PTPA belongs to a novel peptidyl-prolyl cis/trans-isomerase (PPIase) family. The PPIase activity of PTPA correlates with its activating activity since both are stimulated by the presence of Mg2+ATP, and a PTPA mutant (Delta208-213) with 400-fold less activity in the activation reaction of PP2A also showed almost no PPIase activity. The point mutant Asp205 --> Gly (in Ypa1) identified this amino acid as essential for both activities. Moreover, PTPA dissociates the inactive form from the complex with the PP2A methylesterase. Finally, Pro190 in the catalytic subunit of PP2A (PP2AC) could be identified as the target Pro isomerized by PTPA/Mg2+ATP since among the 14 Pro residues present in 12 synthesized peptides representing the microenvironments of these prolines in PP2AC, only Pro190 could be isomerized by PTPA/Mg2+ATP. This Pro190 is present in a predicted loop structure near the catalytic center of PP2AC and, if mutated into a Phe, the phosphatase is inactive and can no longer be activated by PTPA/Mg2+ATP.


Subject(s)
Peptidylprolyl Isomerase/physiology , Phosphoprotein Phosphatases/metabolism , Proteins/physiology , Adenosine Triphosphate/physiology , Animals , COS Cells , Catalytic Domain , Chlorocebus aethiops , Cyclophilin A/genetics , Cyclophilin A/physiology , Peptidyl-Prolyl Isomerase F , Cyclophilins/genetics , Cyclophilins/physiology , Humans , Kinetics , Magnesium/physiology , Multigene Family , Mutagenesis, Site-Directed , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Proline/genetics , Proline/metabolism , Protein Phosphatase 2 , Proteins/genetics , Rabbits , Substrate Specificity , Tacrolimus Binding Protein 1A/genetics , Tacrolimus Binding Protein 1A/physiology
17.
Am J Physiol Cell Physiol ; 289(6): C1476-84, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16049053

ABSTRACT

Ca+-induced Ca2+ release (CICR) in the heart involves local Ca2+ signaling between sarcolemmal L-type Ca2+ channels (dihydropyridine receptors, DHPRs) and type 2 ryanodine receptors (RyR2s) in the sarcoplasmic reticulum (SR). We reconstituted cardiac-like CICR by expressing a cardiac dihydropyridine-insensitive (T1066Y/Q1070M) alpha1-subunit (alpha1CYM) and RyR2 in myotubes derived from RyR1-knockout (dyspedic) mice. Myotubes expressing alpha1CYM and RyR2 were vesiculated and exhibited spontaneous Ca2+ oscillations that resulted in chaotic and uncontrolled contractions. Coexpression of FKBP12.6 (but not FKBP12.0) with alpha1CYM and RyR2 eliminated vesiculations and reduced the percentage of myotubes exhibiting uncontrolled global Ca2+ oscillations (63% and 13% of cells exhibited oscillations in the absence and presence of FKBP12.6, respectively). alpha1CYM/RyR2/FKBP12.6-expressing myotubes exhibited robust and rapid electrically evoked Ca2+ transients that required extracellular Ca2+. Depolarization-induced Ca2+ release in alpha1CYM/RyR2/FKBP12.6-expressing myotubes exhibited a bell-shaped voltage dependence that was fourfold larger than that of myotubes expressing alpha1CYM alone (maximal fluorescence change was 2.10 +/- 0.39 and 0.54 +/- 0.07, respectively), despite similar Ca2+ current densities. In addition, the gain of CICR in alpha1CYM/RyR2/FKBP12.6-expressing myotubes exhibited a nonlinear voltage dependence, being considerably larger at threshold potentials. We used this molecular model of local alpha1C-RyR2 signaling to assess the ability of FKBP12.6 to inhibit spontaneous Ca2+ release via a phosphomimetic mutation in RyR2 (S2808D). Electrically evoked Ca2+ release and the incidence of spontaneous Ca2+ oscillations did not differ in wild-type RyR2- and S2808D-expressing myotubes over a wide range of FKBP12.6 expression. Thus a negative charge at S2808 does not alter in situ regulation of RyR2 by FKBP12.6.


Subject(s)
Calcium Channels, L-Type/physiology , Calcium Signaling , Muscle Fibers, Skeletal/physiology , Myocardium/metabolism , Ryanodine Receptor Calcium Release Channel/physiology , Tacrolimus Binding Proteins/physiology , Animals , Animals, Newborn , Calcium Channels, L-Type/genetics , Cells, Cultured , Electric Stimulation , Mice , Mice, Knockout , Muscle Contraction , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Mutation , Phosphorylation , Protein Subunits/genetics , Protein Subunits/physiology , Ryanodine Receptor Calcium Release Channel/genetics , Tacrolimus Binding Protein 1A/physiology , Tacrolimus Binding Proteins/genetics
18.
Bone ; 36(2): 243-53, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15780950

ABSTRACT

FK506 is a commonly used immunosuppressant that mediates its action by exclusively interacting with the cytosolic immunophilin, FK506 binding protein 12 (FKBP12). Although FK506-induced acute osteoporosis is now well recognised, its precise mode of action in osteoblasts remains unclear. Therefore, in the present study we characterised FKBP12 in osteoblasts and investigated the role of FK506 in modulating osteoblast-specific transcription factors, core-binding factor alpha1 (Cbfa1) and osterix gene expression in ROS 17/2.8 cells. RT-PCR, immunolocalisation and Western blotting studies were employed to identify and characterise FKBP12 in rat primary osteoblasts and osteoblast-like osteosarcoma ROS 17/2.8 cells. Western blotting extracts of these cells revealed the 12 kDa and hitherto unreported 10 kDa FKBP isoform that were immunolocalised predominantly to the cytosol. The transient exposure of ROS 17/2.8 cells to H2O2 (100 microM) was found to elevate FKBP12 mRNA after 10 min and protein expression after 24 h. Both PTH (10(-9) M) and 1,25 (OH)2D3 (Vitamin D3) (10(-7) M) suppressed FKBP12 protein expression. FK506 in the therapeutic range (25 nmol/L) suppressed expression of Cbfa1 and osterix mRNA. The inhibition of Cbfa1 isoforms II/III expression was evident at 30 min and the extent of inhibition was sustained at 6 h. Osterix inhibition was also seen after 30 min, however, it became maximal after 6 h. The dose-dependant inhibition of osterix in these cells, carried out using 1.25, 12.5 and 125 nmol/L of FK506 was maximal at 1.25 nmol/L. Cbfa1 isoforms II/III were also maximally inhibited at 1.25 nmol/L; interestingly, the inhibition became less marked at higher concentrations of FK506. Similar dose of FK506 was found to inhibit ROS 17/2.8 cell proliferation; the inhibitory effect however was greater in insulin-stimulated cells. The results of this study suggest that immunosuppressant-induced osteoporosis, which is known to involve accelerated bone resorption by increase in osteoclastogenesis, may in fact also be accentuated by the inhibition of osteoblast differentiation and function.


Subject(s)
Neoplasm Proteins/biosynthesis , Tacrolimus Binding Protein 1A/physiology , Transcription Factors/biosynthesis , Alkaline Phosphatase/metabolism , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Core Binding Factor Alpha 1 Subunit , Core Binding Factors , Cytosol/enzymology , Cytosol/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Molecular Sequence Data , Neoplasm Proteins/genetics , Protein Isoforms/physiology , RNA, Messenger/biosynthesis , Rats , Tacrolimus/metabolism , Tacrolimus/pharmacology , Tacrolimus Binding Protein 1A/biosynthesis , Tacrolimus Binding Protein 1A/metabolism , Transcription Factors/genetics
19.
J Physiol ; 556(Pt 3): 919-34, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14966299

ABSTRACT

This study investigated the function of FK506-binding protein (FKBP12.6) using adenoviral-mediated gene transfer to over-express FKBP12.6 (Ad-FKBP12.6) in adult rabbit ventricular cardiomyocytes. Infection with a beta-galactosidase-expressing adenovirus (Ad-LacZ) was used as a control. Peak-systolic intracellular [Ca(2+)] (measured with Fura-2) was higher in the Ad-FKBP12.6 group compared to Ad-LacZ (1 Hz field stimulation at 37 degrees C). The amplitude of caffeine-induced Ca(2+) release was also greater, indicating a higher SR Ca(2+) content in the Ad-FKBP12.6 group. Voltage clamp experiments indicated that FKBP12.6 over-expression did not change L-type Ca(2+) current amplitude or Ca(2+) efflux rates via the Na(+)-Ca(2+) exchanger. Ca(2+) transients comparable to those after Ad-FKBP12.6 transfection could be obtained by enhancing SR Ca(2+) content of Ad-LacZ infected cells with periods of high frequency stimulation. Line-scan confocal microscopy (Fluo-3 fluorescence) of intact cardiomyocytes stimulated at 0.5 Hz (20-21 degrees C) revealed a higher degree of synchronicity of SR Ca(2+) release and fewer non-responsive Ca(2+) release sites in the Ad-FKBP12.6 group compared to control. Ca(2+) spark morphology was measured in beta-escin-permeabilized cardiomyocytes at a free [Ca(2+)](i) of 150 nm. The average values of the spark parameters (amplitude, duration, width and frequency) were reduced in the Ad-FKBP12.6 group. Increasing [Ca(2+)](i) to 400 nm caused coherent propagating Ca(2+) waves in the Ad-FKBP12.6 group but only limited Ca(2+) release events were recorded in the control group. These data indicate that FKBP12.6 over-expression enhances Ca(2+) transient amplitude predominately by increasing SR Ca(2+) content. Moreover, there is also evidence that FKBP12.6 can enhance the coupling between SR Ca(2+) release sites independently of SR content.


Subject(s)
Calcium Signaling/physiology , Myocytes, Cardiac/physiology , Tacrolimus Binding Protein 1A/physiology , Adenoviridae/genetics , Animals , Caffeine/pharmacology , Calcium/metabolism , Calcium/pharmacology , Calcium Signaling/drug effects , Cell Membrane Permeability/drug effects , Cell Shape/drug effects , Cells, Cultured , Electric Stimulation , Escin/pharmacology , Fura-2/chemistry , Gene Expression , Genetic Vectors/genetics , Humans , Membrane Potentials/physiology , Microscopy, Confocal , Microscopy, Fluorescence , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Rabbits , Sarcoplasmic Reticulum/chemistry , Sarcoplasmic Reticulum/metabolism , Spectrometry, Fluorescence , Tacrolimus Binding Protein 1A/genetics , Tetrodotoxin/pharmacology , Thapsigargin/pharmacology , Transfection
20.
Front Biosci ; 9: 619-31, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14766396

ABSTRACT

FKBP12 as an immunophilin that binds to two well-known immunosuppressive macrolides, FK506 and rapamycin, has attracted immense attention and its role in mediating the immunosuppressive functions of these macrolides has been extensively studied. Since FKBP12 is a well-conserved protein among many species and is also highly expressed in almost all cells, it must play important roles in cellular function in the absence of macrolides. In one such a role, FKBP12 interacts with and regulates the functional state of the ryanodine Ca2+ channel receptor by altering protein conformation and coordinating multi-protein complex formation. This review summarizes another physiological role of FKBP12 as an interactor and a regulator of the type I serine/threonine kinase receptors of TGF-beta superfamily. Current data, derived from detailed biochemical studies as well as from functional studies in various systems, suggest that FKBP12 functions as a "guardian" for the type I receptors to prevent them from leaky signaling under sub-optimal ligand concentrations, thereby providing a molecular "gradient reader" for TGF-beta family morphogens. This aspect of FKBP12 function may be critical for cellular responsiveness to morphogenetic gradients of the TGF-beta family members during early development, serving to assure the translation of different ligand concentrations into different signaling readouts.


Subject(s)
Receptors, Transforming Growth Factor beta/metabolism , Tacrolimus Binding Protein 1A/metabolism , Animals , Calcium Channels/chemistry , Forecasting , Humans , Immunophilins/metabolism , Macrolides/pharmacology , Protein Structure, Tertiary/physiology , Tacrolimus Binding Protein 1A/drug effects , Tacrolimus Binding Protein 1A/physiology , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...