93 results on '"Honkanen RE"'
Search Results
2. Polyamines Regulate Serine/Threonine Protein Phosphatases in Insulin-Secreting Cells
- Author
-
Honkanen Re and Sjöholm A
- Subjects
Eflornithine ,Spermidine ,Endocrinology, Diabetes and Metabolism ,Decarboxylase inhibitor ,Spermine ,Protein Serine-Threonine Kinases ,Biology ,Arginine ,Histones ,Islets of Langerhans ,chemistry.chemical_compound ,Endocrinology ,Insulin Secretion ,Okadaic Acid ,Phosphoprotein Phosphatases ,Polyamines ,Putrescine ,Tumor Cells, Cultured ,Internal Medicine ,Humans ,Insulin ,Protein phosphorylation ,Enzyme Inhibitors ,Phosphorylation ,Hepatology ,Kinase ,Enzyme Activation ,Pancreatic Neoplasms ,chemistry ,Biochemistry ,Insulinoma ,Polyamine ,Protein Processing, Post-Translational - Abstract
Reversible protein phosphorylation is an important mechanism by which cells transduce external signals into biologic responses. Levels of protein phosphorylation are determined by the balanced actions of both protein kinases and protein phosphatases (PPases). However, compared with protein kinases, regulation of PPases has been relatively neglected. The insulin secretagogue L-arginine, an immediate metabolic precursor to polyamines, causes a rapid and transient decrease in PPase-1 activity in insulin-secreting RINm5F cells. We here show that polyamines dose-dependently suppress PPase-1-like activity when added to RINm5F cell homogenates at physiologic concentrations (spermine > spermidine > putrescine), while having minor and inconsistent effects on PPase-2A-like activity. The IC50 value for spermine on PPase-1-like activity was approximately 4 mM. The inhibitory effect was reproduced and of comparable magnitude on purified PPases types 1 and 2A. On the other hand, when endogenous polyamine pools were exhausted by 4 days of exposure to the specific L-ornithine decarboxylase inhibitor DL-alpha-difluoromethylornithine, there was an increase in PPase-2A-like activity. Quantitative Western analysis revealed that the amount of PPase-2A protein did not change after this treatment. It is concluded that polyamines cause time-and concentration-dependent inhibitory effects on RINm5F cell PPase activities, which may contribute to the increase in phosphorylation state that occurs after secretory stimulation.
- Published
- 2000
3. B56δ long-disordered arms form a dynamic PP2A regulation interface coupled with global allostery and Jordan's syndrome mutations.
- Author
-
Wu CG, Balakrishnan VK, Merrill RA, Parihar PS, Konovolov K, Chen YC, Xu Z, Wei H, Sundaresan R, Cui Q, Wadzinski BE, Swingle MR, Musiyenko A, Chung WK, Honkanen RE, Suzuki A, Huang X, Strack S, and Xing Y
- Subjects
- Jordan, Phosphorylation, Mutation, Holoenzymes genetics, Holoenzymes metabolism, Protein Phosphatase 2 metabolism
- Abstract
Intrinsically disordered regions (IDR) and short linear motifs (SLiMs) play pivotal roles in the intricate signaling networks governed by phosphatases and kinases. B56δ (encoded by PPP2R5D ) is a regulatory subunit of protein phosphatase 2A (PP2A) with long IDRs that harbor a substrate-mimicking SLiM and multiple phosphorylation sites. De novo missense mutations in PPP2R5D cause intellectual disabilities (ID), macrocephaly, Parkinsonism, and a broad range of neurological symptoms. Our single-particle cryo-EM structures of the PP2A-B56δ holoenzyme reveal that the long, disordered arms at the B56δ termini fold against each other and the holoenzyme core. This architecture suppresses both the phosphatase active site and the substrate-binding protein groove, thereby stabilizing the enzyme in a closed latent form with dual autoinhibition. The resulting interface spans over 190 Å and harbors unfavorable contacts, activation phosphorylation sites, and nearly all residues with ID-associated mutations. Our studies suggest that this dynamic interface is coupled to an allosteric network responsive to phosphorylation and altered globally by mutations. Furthermore, we found that ID mutations increase the holoenzyme activity and perturb the phosphorylation rates, and the severe variants significantly increase the mitotic duration and error rates compared to the normal variant., Competing Interests: Competing interests statement:The authors declare no competing interest.
- Published
- 2024
- Full Text
- View/download PDF
4. Quantitative proteomics and phosphoproteomics of PP2A-PPP2R5D variants reveal deregulation of RPS6 phosphorylation via converging signaling cascades.
- Author
-
Smolen KA, Papke CM, Swingle MR, Musiyenko A, Li C, Salter EA, Camp AD, Honkanen RE, and Kettenbach AN
- Subjects
- Humans, Autism Spectrum Disorder, HEK293 Cells, Mechanistic Target of Rapamycin Complex 1 genetics, Mechanistic Target of Rapamycin Complex 1 metabolism, Phosphorylation, Protein Phosphatase 2 genetics, Protein Phosphatase 2 metabolism, Proteomics, Ribosomal Protein S6 genetics, Ribosomal Protein S6 metabolism, Abnormalities, Multiple metabolism, Abnormalities, Multiple pathology
- Abstract
Genetic germline variants of PPP2R5D (encoding: phosphoprotein phosphatase 2 regulatory protein 5D) result in PPP2R5D-related disorder (Jordan's Syndrome), which is characterized by intellectual disability, hypotonia, seizures, macrocephaly, autism spectrum disorder, and delayed motor skill development. The disorder originates from de novo single nucleotide mutations, generating missense variants that act in a dominant manner. Pathogenic mutations altering 13 different amino acids have been identified, with the E198K variant accounting for ∼40% of reported cases. However, the generation of a heterozygous E198K variant cell line to study the molecular effects of the pathogenic mutation has been challenging. Here, we use CRISPR-PRIME genomic editing to introduce a transition (c.592G>A) in a single PPP2R5D allele in HEK293 cells, generating E198K-heterozygous lines to complement existing E420K variant lines. We generate global protein and phosphorylation profiles of WT, E198K, and E420K cell lines and find unique and shared changes between variants and WT cells in kinase- and phosphatase-controlled signaling cascades. We observed ribosomal protein S6 (RPS6) hyperphosphorylation as a shared signaling alteration, indicative of increased ribosomal protein S6-kinase activity. Treatment with rapamycin or an RPS6-kinase inhibitor (LY2584702) suppressed RPS6 phosphorylation in both, suggesting upstream activation of mTORC1/p70S6K. Intriguingly, our data suggests ERK-dependent activation of mTORC1 in both E198K and E420K variant cells, with additional AKT-mediated mTORC1 activation in the E420K variant. Thus, although upstream activation of mTORC1 differs between PPP2R5D-related disorder genotypes, inhibition of mTORC1 or RPS6 kinases warrants further investigation as potential therapeutic strategies for patients., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
5. Editorial: Deciphering the etiology of rare genetic disorders associated with protein phosphatases.
- Author
-
Janssens V, Wadzinski BE, Li C, and Honkanen RE
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
6. Quantum-based modeling implies that bidentate Arg 89 -substrate binding enhances serine/threonine protein phosphatase-2A(PPP2R5D/PPP2R1A/PPP2CA)-mediated dephosphorylation.
- Author
-
Salter EA, Wierzbicki A, Honkanen RE, and Swingle MR
- Abstract
PP2A-serine/threonine protein phosphatases function as heterotrimeric holoenzymes, composed of a common scaffold (A-subunit encoded by PPP2R1A/PPP2R1B), a common catalytic (C-subunit encoded by PPP2CA/PPP2CB), and one of many variable regulatory (B) subunits. The site of phosphoprotein phosphatase (PPP) hydrolysis features a bimetal system (M
1 /M2 ), an associated bridge hydroxide [W1 (OH- )], and a highly-conserved core sequence. In the presumptive common mechanism, the phosphoprotein's seryl/threonyl phosphate coordinates the M1 /M2 system, W1 (OH- ) attacks the central P atom, rupturing the antipodal bond, and simultaneously, a histidine/aspartate tandem protonates the exiting seryl/threonyl alkoxide. Based on studies of PPP5C, a conserved arginine proximal to M1 is also expected to bind the substrate's phosphate group in a bidentate fashion. However, in PP2A isozymes, the role of the arginine (Arg89 ) in hydrolysis is not clear because two independent structures for PP2A(PPP2R5C) and PP2A(PPP2R5D) show that Arg89 engages in a weak salt bridge at the B:C interface. These observations raise the question of whether hydrolysis proceeds with or without direct involvement of Arg89 . The interaction of Arg89 with B:Glu198 in PP2A(PPP2R5D) is significant because the pathogenic E198K variant of B56δ is associated with irregular protein phosphorylation levels and consequent developmental disorders (Jordan's Syndrome; OMIM #616355). In this study, we perform quantum-based hybrid [ONIOM(UB3LYP/6-31G(d):UPM7)] calculations on 39-residue models of the PP2A(PPP2R5D)/pSer (phosphoserine) system to estimate activation barriers for hydrolysis in the presence of bidentate Arg89 -substrate binding and when Arg89 is otherwise engaged in the salt-bridge interaction. Our solvation-corrected results yield ΔH‡ ≈ ΔE‡ = +15.5 kcal/mol for the former case, versus +18.8 kcal/mol for the latter, indicating that bidentate Arg89 -substrate binding is critical for optimal catalytic function of the enzyme. We speculate that PP2A(PPP2R5D) activity is suppressed by B:Glu198 sequestration of C:Arg89 under native conditions, whereas the PP2A(PPP2R5D)-holoenzyme containing the E198K variant has a positively-charged lysine in this position that alters normal function., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Salter, Wierzbicki, Honkanen and Swingle.)- Published
- 2023
- Full Text
- View/download PDF
7. FAK Activation Promotes SMC Dedifferentiation via Increased DNA Methylation in Contractile Genes.
- Author
-
Jeong K, Murphy JM, Kim JH, Campbell PM, Park H, Rodriguez YAR, Choi CS, Kim JS, Park S, Kim HJ, Scammell JG, Weber DS, Honkanen RE, Schlaepfer DD, Ahn EE, and Lim SS
- Subjects
- Animals, Cells, Cultured, Contractile Proteins metabolism, DNA Methyltransferase 3A genetics, DNA Methyltransferase 3A metabolism, Focal Adhesion Kinase 1 genetics, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Mice, Mice, Inbred C57BL, Muscle, Smooth, Vascular cytology, Myocytes, Smooth Muscle cytology, Myocytes, Smooth Muscle physiology, Proteolysis, Ubiquitination, Up-Regulation, Cell Dedifferentiation, Contractile Proteins genetics, DNA Methylation, Focal Adhesion Kinase 1 metabolism, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle metabolism
- Abstract
Rationale: Vascular smooth muscle cells (SMCs) exhibit remarkable plasticity and can undergo dedifferentiation upon pathological stimuli associated with disease and interventions., Objective: Although epigenetic changes are critical in SMC phenotype switching, a fundamental regulator that governs the epigenetic machineries regulating the fate of SMC phenotype has not been elucidated., Methods and Results: Using SMCs, mouse models, and human atherosclerosis specimens, we found that FAK (focal adhesion kinase) activation elicits SMC dedifferentiation by stabilizing DNMT3A (DNA methyltransferase 3A). FAK in SMCs is activated in the cytoplasm upon serum stimulation in vitro or vessel injury and active FAK prevents DNMT3A from nuclear FAK-mediated degradation. However, pharmacological or genetic FAK catalytic inhibition forced FAK nuclear localization, which reduced DNMT3A protein via enhanced ubiquitination and proteasomal degradation. Reduced DNMT3A protein led to DNA hypomethylation in contractile gene promoters, which increased SMC contractile protein expression. RNA-sequencing identified SMC contractile genes as a foremost upregulated group by FAK inhibition from injured femoral artery samples compared with vehicle group. DNMT3A knockdown in injured arteries reduced DNA methylation and enhanced contractile gene expression supports the notion that nuclear FAK-mediated DNMT3A degradation via E3 ligase TRAF6 (TNF [tumor necrosis factor] receptor-associated factor 6) drives differentiation of SMCs. Furthermore, we observed that SMCs of human atherosclerotic lesions exhibited decreased nuclear FAK, which was associated with increased DNMT3A levels and decreased contractile gene expression., Conclusions: This study reveals that nuclear FAK induced by FAK catalytic inhibition specifically suppresses DNMT3A expression in injured vessels resulting in maintaining SMC differentiation by promoting the contractile gene expression. Thus, FAK inhibitors may provide a new treatment option to block SMC phenotypic switching during vascular remodeling and atherosclerosis.
- Published
- 2021
- Full Text
- View/download PDF
8. Phosphatase inhibition by LB-100 enhances BMN-111 stimulation of bone growth.
- Author
-
Shuhaibar LC, Kaci N, Egbert JR, Horville T, Loisay L, Vigone G, Uliasz TF, Dambroise E, Swingle MR, Honkanen RE, Biosse Duplan M, Jaffe LA, and Legeai-Mallet L
- Subjects
- Animals, Bone Diseases, Developmental genetics, Cartilage drug effects, Cartilage growth & development, Cell Differentiation drug effects, Chondrocytes drug effects, Drug Synergism, Growth Plate drug effects, Growth Plate growth & development, Mice, Natriuretic Peptide, C-Type pharmacology, Organ Size, Phosphorylation, Primary Cell Culture, Receptors, Atrial Natriuretic Factor genetics, Tibia drug effects, Tibia growth & development, Achondroplasia genetics, Bone Development drug effects, Enzyme Inhibitors pharmacology, Natriuretic Peptide, C-Type analogs & derivatives, Phosphoric Monoester Hydrolases antagonists & inhibitors, Piperazines pharmacology, Receptor, Fibroblast Growth Factor, Type 3 genetics, Receptors, Atrial Natriuretic Factor agonists
- Abstract
Activating mutations in fibroblast growth factor receptor 3 (FGFR3) and inactivating mutations in the natriuretic peptide receptor 2 (NPR2) guanylyl cyclase both result in decreased production of cyclic GMP in chondrocytes and severe short stature, causing achondroplasia (ACH) and acromesomelic dysplasia, type Maroteaux, respectively. Previously, we showed that an NPR2 agonist BMN-111 (vosoritide) increases bone growth in mice mimicking ACH (Fgfr3Y367C/+). Here, because FGFR3 signaling decreases NPR2 activity by dephosphorylating the NPR2 protein, we tested whether a phosphatase inhibitor (LB-100) could enhance BMN-111-stimulated bone growth in ACH. Measurements of cGMP production in chondrocytes of living tibias, and of NPR2 phosphorylation in primary chondrocytes, showed that LB-100 counteracted FGF-induced dephosphorylation and inactivation of NPR2. In ex vivo experiments with Fgfr3Y367C/+ mice, the combination of BMN-111 and LB-100 increased bone length and cartilage area, restored chondrocyte terminal differentiation, and increased the proliferative growth plate area, more than BMN-111 alone. The combination treatment also reduced the abnormal elevation of MAP kinase activity in the growth plate of Fgfr3Y367C/+ mice and improved the skull base anomalies. Our results provide a proof of concept that a phosphatase inhibitor could be used together with an NPR2 agonist to enhance cGMP production as a therapy for ACH.
- Published
- 2021
- Full Text
- View/download PDF
9. A disorder-related variant (E420K) of a PP2A-regulatory subunit (PPP2R5D) causes constitutively active AKT-mTOR signaling and uncoordinated cell growth.
- Author
-
Papke CM, Smolen KA, Swingle MR, Cressey L, Heng RA, Toporsian M, Deng L, Hagen J, Shen Y, Chung WK, Kettenbach AN, and Honkanen RE
- Subjects
- Autistic Disorder genetics, Autistic Disorder pathology, CRISPR-Cas Systems genetics, Genetic Diseases, Inborn pathology, Humans, Intellectual Disability genetics, Intellectual Disability pathology, Megalencephaly genetics, Megalencephaly pathology, Mutation genetics, Polymorphism, Single Nucleotide genetics, Proto-Oncogene Proteins c-akt genetics, Genetic Diseases, Inborn genetics, Genetic Predisposition to Disease, Protein Phosphatase 2 genetics, Proteomics, TOR Serine-Threonine Kinases genetics
- Abstract
Functional genomic approaches have facilitated the discovery of rare genetic disorders and improved efforts to decipher their underlying etiology. PPP2R5D-related disorder is an early childhood onset condition characterized by intellectual disability, hypotonia, autism-spectrum disorder, macrocephaly, and dysmorphic features. The disorder is caused by de novo single nucleotide changes in PPP2R5D, which generate heterozygous dominant missense variants. PPP2R5D is known to encode a B'-type (B'56δ) regulatory subunit of a PP2A-serine/threonine phosphatase. To help elucidate the molecular mechanisms altered in PPP2R5D-related disorder, we used a CRISPR-single-base editor to generate HEK-293 cells in which a single transition (c.1258G>A) was introduced into one allele, precisely recapitulating a clinically relevant E420K variant. Unbiased quantitative proteomic and phosphoproteomic analyses of endogenously expressed proteins revealed heterozygous-dominant changes in kinase/phosphatase signaling. These data combined with orthogonal validation studies revealed a previously unrecognized interaction of PPP2R5D with AKT in human cells, leading to constitutively active AKT-mTOR signaling, increased cell size, and uncoordinated cellular growth in E420K-variant cells. Rapamycin reduced cell size and dose-dependently reduced RPS6 phosphorylation in E420K-variant cells, suggesting that inhibition of mTOR1 can suppress both the observed RPS6 hyperphosphorylation and increased cell size. Together, our findings provide a deeper understanding of PPP2R5D and insight into how the E420K-variant alters signaling networks influenced by PPP2R5D. Our comprehensive approach, which combines precise genome editing, isobaric tandem mass tag labeling of peptides generated from endogenously expressed proteins, and concurrent liquid chromatography-mass spectrometry (LC-MS
3 ), also provides a roadmap that can be used to rapidly explore the etiologies of additional genetic disorders., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
10. S100A6 is a positive regulator of PPP5C-FKBP51-dependent regulation of endothelial calcium signaling.
- Author
-
Haldar B, Hamilton CL, Solodushko V, Abney KA, Alexeyev M, Honkanen RE, Scammell JG, and Cioffi DL
- Subjects
- Animals, Cells, Cultured, Endothelium, Vascular cytology, Lung blood supply, Nuclear Proteins metabolism, Phosphoprotein Phosphatases metabolism, Protein Binding, Protein Transport, Rats, TRPC Cation Channels metabolism, Tacrolimus Binding Proteins metabolism, Calcium Signaling, Cell Cycle Proteins metabolism, Endothelial Cells metabolism, S100 Calcium Binding Protein A6 metabolism
- Abstract
I
SOC is a cation current permeating the ISOC channel. In pulmonary endothelial cells, ISOC activation leads to formation of inter-endothelial cell gaps and barrier disruption. The immunophilin FK506-binding protein 51 (FKBP51), in conjunction with the serine/threonine protein phosphatase 5C (PPP5C), inhibits ISOC . Free PPP5C assumes an autoinhibitory state, which has low "basal" catalytic activity. Several S100 protein family members bind PPP5C increasing PPP5C catalytic activity in vitro. One of these family members, S100A6, exhibits a calcium-dependent translocation to the plasma membrane. The goal of this study was to determine whether S100A6 activates PPP5C in pulmonary endothelial cells and contributes to ISOC inhibition by the PPP5C-FKBP51 axis. We observed that S100A6 activates PPP5C to dephosphorylate tau T231. Following ISOC activation, cytosolic S100A6 translocates to the plasma membrane and interacts with the TRPC4 subunit of the ISOC channel. Global calcium entry and ISOC are decreased by S100A6 in a PPP5C-dependent manner and by FKBP51 in a S100A6-dependent manner. Further, calcium entry-induced endothelial barrier disruption is decreased by S100A6 dependent upon PPP5C, and by FKBP51 dependent upon S100A6. Overall, these data reveal that S100A6 plays a key role in the PPP5C-FKBP51 axis to inhibit ISOC and protect the endothelial barrier against calcium entry-induced disruption., (© 2020 Federation of American Societies for Experimental Biology.)- Published
- 2020
- Full Text
- View/download PDF
11. Targeting phosphatases in cancer: suppression of many versus the ablation of one.
- Author
-
D'Arcy BM, Prakash A, and Honkanen RE
- Published
- 2019
- Full Text
- View/download PDF
12. Development of a Synthetic 3-ketosteroid Δ 1 -dehydrogenase for the Generation of a Novel Catabolic Pathway Enabling Cholesterol Degradation in Human Cells.
- Author
-
D'Arcy BM, Swingle MR, Schambeau L, Pannell L, Prakash A, and Honkanen RE
- Subjects
- Cell Line, Escherichia coli, Genetic Engineering, Humans, Oxidoreductases genetics, Proof of Concept Study, Cholesterol metabolism, Oxidoreductases chemistry, Oxidoreductases metabolism
- Abstract
Cholesterol is an essential component of membranes, which is acquired by cells via receptor-mediated endocytosis of lipoproteins or via de novo synthesis. In specialized cells, anabolic enzymes metabolize cholesterol, generating steroid hormones or bile acids. However, surplus cholesterol cannot be catabolized due to the lack of enzymes capable of degrading the cholestane ring. The inability to degrade cholesterol becomes evident in the development and progression of cardiovascular disease, where the accumulation of cholesterol/cholesteryl-esters in macrophages can elicit a maladaptive immune response leading to the development and progression of atherosclerosis. The discovery of cholesterol catabolic pathways in Actinomycetes led us to the hypothesis that if enzymes enabling cholesterol catabolism could be genetically engineered and introduced into human cells, the atherosclerotic process may be prevented or reversed. Comparison of bacterial enzymes that degrade cholesterol to obtain carbon and generate energy with the action of human enzymes revealed that humans lack a 3-ketosteroid Δ
1 -dehydrogenase (Δ1 -KstD), which catalyzes the C-1 and C-2 desaturation of ring A. Here we describe the construction, heterologous expression, and actions of a synthetic humanized Δ1 -KstD expressed in Hep3B and U-937 cells, providing proof that one of three key enzymes required for cholesterol ring opening can be functionally expressed in human cells.- Published
- 2019
- Full Text
- View/download PDF
13. The Antitumor Drug LB-100 Is a Catalytic Inhibitor of Protein Phosphatase 2A (PPP2CA) and 5 (PPP5C) Coordinating with the Active-Site Catalytic Metals in PPP5C.
- Author
-
D'Arcy BM, Swingle MR, Papke CM, Abney KA, Bouska ES, Prakash A, and Honkanen RE
- Subjects
- Amino Acid Sequence genetics, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Catalysis, Catalytic Domain drug effects, Cell Line, Tumor, Humans, Metals chemistry, Methylation, Mutagenesis, Site-Directed, Neoplasms genetics, Neoplasms pathology, Nuclear Proteins antagonists & inhibitors, Phosphoprotein Phosphatases antagonists & inhibitors, Piperazines pharmacology, Protein Phosphatase 2 antagonists & inhibitors, Protein Phosphatase 2 genetics, Bridged Bicyclo Compounds, Heterocyclic chemistry, Neoplasms drug therapy, Nuclear Proteins chemistry, Phosphoprotein Phosphatases chemistry, Piperazines chemistry, Protein Phosphatase 2 chemistry
- Abstract
LB-100 is an experimental cancer therapeutic with cytotoxic activity against cancer cells in culture and antitumor activity in animals. The first phase I trial (NCT01837667) evaluating LB-100 recently concluded that safety and efficacy parameters are favorable for further clinical testing. Although LB-100 is widely reported as a specific inhibitor of serine/threonine phosphatase 2A (PP2AC/ PPP2CA:PPP2CB ), we could find no experimental evidence in the published literature demonstrating the specific engagement of LB-100 with PP2A in vitro , in cultured cells, or in animals. Rather, the premise for LB-100 targeting PP2AC is derived from studies that measure phosphate released from a phosphopeptide (K-R-pT-I-R-R) or inferred from the ability of LB-100 to mimic activity previously reported to result from the inhibition of PP2AC by other means. PP2AC and PPP5C share a common catalytic mechanism. Here, we demonstrate that the phosphopeptide used to ascribe LB-100 specificity for PP2A is also a substrate for PPP5C. Inhibition assays using purified enzymes demonstrate that LB-100 is a catalytic inhibitor of both PP2AC and PPP5C. The structure of PPP5C cocrystallized with LB-100 was solved to a resolution of 1.65Å, revealing that the 7-oxabicyclo[2.2.1]heptane-2,3-dicarbonyl moiety coordinates with the metal ions and key residues that are conserved in both PP2AC and PPP5C. Cell-based studies revealed some known actions of LB-100 are mimicked by the genetic disruption of PPP5C These data demonstrate that LB-100 is a catalytic inhibitor of both PP2AC and PPP5C and suggest that the observed antitumor activity might be due to an additive effect achieved by suppressing both PP2A and PPP5C., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
14. Inhibitors of Serine/Threonine Protein Phosphatases: Biochemical and Structural Studies Provide Insight for Further Development.
- Author
-
Swingle MR and Honkanen RE
- Subjects
- Animals, Catalysis, Catalytic Domain, Enzyme Inhibitors metabolism, Enzyme Inhibitors pharmacology, Humans, Isoenzymes antagonists & inhibitors, Isoenzymes chemistry, Isoenzymes genetics, Isoenzymes metabolism, Phosphoprotein Phosphatases chemistry, Phosphoprotein Phosphatases genetics, Phosphoprotein Phosphatases metabolism, Protein Binding, Protein Domains, Enzyme Inhibitors chemistry, Phosphoprotein Phosphatases antagonists & inhibitors
- Abstract
Background: The reversible phosphorylation of proteins regulates many key functions in eukaryotic cells. Phosphorylation is catalyzed by protein kinases, with the majority of phosphorylation occurring on side chains of serine and threonine residues. The phosphomonoesters generated by protein kinases are hydrolyzed by protein phosphatases. In the absence of a phosphatase, the half-time for the hydrolysis of alkyl phosphate dianions at 25º C is over 1 trillion years; knon ~2 x 10-20 sec-1. Therefore, ser/thr phosphatases are critical for processes controlled by reversible phosphorylation., Methods: This review is based on the literature searched in available databases. We compare the catalytic mechanism of PPP-family phosphatases (PPPases) and the interactions of inhibitors that target these enzymes., Results: PPPases are metal-dependent hydrolases that enhance the rate of hydrolysis ([kcat/kM]/knon ) by a factor of ~1021, placing them among the most powerful known catalysts on earth. Biochemical and structural studies indicate that the remarkable catalytic proficiencies of PPPases are achieved by 10 conserved amino acids, DXH(X)~26DXXDR(X)~20- 26NH(X)~50H(X)~25-45R(X)~30-40H. Six act as metal-coordinating residues. Four position and orient the substrate phosphate. Together, two metal ions and the 10 catalytic residues position the phosphoryl group and an activated bridging water/hydroxide nucleophile for an inline attack upon the substrate phosphorous atom. The PPPases are conserved among species, and many structurally diverse natural toxins co-evolved to target these enzymes., Conclusion: Although the catalytic site is conserved, opportunities for the development of selective inhibitors of this important group of metalloenzymes exist., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.net.)
- Published
- 2019
- Full Text
- View/download PDF
15. Serine/threonine phosphatase 5 (PP5C/PPP5C) regulates the ISOC channel through a PP5C-FKBP51 axis.
- Author
-
Hamilton CL, Abney KA, Vasauskas AA, Alexeyev M, Li N, Honkanen RE, Scammell JG, and Cioffi DL
- Abstract
Pulmonary endothelial cells express a store-operated calcium entry current ( I
soc ), which contributes to inter-endothelial cell gap formation. Isoc is regulated by a heterocomplex of proteins that includes the immunophilin FKBP51. FKBP51 inhibits Isoc by mechanisms that are not fully understood. In pulmonary artery endothelial cells (PAECs) we have shown that FKBP51 increases microtubule polymerization, an event that is critical for Isoc inhibition by FKBP51. In neurons, FKBP51 promotes microtubule stability through facilitation of tau dephosphorylation. However, FKBP51 does not possess phosphatase activity. Protein phosphatase 5 (PP5C/PPP5C) can dephosphorylate tau, and similar to FKBP51, PP5C possesses tetratricopeptide repeats (TPR) that mediate interaction with heat shock protein-90 (HSP90) chaperone/scaffolding complexes. We therefore tested whether PP5C contributes to FKBP51-mediated inhibition of Isoc . Both siRNA-mediated suppression of PP5C expression in PAECs and genetic disruption of PP5C in HEK293 cells attenuate FKBP51-mediated inhibition of Isoc . Reintroduction of catalytically competent, but not catalytically inactive PP5C, restored FKBP51-mediated inhibition of Isoc . PAEC cell fractionation studies identified both PP5C and the ISOC heterocomplex in the same membrane fractions. Further, PP5C co-precipitates with TRPC4, an essential subunit of ISOC channel. Finally, to determine if PP5C is required for FKBP51-mediated inhibition of calcium entry-induced inter-endothelial cell gap formation, we measured gap area by wide-field microscopy and performed biotin gap quantification assay and electric cell-substrate impedance sensing (ECIS®). Collectively, the data presented indicate that suppression of PP5C expression negates the protective effect of FKBP51. These observations identify PP5C as a novel member of the ISOC heterocomplex that is required for FKBP51-mediated inhibition of Isoc .- Published
- 2018
- Full Text
- View/download PDF
16. PP1:Tautomycetin Complex Reveals a Path toward the Development of PP1-Specific Inhibitors.
- Author
-
Choy MS, Swingle M, D'Arcy B, Abney K, Rusin SF, Kettenbach AN, Page R, Honkanen RE, and Peti W
- Subjects
- Amino Acid Sequence, Humans, Lipids, Models, Molecular, Protein Phosphatase 1 chemistry, Substrate Specificity, Enzyme Inhibitors pharmacology, Furans metabolism, Protein Phosphatase 1 antagonists & inhibitors, Protein Phosphatase 1 metabolism
- Abstract
Selective inhibitors for each serine/threonine phosphatase (PPP) are essential to investigate the biological actions of PPPs and to guide drug development. Biologically diverse organisms (e.g., cyanobacteria, dinoflagellates, beetles) produce structurally distinct toxins that are catalytic inhibitors of PPPs. However, most toxins exhibit little selectivity, typically inhibiting multiple family members with similar potencies. Thus, the use of these toxins as chemical tools to study the relationship between individual PPPs and their biological substrates, and how disruptions in these relationships contributes to human disease, is severely limited. Here, we show that tautomycetin (TTN) is highly selective for a single PPP, protein phosphatase 1 (PP1/PPP1C). Our structure of the PP1:TTN complex reveals that PP1 selectivity is defined by a covalent bond between TTN and a PP1-specific cysteine residue, Cys127. Together, these data provide key molecular insights needed for the development of novel probes targeting single PPPs, especially PP1.
- Published
- 2017
- Full Text
- View/download PDF
17. An Ultra-High-Throughput Screen for Catalytic Inhibitors of Serine/Threonine Protein Phosphatases Types 1 and 5 (PP1C and PP5C).
- Author
-
Swingle M, Volmar CH, Saldanha SA, Chase P, Eberhart C, Salter EA, D'Arcy B, Schroeder CE, Golden JE, Wierzbicki A, Hodder P, and Honkanen RE
- Subjects
- Catalysis, Enzyme Assays, Enzyme Inhibitors chemistry, Humans, Miniaturization, Phosphoproteins metabolism, Protein Phosphatase 1 metabolism, Radiopharmaceuticals chemistry, Reproducibility of Results, Substrate Specificity, Enzyme Inhibitors analysis, Enzyme Inhibitors pharmacology, High-Throughput Screening Assays methods, Protein Phosphatase 1 antagonists & inhibitors
- Abstract
Although there has been substantial success in the development of specific inhibitors for protein kinases, little progress has been made in the identification of specific inhibitors for their protein phosphatase counterparts. Inhibitors of PP1 and PP5 are desired as probes for research and to test their potential for drug development. We developed and miniaturized (1536-well plate format) nearly identical homogeneous, fluorescence intensity (FLINT) enzymatic assays to detect inhibitors of PP1 or PP5. The assays were used in an ultra-high-throughput screening (uHTS) campaign, testing >315,000 small-molecule compounds. Both assays demonstrated robust performance, with a Z' of 0.92 ± 0.03 and 0.95 ± 0.01 for the PP1 and PP5 assays, respectively. Screening the same library with both assays aided the identification of class inhibitors and assay artifacts. Confirmation screening and hit prioritization assays used [
32 P/33 P]-radiolabel protein substrates, revealing excellent agreement between the FLINT and radiolabel assays. This screening campaign led to the discovery of four novel unrelated small-molecule inhibitors of PP1 and ~30 related small-molecule inhibitors of PP5. The results suggest that this uHTS approach is suitable for identifying selective chemical probes that inhibit PP1 or PP5 activity, and it is likely that similar assays can be developed for other PPP-family phosphatases.- Published
- 2017
- Full Text
- View/download PDF
18. Crystal structures and mutagenesis of PPP-family ser/thr protein phosphatases elucidate the selectivity of cantharidin and novel norcantharidin-based inhibitors of PP5C.
- Author
-
Chattopadhyay D, Swingle MR, Salter EA, Wood E, D'Arcy B, Zivanov C, Abney K, Musiyenko A, Rusin SF, Kettenbach A, Yet L, Schroeder CE, Golden JE, Dunham WH, Gingras AC, Banerjee S, Forbes D, Wierzbicki A, and Honkanen RE
- Subjects
- Amino Acid Sequence, Binding Sites, Catalytic Domain, Cloning, Molecular, Crystallography, X-Ray, Escherichia coli genetics, Escherichia coli metabolism, Gene Expression, Humans, Kinetics, Molecular Docking Simulation, Mutagenesis, Site-Directed, Nuclear Proteins antagonists & inhibitors, Nuclear Proteins genetics, Nuclear Proteins metabolism, Phosphoprotein Phosphatases antagonists & inhibitors, Phosphoprotein Phosphatases genetics, Phosphoprotein Phosphatases metabolism, Protein Binding, Protein Domains, Protein Phosphatase 1 antagonists & inhibitors, Protein Phosphatase 1 genetics, Protein Phosphatase 1 metabolism, Protein Structure, Secondary, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Sequence Alignment, Structure-Activity Relationship, Bridged Bicyclo Compounds, Heterocyclic chemistry, Cantharidin chemistry, Enzyme Inhibitors chemistry, Nuclear Proteins chemistry, Phosphoprotein Phosphatases chemistry, Protein Phosphatase 1 chemistry
- Abstract
Cantharidin is a natural toxin and an active constituent in a traditional Chinese medicine used to treat tumors. Cantharidin acts as a semi-selective inhibitor of PPP-family ser/thr protein phosphatases. Despite sharing a common catalytic mechanism and marked structural similarity with PP1C, PP2AC and PP5C, human PP4C was found to be insensitive to the inhibitory activity of cantharidin. To explore the molecular basis for this selectivity, we synthesized and tested novel C5/C6-derivatives designed from quantum-based modeling of the interactions revealed in the co-crystal structures of PP5C in complex with cantharidin. Structure-activity relationship studies and analysis of high-resolution (1.25Å) PP5C-inhibitor co-crystal structures reveal close contacts between the inhibitor bridgehead oxygen and both a catalytic metal ion and a non-catalytic phenylalanine residue, the latter of which is substituted by tryptophan in PP4C. Quantum chemistry calculations predicted that steric clashes with the bulkier tryptophan side chain in PP4C would force all cantharidin-based inhibitors into an unfavorable binding mode, disrupting the strong coordination of active site metal ions observed in the PP5C co-crystal structures, thereby rendering PP4C insensitive to the inhibitors. This prediction was confirmed by inhibition studies employing native human PP4C. Mutation of PP5C (F446W) and PP1C (F257W), to mimic the PP4C active site, resulted in markedly suppressed sensitivity to cantharidin. These observations provide insight into the structural basis for the natural selectivity of cantharidin and provide an avenue for PP4C deselection. The novel crystal structures also provide insight into interactions that provide increased selectivity of the C5/C6 modifications for PP5C versus other PPP-family phosphatases., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
19. Modeling the antiferromagnetic MnIIMnII system within the protein phosphatase-5 catalytic site.
- Author
-
Salter EA, Honkanen RE, and Wierzbicki A
- Subjects
- Nuclear Proteins metabolism, Phosphoprotein Phosphatases metabolism, Catalytic Domain, Enzyme Inhibitors chemistry, Manganese chemistry, Models, Molecular, Nuclear Proteins chemistry, Phosphoprotein Phosphatases chemistry
- Abstract
Protein phosphatase-5 (PP5), a novel target for inhibition in a search for new antitumor drugs, contains a homobimetallic Mn(II)Mn(II) system in its catalytic site. The ground electronic state is an antiferromagnetically-coupled singlet. We report optimizations of a known inhibitor within a 42-residue model of the PP5 catalytic site under several two-level hybrid ONIOM computational models. Using the high-resolution crystal structure of a PP5/inhibitor complex as reference, we compare geometric parameters as the qualities of the "high-level" and "low-level" wavefunctions are successively improved by using the correct antiferromagnetic (AF) singlet state. We find that the UB3LYP AF wavefunction for the high-level region is necessary for experimental fidelity. A closed-shell semi-empirical method (RPM6) can be used for the low-quality part of the hybrid scheme to afford geometries which are qualitatively on par with that obtained using the more time-consuming open-shell UB3LYP AF wavefunction. As the AF state can be elusive for such a large system, the ferromagnetic (F) state can also be used in the low-quality calculations without impacting the geometry.
- Published
- 2015
- Full Text
- View/download PDF
20. Development and validation of a robust and sensitive assay for the discovery of selective inhibitors for serine/threonine protein phosphatases PP1α (PPP1C) and PP5 (PPP5C).
- Author
-
Swingle MR and Honkanen RE
- Subjects
- Cantharidin chemistry, Dimethyl Sulfoxide chemistry, Fluorescence, Humans, Indicators and Reagents, Kinetics, Octoxynol, Reproducibility of Results, Small Molecule Libraries, Substrate Specificity, Surface-Active Agents, Enzyme Inhibitors pharmacology, High-Throughput Screening Assays methods, Nuclear Proteins antagonists & inhibitors, Phosphoprotein Phosphatases antagonists & inhibitors, Protein Phosphatase 1 antagonists & inhibitors
- Abstract
Protein phosphatase types 1 α (PP1α/PPP1C) and 5 (PP5/PPP5C) are members of the PPP family of serine/threonine protein phosphatases. PP1 and PP5 share a common catalytic mechanism, and several natural compounds, including okadaic acid, microcystin, and cantharidin, act as strong inhibitors of both enzymes. However, to date there have been no reports of compounds that can selectively inhibit PP1 or PP5, and specific or highly selective inhibitors for either PP1 or PP5 are greatly desired by both the research and pharmaceutical communities. Here we describe the development and optimization of a sensitive and robust (representative PP5C assay data: Z'=0.93; representative PP1Cα assay data: Z'=0.90) fluorescent phosphatase assay that can be used to simultaneously screen chemical libraries and natural product extracts for the presence of catalytic inhibitors of PP1 and PP5.
- Published
- 2014
- Full Text
- View/download PDF
21. Protein phosphatases in pancreatic islets.
- Author
-
Ortsäter H, Grankvist N, Honkanen RE, and Sjöholm Å
- Subjects
- Diabetes Mellitus metabolism, Glucose metabolism, Humans, Insulin Secretion, Insulin-Secreting Cells metabolism, Models, Biological, Phosphoprotein Phosphatases classification, Phosphorylation, Insulin metabolism, Islets of Langerhans metabolism, Phosphoprotein Phosphatases metabolism, Phosphoproteins metabolism
- Abstract
The prevalence of diabetes is increasing rapidly worldwide. A cardinal feature of most forms of diabetes is the lack of insulin-producing capability, due to the loss of insulin-producing β-cells, impaired glucose-sensitive insulin secretion from the β-cell, or a combination thereof, the reasons for which largely remain elusive. Reversible phosphorylation is an important and versatile mechanism for regulating the biological activity of many intracellular proteins, which, in turn, controls a variety of cellular functions. For instance, significant changes in protein kinase activities and in protein phosphorylation patterns occur subsequent to the stimulation of insulin release by glucose. Therefore, the molecular mechanisms regulating the phosphorylation of proteins involved in the insulin secretory process by the β-cell have been extensively investigated. However, far less is known about the role and regulation of protein dephosphorylation by various protein phosphatases. Herein, we review extant data implicating serine/threonine and tyrosine phosphatases in various aspects of healthy and diabetic islet biology, ranging from control of hormonal stimulus-secretion coupling to mitogenesis and apoptosis., (© 2014 Society for Endocrinology.)
- Published
- 2014
- Full Text
- View/download PDF
22. Mitogen-activated protein kinases and protein phosphatase 5 mediate glucocorticoid-induced cytotoxicity in pancreatic islets and β-cells.
- Author
-
Fransson L, Rosengren V, Saha TK, Grankvist N, Islam T, Honkanen RE, Sjöholm Å, and Ortsäter H
- Subjects
- Animals, Apoptosis drug effects, Caspase 3 genetics, Caspase 3 metabolism, DNA Fragmentation drug effects, Gene Expression Regulation, Insulin-Secreting Cells cytology, Insulin-Secreting Cells metabolism, JNK Mitogen-Activated Protein Kinases antagonists & inhibitors, JNK Mitogen-Activated Protein Kinases genetics, JNK Mitogen-Activated Protein Kinases metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Nuclear Proteins genetics, Phosphoprotein Phosphatases genetics, Phosphorylation, Protein Kinase Inhibitors pharmacology, Signal Transduction, Stress, Physiological drug effects, p38 Mitogen-Activated Protein Kinases antagonists & inhibitors, p38 Mitogen-Activated Protein Kinases genetics, Dexamethasone pharmacology, Glucocorticoids pharmacology, Insulin-Secreting Cells drug effects, Nuclear Proteins metabolism, Phosphoprotein Phosphatases metabolism, p38 Mitogen-Activated Protein Kinases metabolism
- Abstract
Glucocorticoid excess is associated with glucose intolerance and diabetes. In addition to inducing insulin resistance, glucocorticoids impair β-cell function and cause β-cell apoptosis. In this study we show that dexamethasone activates mitogen-activated protein kinases (MAPKs) signaling in MIN6 β-cells, as evident by enhanced phosphorylation of p38 MAPK and c-Jun N-terminal kinase (JNK). In contrast, the integrated stress response pathway was inhibited by dexamethasone. A p38 MAPK inhibitor attenuated dexamethasone-induced apoptosis in β-cells and isolated islets and decreased glucocorticoid receptor phosphorylation at S220. In contrast, a JNK inhibitor augmented DNA fragmentation and dexamethasone-induced formation of cleaved caspase 3. We also show that inhibition of protein phosphatase 5 (PP5) augments apoptosis in dexamethasone-exposed islets and β-cells, with a concomitant activation of p38 MAPK. In conclusion, our data provide evidence that in islets and β-cells, p38 MAPK and JNK phosphorylation work in concert with PP5 to regulate the cytotoxic effects exerted by glucocorticoids., (Copyright © 2013 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
23. Small G proteins Rac1 and Ras regulate serine/threonine protein phosphatase 5 (PP5)·extracellular signal-regulated kinase (ERK) complexes involved in the feedback regulation of Raf1.
- Author
-
Mazalouskas MD, Godoy-Ruiz R, Weber DJ, Zimmer DB, Honkanen RE, and Wadzinski BE
- Subjects
- Amino Acid Substitution, Animals, Cattle, Cell Line, Extracellular Signal-Regulated MAP Kinases genetics, Glycoproteins genetics, Humans, Multienzyme Complexes genetics, Mutation, Missense, Phosphorylation, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-raf genetics, Proto-Oncogene Proteins p21(ras) genetics, Rats, rac1 GTP-Binding Protein genetics, ras Proteins genetics, Extracellular Signal-Regulated MAP Kinases metabolism, Glycoproteins metabolism, Multienzyme Complexes metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-raf metabolism, Proto-Oncogene Proteins p21(ras) metabolism, rac1 GTP-Binding Protein metabolism, ras Proteins metabolism
- Abstract
Serine/threonine protein phosphatase 5 (PP5, PPP5C) is known to interact with the chaperonin heat shock protein 90 (HSP90) and is involved in the regulation of multiple cellular signaling cascades that control diverse cellular processes, such as cell growth, differentiation, proliferation, motility, and apoptosis. Here, we identify PP5 in stable complexes with extracellular signal-regulated kinases (ERKs). Studies using mutant proteins reveal that the formation of PP5·ERK1 and PP5·ERK2 complexes partially depends on HSP90 binding to PP5 but does not require PP5 or ERK1/2 activity. However, PP5 and ERK activity regulates the phosphorylation state of Raf1 kinase, an upstream activator of ERK signaling. Whereas expression of constitutively active Rac1 promotes the assembly of PP5·ERK1/2 complexes, acute activation of ERK1/2 fails to influence the phosphatase-kinase interaction. Introduction of oncogenic HRas (HRas(V12)) has no effect on PP5-ERK1 binding but selectively decreases the interaction of PP5 with ERK2, in a manner that is independent of PP5 and MAPK/ERK kinase (MEK) activity, yet paradoxically requires ERK2 activity. Additional studies conducted with oncogenic variants of KRas4B reveal that KRas(L61), but not KRas(V12), also decreases the PP5-ERK2 interaction. The expression of wild type HRas or KRas proteins fails to reduce PP5-ERK2 binding, indicating that the effect is specific to HRas(V12) and KRas(L61) gain-of-function mutations. These findings reveal a novel, differential responsiveness of PP5-ERK1 and PP5-ERK2 interactions to select oncogenic Ras variants and also support a role for PP5·ERK complexes in regulating the feedback phosphorylation of PP5-associated Raf1.
- Published
- 2014
- Full Text
- View/download PDF
24. Genetic disruption of protein phosphatase 5 in mice prevents high-fat diet feeding-induced weight gain.
- Author
-
Grankvist N, Honkanen RE, Sjöholm Å, and Ortsäter H
- Subjects
- Animals, Blood Glucose, Diet, High-Fat adverse effects, Insulin blood, Insulin Resistance genetics, Mice, Mice, Knockout, Nuclear Proteins deficiency, Obesity etiology, Obesity genetics, Phosphoprotein Phosphatases deficiency, Nuclear Proteins genetics, Phosphoprotein Phosphatases genetics, Weight Gain genetics
- Abstract
The role of serine/threonine protein phosphatase 5 (PP5) in the development of obesity and insulin resistance associated with high-fat diet-feeding (HFD) was examined using PP5-deficient mice (Ppp5c(-/-)). Despite similar caloric intake, Ppp5c(-/-) mice on HFD gained markedly less weight and did not accumulate visceral fat compared to wild-type littermates (Ppp5c(+/+)). On a control diet, Ppp5c(-/-) mice had markedly improved glucose control compared to Ppp5c(+/+) mice, an effect diminished by HFD. However, even after 10 weeks of HFD glucose control in Ppp5c(-/-) mice was similar to that observed in Ppp5c(+/+) mice on the control diet. Thus, PP5 deficiency confers protection against HFD-induced weight gain in mice., (Copyright © 2013 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
25. Suppression of Ser/Thr phosphatase 4 (PP4C/PPP4C) mimics a novel post-mitotic action of fostriecin, producing mitotic slippage followed by tetraploid cell death.
- Author
-
Theobald B, Bonness K, Musiyenko A, Andrews JF, Urban G, Huang X, Dean NM, and Honkanen RE
- Subjects
- Dose-Response Relationship, Drug, HeLa Cells, Humans, Molecular Mimicry, Nuclear Proteins antagonists & inhibitors, Nuclear Proteins metabolism, Phosphoprotein Phosphatases metabolism, Protein Phosphatase 1 antagonists & inhibitors, Protein Phosphatase 1 metabolism, Protein Phosphatase 2 antagonists & inhibitors, Protein Phosphatase 2 metabolism, Tetraploidy, Antineoplastic Agents pharmacology, Apoptosis drug effects, Cell Cycle drug effects, Mitosis drug effects, Phosphoprotein Phosphatases antagonists & inhibitors, Polyenes pharmacology, Pyrones pharmacology
- Abstract
Unlabelled: Fostriecin is a natural product purified from Sterptomyces extracts with antitumor activity sufficient to warrant human clinical trials. Unfortunately, difficulties associated with supply and stable drug formulation stalled further development. At a molecular level, fostriecin is known to act as a catalytic inhibitor of four PPP-family phosphatases, and reports describing the design of molecules in this class suggest derivatives targeting enzymes within the fostriecin-sensitive subfamily can be successful. However, it is not clear if the tumor-selective cytotoxicity of fostriecin results from the inhibition of a specific phosphatase, multiple phosphatases, or a limited subset of fostriecin sensitive phosphatases. How the inhibition of sensitive phosphatases contributes to tumor-selective cytotoxicity is also not clear. Here, high-content time-lapse imaging of live cells revealed novel insight into the cellular actions of fostriecin, showing that fostriecin-induced apoptosis is not simply induced following a sustained mitotic arrest. Rather, apoptosis occurred in an apparent second interphase produced when tetraploid cells undergo mitotic slippage. Comparison of the actions of fostriecin and antisense-oligonucleotides specifically targeting human fostriecin-sensitive phosphatases revealed that the suppression PP4C alone is sufficient to mimic many actions of fostriecin. Importantly, targeted suppression of PP4C induced apoptosis, with death occurring in tetraploid cells following mitotic slippage. This effect was not observed following the suppression of PP1C, PP2AC, or PP5C. These data clarify PP4C as a fostriecin-sensitive phosphatase and demonstrate that the suppression of PP4C triggers mitotic slippage/apoptosis., Implications: Future development of fostriecin class inhibitors should consider PP4C as a potentially important target. Mol Cancer Res; 11(8); 845-55. ©2013 AACR., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
26. DNAJB6 chaperones PP2A mediated dephosphorylation of GSK3β to downregulate β-catenin transcription target, osteopontin.
- Author
-
Mitra A, Menezes ME, Pannell LK, Mulekar MS, Honkanen RE, Shevde LA, and Samant RS
- Subjects
- Animals, Cell Line, Tumor, Down-Regulation, Epithelial-Mesenchymal Transition, Female, Gene Expression Regulation, Neoplastic, Glycogen Synthase Kinase 3 beta, HSC70 Heat-Shock Proteins metabolism, HSP40 Heat-Shock Proteins genetics, HSP40 Heat-Shock Proteins metabolism, Humans, Lymphoid Enhancer-Binding Factor 1 genetics, Lymphoid Enhancer-Binding Factor 1 metabolism, Melanoma metabolism, Melanoma secondary, Mice, Mice, Nude, Molecular Chaperones genetics, Molecular Chaperones metabolism, Neoplasm Transplantation, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Okadaic Acid pharmacology, Oligonucleotide Array Sequence Analysis, Osteopontin metabolism, Phosphorylation, Protein Binding, Protein Interaction Domains and Motifs, Protein Phosphatase 2 antagonists & inhibitors, Protein Processing, Post-Translational, Protein Structure, Tertiary, Skin Neoplasms metabolism, Skin Neoplasms pathology, T Cell Transcription Factor 1 genetics, T Cell Transcription Factor 1 metabolism, Transcription, Genetic, Transcriptome, Glycogen Synthase Kinase 3 metabolism, HSP40 Heat-Shock Proteins physiology, Molecular Chaperones physiology, Nerve Tissue Proteins physiology, Osteopontin genetics, Protein Phosphatase 2 metabolism, beta Catenin metabolism
- Abstract
Elevated levels of the oncoprotein, osteopontin (OPN), are associated with poor outcome of several types of cancers including melanoma. We have previously reported an important involvement of DNAJB6, a member of heat-shock protein 40 (HSP40) family, in negatively impacting tumor growth. The current study was prompted by our observations reported here which revealed a reciprocal relationship between DNAJB6 and OPN in melanoma specimens. The 'J domain' is the most conserved domain of HSP40 family of proteins. Hence, we assessed the functional role of the J domain in activities of DNAJB6. We report that the J domain of DNAJB6 is involved in mediating OPN suppression. Deletion of the J domain renders DNAJB6 incapable of impeding malignancy and suppressing OPN. Our mechanistic investigations reveal that DNAJB6 binds HSPA8 (heat-shock cognate protein, HSC70) and causes dephosphorylation of glycogen synthase kinase 3β (GSK3β) at Ser 9 by recruiting protein phosphatase, PP2A. This dephosphorylation activates GSK3β, leading to degradation of β-catenin and subsequent loss of TCF/LEF (T cell factor1/lymphoid enhancer factor1) activity. Deletion of the J domain abrogates assembly of this multiprotein complex and renders GSK3β inactive, thus, stabilizing β-catenin, a transcription co-activator for OPN expression. Our in-vitro and in-vivo functional analyses show that silencing OPN expression in the background of deletion of the J domain renders the resultant tumor cells less malignant despite the presence of stabilized β-catenin. Thus, we have uncovered a new mechanism for regulation of GSK3β activity leading to inhibition of Wnt/β-catenin signaling.
- Published
- 2012
- Full Text
- View/download PDF
27. Cytokines alter glucocorticoid receptor phosphorylation in airway cells: role of phosphatases.
- Author
-
Bouazza B, Krytska K, Debba-Pavard M, Amrani Y, Honkanen RE, Tran J, and Tliba O
- Subjects
- Androstadienes pharmacology, Cells, Cultured, Fluticasone, Gene Knockdown Techniques, Glucocorticoids pharmacology, Glucocorticoids physiology, Humans, Mutation, Missense, Myocytes, Smooth Muscle metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism, Phosphoprotein Phosphatases genetics, Phosphoprotein Phosphatases metabolism, Phosphorylation, RNA Interference, Receptors, Glucocorticoid genetics, Cytokines physiology, Myocytes, Smooth Muscle enzymology, Nuclear Proteins physiology, Phosphoprotein Phosphatases physiology, Protein Processing, Post-Translational, Receptors, Glucocorticoid metabolism, Respiratory System cytology
- Abstract
Corticosteroid insensitivity (CSI) represents a profound challenge in managing patients with asthma. We recently demonstrated that short exposure of airway smooth muscle cells (ASMCs) to proasthmatic cytokines drastically reduced their responsiveness to glucocorticoids (GCs), an effect that was partially mediated via interferon regulatory factor-1, suggesting the involvement of additional mechanisms (Am J Respir Cell Mol Biol 2008;38:463-472). Although GC receptor (GR) can be phosphorylated at multiple serines in the N-terminal region, the major phosphorylation sites critical for GR transcriptional activity are serines 211 (Ser211) and 226 (Ser226). We tested the novel hypothesis that cytokine-induced CSI in ASMCs is due to an impaired GR phosphorylation. Cells were treated with TNF-α (10 ng/ml) and IFN-γ (500 UI/ml) for 6 hours and/or fluticasone (100 nm) added 2 hours before. GR was constitutively phosphorylated at Ser226 but not at Ser211 residues. Cytokines dramatically suppressed fluticasone-induced phosphorylation of GR on Ser211 but not on Ser226 residues while increasing the expression of Ser/Thr protein phosphatase (PP)5 but not that of PP1 or PP2A. Transfection studies using a reporter construct containing GC responsive elements showed that the specific small interfering RNA-induced mRNA knockdown of PP5, but not that of PP1 or PP2A, partially prevented the cytokine suppressive effects on GR-meditated transactivation activity. Similarly, cytokines failed to inhibit GC-induced GR-Ser211 phosphorylation when expression of PP5 was suppressed. We propose that the novel mechanism that proasthmatic cytokine-induced CSI in ASMCs is due, in part, to PP5-mediated impairment of GR-Ser211 phosphorylation.
- Published
- 2012
- Full Text
- View/download PDF
28. Serine/threonine protein phosphatase 5 regulates glucose homeostasis in vivo and apoptosis signalling in mouse pancreatic islets and clonal MIN6 cells.
- Author
-
Grankvist N, Amable L, Honkanen RE, Sjöholm A, and Ortsäter H
- Subjects
- Animals, Apoptosis, Base Sequence, Glucose Tolerance Test, Homeostasis, Male, Mice, Mitogen-Activated Protein Kinases metabolism, Nuclear Proteins pharmacology, Phosphoprotein Phosphatases pharmacology, Signal Transduction, Diabetes Mellitus, Experimental metabolism, Diabetes Mellitus, Type 2 metabolism, Islets of Langerhans metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Nuclear Proteins metabolism, Phosphoprotein Phosphatases metabolism, Reactive Oxygen Species metabolism
- Abstract
Aims/hypothesis: During the development of type 2 diabetes mellitus, beta cells are often exposed to a high glucose/hyperlipidaemic environment, in which the levels of reactive oxygen species (ROS) are elevated. In turn, ROS can trigger an apoptotic response leading to beta cell death, by activating mitogen-activated protein kinase (MAPK) signalling cascades. Here we test the hypothesis that serine/threonine protein phosphatase 5 (PP5) acts to suppress proapoptotic c-Jun N-terminal kinase (JNK) signalling in beta cells., Methods: Ppp5c(-/-) and Ppp5c(+/+) mice were subjected to intraperitoneal glucose (IPGTT) or insulin tolerance tests. Pancreatic islets from Ppp5c(-/-) and Ppp5c(+/+) mice or MIN6 cells treated with short-interfering RNA targeting PP5 were exposed to palmitate or H(2)O(2) to activate MAPK signalling. Changes in protein phosphorylation, mRNA expression, apoptosis and insulin secretion were detected by western blot analysis, quantitative RT-PCR or ELISA., Results: Ppp5c(-/-) mice weighed less and exhibited reduced fasting glycaemia and improved glucose tolerance during IPGTT, but retained normal insulin sensitivity and islet volume. Comparison of MAPK signalling in islets from Ppp5c(-/-) mice and MIN6 cells revealed that the lack of PP5 was associated with enhanced H(2)O(2)-induced phosphorylation of JNK and c-Jun. Cells with reduced PP5 also showed enhanced JNK phosphorylation and apoptosis after palmitate treatment. PP5 suppression in MIN6 cells correlated with hypersecretion of insulin in response to glucose., Conclusions/interpretation: PP5 deficiency in mice is associated with reduced weight gain, lower fasting glycaemia, and improved glucose tolerance during IPGTT. At a molecular level, PP5 helps suppress apoptosis in beta cells by a mechanism that involves regulation of JNK phosphorylation.
- Published
- 2012
- Full Text
- View/download PDF
29. Calcineurin regulates homologous desensitization of natriuretic peptide receptor-A and inhibits ANP-induced testosterone production in MA-10 cells.
- Author
-
Henesy MB, Britain AL, Zhu B, Amable L, Honkanen RE, Corbin JD, Francis SH, and Rich TC
- Subjects
- Animals, Calcineurin genetics, Cell Line, Tumor, Computer Simulation, Cyclic GMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 5 metabolism, Genes, Reporter, Humans, Kinetics, Male, Mice, Models, Theoretical, NFATC Transcription Factors genetics, Phosphoprotein Phosphatases metabolism, Phosphorylation, RNA Interference, Signal Transduction drug effects, Atrial Natriuretic Factor pharmacology, Calcineurin metabolism, Receptors, Atrial Natriuretic Factor metabolism, Testosterone biosynthesis
- Abstract
Receptor desensitization is a ubiquitous regulatory mechanism that defines the activatable pool of receptors, and thus, the ability of cells to respond to environmental stimuli. In recent years, the molecular mechanisms controlling the desensitization of a variety of receptors have been established. However, little is known about the molecular mechanisms that underlie desensitization of natriuretic peptide receptors, including natriuretic peptide receptor-A (NPR-A). Here we report that calcineurin (protein phosphatase 2B, PP2B, PPP3C) regulates homologous desensitization of NPR-A in murine Leydig tumor (MA-10) cells. We demonstrate that both pharmacological inhibition of calcineurin activity and siRNA-mediated suppression of calcineurin expression potentiate atrial natriuretic peptide (ANP)-induced cGMP synthesis. Treatment of MA-10 cells with inhibitors of other phosphoprotein phosphatases had little or no effect on ANP-induced cGMP accumulation. In addition, overexpression of calcineurin blunts ANP-induced cGMP synthesis. We also present data indicating that the inhibition of calcineurin potentiates ANP-induced testosterone production. To better understand the contribution of calcineurin in the regulation of NPR-A activity, we examined the kinetics of ANP-induced cGMP signals. We observed transient ANP-induced cGMP signals, even in the presence of phosphodiesterase inhibitors. Inhibition of both calcineurin and phosphodiesterase dramatically slowed the decay in the response. These observations are consistent with a model in which calcineurin mediated dephosphorylation and desensitization of NPR-A is associated with significant inhibition of cGMP synthesis. PDE activity hydrolyzes cGMP, thus lowering intracellular cGMP toward the basal level. Taken together, these data suggest that calcineurin plays a previously unrecognized role in the desensitization of NPR-A and, thereby, inhibits ANP-mediated increases in testosterone production.
- Published
- 2012
- Full Text
- View/download PDF
30. Disruption of serine/threonine protein phosphatase 5 (PP5:PPP5c) in mice reveals a novel role for PP5 in the regulation of ultraviolet light-induced phosphorylation of serine/threonine protein kinase Chk1 (CHEK1).
- Author
-
Amable L, Grankvist N, Largen JW, Ortsäter H, Sjöholm Å, and Honkanen RE
- Subjects
- Actins genetics, Alleles, Animals, Blastocyst metabolism, Breeding, Cell Line, Checkpoint Kinase 1, Female, Fibroblasts drug effects, Fibroblasts metabolism, Fibroblasts radiation effects, HeLa Cells, Humans, Hydroxyurea pharmacology, Integrases genetics, Male, Mice, Nuclear Proteins metabolism, Phenotype, Phosphoprotein Phosphatases metabolism, Phosphorylation drug effects, Phosphorylation genetics, Phosphorylation radiation effects, Promoter Regions, Genetic genetics, Protein Kinases chemistry, Receptors, Neuropeptide Y metabolism, Serine metabolism, Tumor Suppressor Protein p53 metabolism, cdc25 Phosphatases metabolism, Nuclear Proteins deficiency, Nuclear Proteins genetics, Phosphoprotein Phosphatases deficiency, Phosphoprotein Phosphatases genetics, Protein Kinases metabolism, Ultraviolet Rays
- Abstract
PP5 is a ubiquitously expressed Ser/Thr protein phosphatase. High levels of PP5 have been observed in human cancers, and constitutive PP5 overexpression aids tumor progression in mouse models of tumor development. However, PP5 is highly conserved among species, and the roles of PP5 in normal tissues are not clear. Here, to help evaluate the biological actions of PP5, a Cre/loxP-conditional mouse line was generated. In marked contrast to the early embryonic lethality associated with the genetic disruption of other PPP family phosphatases (e.g. PP2A and PP4), intercrosses with mouse lines that ubiquitously express Cre recombinase starting early in development (e.g. MeuCre40 and ACTB-Cre) produced viable and fertile PP5-deficient mice. Phenotypic differences caused by the total disruption of PP5 were minor, suggesting that small molecule inhibitors of PP5 will not have widespread systemic toxicity. Examination of roles for PP5 in fibroblasts generated from PP5-deficient embryos (PP5(-/-) mouse embryonic fibroblasts) confirmed some known roles and identified new actions for PP5. PP5(-/-) mouse embryonic fibroblasts demonstrated increased sensitivity to UV light, hydroxyurea, and camptothecin, which are known activators of ATR (ataxia-telangiectasia and Rad3-related) kinase. Further study revealed a previously unrecognized role for PP5 downstream of ATR activation in a UV light-induced response. The genetic disruption of PP5 is associated with enhanced and prolonged phosphorylation of a single serine (Ser-345) on Chk1, increased phosphorylation of the p53 tumor suppressor protein (p53) at serine 18, and increased p53 protein levels. A comparable role for PP5 in the regulation of Chk1 phosphorylation was also observed in human cells.
- Published
- 2011
- Full Text
- View/download PDF
31. Cardioprotection by mild hypothermia during ischemia involves preservation of ERK activity.
- Author
-
Yang X, Liu Y, Yang XM, Hu F, Cui L, Swingle MR, Honkanen RE, Soltani P, Tissier R, Cohen MV, and Downey JM
- Subjects
- Animals, Blotting, Western, Enzyme Inhibitors pharmacology, Myocardial Ischemia prevention & control, Rabbits, Signal Transduction drug effects, Extracellular Signal-Regulated MAP Kinases metabolism, Hypothermia, Induced, Myocardial Ischemia metabolism, Signal Transduction physiology
- Abstract
Cooling the ischemic heart by just a few degrees protects it from infarction without affecting its mechanical function, but the mechanism of this protection is unknown. We investigated whether signal transduction pathways might be involved in the anti-infarct effect of mild hypothermia (35°C). Isolated rabbit hearts underwent 30 min of coronary artery occlusion/2 h of reperfusion. They were either maintained at 38.5°C or cooled to 35°C just before and only during ischemia. Infarct size was measured. The effects of the protein kinase C inhibitor chelerythrine, the nitric oxide synthase inhibitor N (ω)-nitro-L: -arginine methyl ester (L: -NAME), the phosphatidylinositol 3-kinase antagonist wortmannin, or either of the mitogen-activated protein kinase kinase 1/2 (MEK1/2) inhibitors PD98059 or U0126 on cooling's protection were examined. Myocardial ATP assays were performed and the level of phosphorylation of extracellular signal-regulated kinase (ERK) and MEK was examined by western blotting. To investigate an effect of cooling on protein phosphatase (PPase), a PPase inhibitor cantharidin was tested in the infarct model and the effect of mild hypothermia on PP2A activity in vitro was measured. Infarct size was 34.4 ± 2.2% of the ischemic zone in normothermic (38.5°C) hearts, but only 15.6 ± 8.7% in hearts cooled to 35°C during ischemia. Mechanical function was unaffected. Neither chelerythrine, L: -NAME, nor wortmannin had any effect, but both PD98059 and U0126 completely eliminated protection. Ischemia rather than reperfusion was the critical time when ERK had to be active to realize protection. Phosphorylation of ERK and MEK fell during normothermic ischemia, but during hypothermic ischemia phosphorylation of ERK remained high while that of MEK was increased. Cooling only slightly delayed the rate at which ATP fell during ischemia, and ERK inhibition did not affect that attenuation suggesting ATP preservation was unrelated to protection. Cantharidin, like cooling, also protected during ischemia but not at reperfusion, and its protection was dependent on ERK phosphorylation. However, mild hypothermia had a negligible effect on PP2A activity in an in vitro assay. Hence, mild hypothermia preserves ERK and MEK activity during ischemia which somehow protects the heart. While a PPase inhibitor mimicked cooling's protection, a direct effect of cooling on PP2A could not be demonstrated.
- Published
- 2011
- Full Text
- View/download PDF
32. Modulation of protein phosphatase 2A activity alters androgen-independent growth of prostate cancer cells: therapeutic implications.
- Author
-
Bhardwaj A, Singh S, Srivastava SK, Honkanen RE, Reed E, and Singh AP
- Subjects
- Apoptosis drug effects, Cell Cycle drug effects, Cell Cycle genetics, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Enzyme Activation drug effects, Extracellular Signal-Regulated MAP Kinases metabolism, Gene Expression Regulation, Neoplastic drug effects, Humans, Male, Prostatic Neoplasms enzymology, Protein Phosphatase 2 antagonists & inhibitors, Protein Phosphatase 2 genetics, Proto-Oncogene Proteins c-akt metabolism, RNA, Small Interfering pharmacology, Receptors, Androgen genetics, Receptors, Androgen metabolism, Signal Transduction drug effects, Androgens metabolism, Enzyme Inhibitors pharmacology, Protein Phosphatase 2 metabolism
- Abstract
Earlier we identified PPP2CA, which encodes for the α-isoform of protein phosphatase 2A (PP2A) catalytic subunit, as one of the downregulated genes in androgen-independent prostate cancer. PP2A is a serine/threonine phosphatase and a potent tumor suppressor involved in broad cellular functions; however, its role in prostate cancer has not yet been determined. Here, we have investigated the effect of PP2A activity modulation on the androgen-independent growth of prostate cancer cells. Our data show that the PPP2CA expression and PP2A activity is downregulated in androgen-independent (C4-2) prostate cancer cells as compared with androgen-dependent (LNCaP) cells. Downregulation of PP2A activity by pharmacologic inhibition or short interfering RNA-mediated PPP2CA silencing sustains the growth of LNCaP cells under an androgen-deprived condition by relieving the androgen deprivation-induced cell-cycle arrest and preventing apoptosis. Immunoblot analyses reveal enhanced phosphorylation of Akt, extracellular signal-regulated kinase (ERK), BAD, increased expression of cyclins (A1/D1), and decreased expression of cyclin inhibitor (p27) on PP2A downregulation. Furthermore, our data show that androgen receptor (AR) signaling is partially maintained in PP2A-inhibited cells through increased AR expression and ligand-independent phosphorylation. Pharmacologic inhibition of Akt, ERK, and AR suggest a role of these signaling pathways in facilitating the androgen-independent growth of LNCaP cells. These observations are supported by the effect of ceramide, a PP2A activator, on androgen-independent C4-2 cells. Ceramide inhibited the growth of C4-2 cells on androgen deprivation, an effect that could be abrogated by PP2A downregulation. Altogether, our findings suggest that modulation of PP2A activity may represent an alternative therapeutic approach for the treatment of advanced androgen-independent prostate cancer.
- Published
- 2011
- Full Text
- View/download PDF
33. Label-free quantitative proteomics and SAINT analysis enable interactome mapping for the human Ser/Thr protein phosphatase 5.
- Author
-
Skarra DV, Goudreault M, Choi H, Mullin M, Nesvizhskii AI, Gingras AC, and Honkanen RE
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Cell Cycle Proteins metabolism, Cell Line, Chaperonins metabolism, HSP90 Heat-Shock Proteins metabolism, Heat-Shock Proteins metabolism, Humans, Mass Spectrometry, Mutation, Nuclear Proteins metabolism, Phosphoprotein Phosphatases metabolism, Protein Binding, Proteomics, Nuclear Proteins analysis, Phosphoprotein Phosphatases analysis, Protein Interaction Mapping
- Abstract
Affinity purification coupled to mass spectrometry (AP-MS) represents a powerful and proven approach for the analysis of protein-protein interactions. However, the detection of true interactions for proteins that are commonly considered background contaminants is currently a limitation of AP-MS. Here using spectral counts and the new statistical tool, Significance Analysis of INTeractome (SAINT), true interaction between the serine/threonine protein phosphatase 5 (PP5) and a chaperonin, heat shock protein 90 (Hsp90), is discerned. Furthermore, we report and validate a new interaction between PP5 and an Hsp90 adaptor protein, stress-induced phosphoprotein 1 (STIP1; HOP). Mutation of PP5, replacing key basic amino acids (K97A and R101A) in the tetratricopeptide repeat (TPR) region known to be necessary for the interactions with Hsp90, abolished both the known interaction of PP5 with cell division cycle 37 homolog and the novel interaction of PP5 with stress-induced phosphoprotein 1. Taken together, the results presented demonstrate the usefulness of label-free quantitative proteomics and statistical tools to discriminate between noise and true interactions, even for proteins normally considered as background contaminants., (Copyright © 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2011
- Full Text
- View/download PDF
34. Total synthesis and evaluation of phostriecin and key structural analogues.
- Author
-
Burke CP, Swingle MR, Honkanen RE, and Boger DL
- Subjects
- Humans, Lactones chemistry, Pyrones, Stereoisomerism, Alkenes chemistry, Alkynes chemistry, Antineoplastic Agents chemical synthesis, Antineoplastic Agents chemistry, Lactones chemical synthesis, Organophosphates chemical synthesis, Organophosphates chemistry, Protein Phosphatase 2 antagonists & inhibitors, Protein Phosphatase 2 chemistry
- Abstract
Full details of the total synthesis of phostriecin (2), the assignment of its relative and absolute stereochemistry, and the resultant structural reassignment of the natural product previously represented as sultriecin (1), a phosphate versus sulfate monoester, are detailed. Studies with authentic material confirmed that phostriecin, but not sultriecin, is an effective and selective inhibitor of protein phosphatase 2A (PP2A) defining a mechanism of action responsible for its antitumor activity. The extension of the studies to the synthesis and evaluation of a series of key synthetic analogues is disclosed that highlights the importance of the natural product phosphate monoester (vs sulfate or free alcohol, both inactive and >250-fold), the α,β-unsaturated lactone (12-fold), and the hydrophobic Z,Z,E-triene tail (C12-C22, ca. 200-fold) including the unique importance of its unsaturation (50-fold, and no longer PP2A selective).
- Published
- 2010
- Full Text
- View/download PDF
35. Structure-activity relationship studies of fostriecin, cytostatin, and key analogs, with PP1, PP2A, PP5, and( beta12-beta13)-chimeras (PP1/PP2A and PP5/PP2A), provide further insight into the inhibitory actions of fostriecin family inhibitors.
- Author
-
Swingle MR, Amable L, Lawhorn BG, Buck SB, Burke CP, Ratti P, Fischer KL, Boger DL, and Honkanen RE
- Subjects
- Alkenes chemistry, Alkenes metabolism, Amino Acid Sequence, Animals, Catalytic Domain drug effects, Catalytic Domain genetics, Cattle, Enzyme Inhibitors metabolism, Molecular Sequence Data, Mutagenesis, Site-Directed, Mutant Chimeric Proteins genetics, Mutant Chimeric Proteins metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism, Organophosphates chemistry, Phosphoprotein Phosphatases genetics, Phosphoprotein Phosphatases metabolism, Polyenes, Protein Binding drug effects, Protein Phosphatase 1 genetics, Protein Phosphatase 1 metabolism, Protein Phosphatase 2 genetics, Protein Phosphatase 2 metabolism, Protein Structure, Tertiary drug effects, Pyrones chemistry, Pyrones metabolism, Rabbits, Structure-Activity Relationship, Alkenes pharmacology, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Mutant Chimeric Proteins antagonists & inhibitors, Nuclear Proteins antagonists & inhibitors, Organophosphates pharmacology, Phosphoprotein Phosphatases antagonists & inhibitors, Protein Phosphatase 1 antagonists & inhibitors, Protein Phosphatase 2 antagonists & inhibitors, Pyrones pharmacology
- Abstract
Fostriecin and cytostatin are structurally related natural inhibitors of serine/threonine phosphatases, with promising antitumor activity. The total synthesis of these antitumor agents has enabled the production of structural analogs, which are useful to explore the biological significance of features contained in the parent compounds. Here, the inhibitory activity of fostriecin, cytostatin, and 10 key structural analogs were tested in side-by-side phosphatase assays to further characterize their inhibitory activity against PP1c (Ser/Thr protein phosphatase 1 catalytic subunit), PP2Ac (Ser/Thr protein phosphatase 2A catalytic subunit), PP5c (Ser/Thr protein phosphatase 5 catalytic subunit), and chimeras of PP1 (Ser/Thr protein phosphatase 1) and PP5 (Ser/Thr protein phosphatase 5), in which key residues predicted for inhibitor contact with PP2A (Ser/Thr protein phosphatase 2A) were introduced into PP1 and PP5 using site-directed mutagenesis. The data confirm the importance of the C9-phosphate and C11-alcohol for general inhibition and further demonstrate the importance of a predicted C3 interaction with a unique cysteine (Cys(269)) in the beta12-beta13 loop of PP2A. The data also indicate that additional features beyond the unsaturated lactone contribute to inhibitory potency and selectivity. Notably, a derivative of fostriecin lacking the entire lactone subunit demonstrated marked potency and selectivity for PP2A, while having substantially reduced and similar activity against PP1 and PP1/PP2A- PP5/PP2A-chimeras that have greatly increased sensitivity to both fostriecin and cytostatin. This suggests that other features [e.g., the (Z,Z,E)-triene] also contribute to inhibitory selectivity. When considered together with previous data, these studies suggest that, despite the high structural conservation of the catalytic site in PP1, PP2A and PP5, the development of highly selective catalytic inhibitors should be feasible.
- Published
- 2009
- Full Text
- View/download PDF
36. Human DNA polymerase eta activity and translocation is regulated by phosphorylation.
- Author
-
Chen YW, Cleaver JE, Hatahet Z, Honkanen RE, Chang JY, Yen Y, and Chou KM
- Subjects
- Ataxia Telangiectasia Mutated Proteins, Caffeine pharmacology, Cell Cycle Proteins genetics, Cell Line, DNA Damage, DNA-Directed DNA Polymerase genetics, Humans, Mutation, Missense, Phosphorylation radiation effects, Protein Kinase C, Protein Serine-Threonine Kinases genetics, RNA, Small Interfering pharmacology, Ultraviolet Rays, DNA Replication, DNA-Directed DNA Polymerase metabolism
- Abstract
Human DNA polymerase eta (pol eta) can replicate across UV-induced pyrimidine dimers, and defects in the gene encoding pol eta result in a syndrome called xeroderma pigmentosum variant (XP-V). XP-V patients are prone to the development of cancer in sun-exposed areas, and cells derived from XP-V patients demonstrate increased sensitivity to UV radiation and a higher mutation rate compared with wild-type cells. pol eta has been shown to replicate across a wide spectrum of DNA lesions introduced by environmental or chemotherapeutic agents, or during nucleotide starvation, suggesting that the biological roles for pol eta are not limited to repair of UV-damaged DNA. The high error rate of pol eta requires that its intracellular activity be tightly regulated. Here, we show that the phosphorylation of pol eta increased after UV irradiation, and that treatment with caffeine, siRNA against ATR, or an inhibitor of PKC (calphostin C), reduced the accumulation of pol eta at stalled replication forks after UV irradiation or treatment with cisplatin and gemcitabine. Site-specific mutagenesis (S587A and T617A) of pol eta at two putative PKC phosphorylation sites located in the protein-protein interaction domain prevented nuclear foci formation induced by UV irradiation or treatment with gemcitabine/cisplatin. In addition, XP-V cell lines stably expressing either the S587A or T617A mutant form of pol eta were more sensitive to UV radiation and gemcitabine/cisplatin than control cells expressing wild-type pol eta. These results suggest that phosphorylation is one mechanism by which the cellular activity of pol eta is regulated.
- Published
- 2008
- Full Text
- View/download PDF
37. The role of serine/threonine protein phosphatase type 5 (PP5) in the regulation of stress-induced signaling networks and cancer.
- Author
-
Golden T, Swingle M, and Honkanen RE
- Subjects
- Animals, Humans, Nuclear Proteins chemistry, Phosphoprotein Phosphatases chemistry, Protein Structure, Quaternary, Cell Transformation, Neoplastic, Neoplasms enzymology, Nuclear Proteins physiology, Phosphoprotein Phosphatases physiology, Signal Transduction physiology, Stress, Physiological enzymology
- Abstract
Although the aberrant actions of protein kinases have long been known to contribute to tumor promotion and carcinogenesis, roles for protein phosphatases in the development of human cancer have only emerged in the last decade. In this review, we discuss the data obtained from studies examining the biological and pathological roles of a serine/threonine protein phosphatase, PP5, which suggest that PP5 is a potentially important regulator of both hormone- and stress-induced signaling networks that enable a cell to respond appropriately to genomic stress.
- Published
- 2008
- Full Text
- View/download PDF
38. Elevated levels of Ser/Thr protein phosphatase 5 (PP5) in human breast cancer.
- Author
-
Golden T, Aragon IV, Rutland B, Tucker JA, Shevde LA, Samant RS, Zhou G, Amable L, Skarra D, and Honkanen RE
- Subjects
- Animals, Cell Death, Cell Line, Tumor, Doxorubicin pharmacology, Humans, Mice, Mice, Nude, Nuclear Proteins, Oxidative Stress, Phosphoprotein Phosphatases, Threonine, Time Factors, Ultraviolet Rays, Up-Regulation, Vinblastine pharmacology, Breast Neoplasms enzymology, Carcinoma, Ductal, Breast enzymology, Carcinoma, Intraductal, Noninfiltrating enzymology
- Abstract
Ser/Thr protein phosphatase 5 (PP5) regulates several signaling-cascades that suppress growth and/or facilitate apoptosis in response to genomic stress. The expression of PP5 is responsive to hypoxia inducible factor-1 (HIF-1) and estrogen, which have both been linked to the progression of human breast cancer. Still, it is not clear if PP5 plays a role in the development of human cancer. Here, immunostaining of breast cancer tissue-microarrays (TMAs) revealed a positive correlation between PP5 over-expression and ductal carcinoma in situ (DCIS; P value 0.0028), invasive ductal carcinoma (IDC; P value 0.012) and IDC with metastases at the time of diagnosis (P value 0.0001). In a mouse xenograft model, the constitutive over-expression of PP5 was associated with an increase in the rate of tumor growth. In a MCF-7 cell culture model over-expression correlated with both an increase in the rate of proliferation and protection from cell death induced by oxidative stress, UVC-irradiation, adriamycin, and vinblastine. PP5 over-expression had no apparent effect on the sensitivity of MCF-7 cells to taxol or rapamycin. Western analysis of extracts from cells over-expressing PP5 revealed a decrease in the phosphorylation of known substrates for PP5. Together, these studies indicate that elevated levels of PP5 protein occur in human breast cancer and suggest that PP5 over-expression may aid tumor progression.
- Published
- 2008
- Full Text
- View/download PDF
39. High yield expression of serine/threonine protein phosphatase type 5, and a fluorescent assay suitable for use in the detection of catalytic inhibitors.
- Author
-
Ni L, Swingle MS, Bourgeois AC, and Honkanen RE
- Subjects
- 4-Nitrophenylphosphatase metabolism, Catalysis, Enzyme Inhibitors chemistry, Escherichia coli metabolism, Fluorescent Dyes, Humans, Hymecromone analogs & derivatives, Hymecromone chemistry, Indicators and Reagents, Nuclear Proteins isolation & purification, Phosphoprotein Phosphatases isolation & purification, Spectrometry, Fluorescence, Enzyme Inhibitors pharmacology, Nuclear Proteins antagonists & inhibitors, Nuclear Proteins biosynthesis, Phosphoprotein Phosphatases antagonists & inhibitors, Phosphoprotein Phosphatases biosynthesis
- Abstract
Protein phosphatase type 5 (PP5) belongs to the PPP family of serine/threonine protein phosphatases and is expressed in most, if not all, human tissues. Although the physiological roles played by PP5 are not yet clear, PP5 is found in association with several proteins that influence intracellular signaling networks initiated by hormones (i.e., glucocorticoids) or cellular stress (i.e., hypoxia, oxidative stress). Recently, studies conducted with short interfering RNA and antisense oligonucleotides indicate that PP5 plays an important role in the regulation of stress-induced signaling cascades that influence both cell growth and the onset of apoptosis. Therefore, the identification of small molecule inhibitors of PP5 is desired for use in studies to further define the biological/pathological roles of PP5. Such inhibitors may also prove useful for development into novel antitumor agents. Here we describe methods to express and purify large amounts of biologically active PP5c, an inhibitor titration-based assay to determine the amount of PP5 in solution, and a fluorescent phosphatase assay that can be used to screen chemical libraries and natural extracts for the presence of catalytic inhibitors.
- Published
- 2007
- Full Text
- View/download PDF
40. Small-molecule inhibitors of ser/thr protein phosphatases: specificity, use and common forms of abuse.
- Author
-
Swingle M, Ni L, and Honkanen RE
- Subjects
- Cantharidin pharmacology, Furans pharmacology, Indole Alkaloids pharmacology, Lactones pharmacology, Lipids pharmacology, Marine Toxins, Microcystins pharmacology, Okadaic Acid pharmacology, Organophosphates pharmacology, Organophosphorus Compounds pharmacology, Oxazoles pharmacology, Peptides, Cyclic pharmacology, Pyrans pharmacology, Pyrones pharmacology, Spiro Compounds pharmacology, Enzyme Inhibitors pharmacology, Phosphoprotein Phosphatases antagonists & inhibitors
- Abstract
Natural product extracts have proven to be a rich source of small molecules that potently inhibit the catalytic activity of certain PPP-family ser/thr protein phosphatases. To date, the list of inhibitors includes okadaic acid (produced by marine dinoflagelates, Prorocentrum sp. and Dinophysis sp.), calyculin A, dragmacidins (isolated from marine sponges), microcystins, nodularins (cyanobacteria, Microcystis sp. and Nodularia sp.), tautomycin, tautomycetin, cytostatins, phospholine, leustroducsins, phoslactomycins, fostriecin (soil bacteria, Streptomyces sp.), and cantharidin (blister beetles, approx 1500 species). Many of these compounds share structural similarities, and several have become readily available for research purposes. Here we will review the specificity of available inhibitors and present methods for their use in studying sensitive phosphatases. Common mistakes in the employment of these compounds will also be addressed briefly, notably the widespread misconception that they only inhibit the activity of PP1 and PP2A. Inhibitors of PP2B (calcineurin) will only be mentioned in passing, except to state that, in our hands, cypermethrin, deltamethrin, and fenvalerate, which are sold as potent inhibitors of PP2B, do not inhibit the catalytic activity of PP2B.
- Published
- 2007
- Full Text
- View/download PDF
41. Total synthesis and evaluation of cytostatin, its C10-C11 diastereomers, and additional key analogues: impact on PP2A inhibition.
- Author
-
Lawhorn BG, Boga SB, Wolkenberg SE, Colby DA, Gauss CM, Swingle MR, Amable L, Honkanen RE, and Boger DL
- Subjects
- Alkenes pharmacology, Animals, Cell Proliferation drug effects, Drug Screening Assays, Antitumor, Enzyme Inhibitors chemistry, Humans, Leukemia L1210, Mice, Models, Molecular, Molecular Conformation, Organophosphates chemistry, Polyenes, Pyrones chemistry, Stereoisomerism, Structure-Activity Relationship, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors pharmacology, Organophosphates chemical synthesis, Organophosphates pharmacology, Phosphoprotein Phosphatases antagonists & inhibitors, Pyrones chemical synthesis, Pyrones pharmacology
- Abstract
The total synthesis of cytostatin, an antitumor agent belonging to the fostriecin family of natural products, is described in full detail. The convergent approach relied on a key epoxide-opening reaction to join the two stereotriad units and a single-step late-stage stereoselective installation of the sensitive (Z,Z,E)-triene through a beta-chelation-controlled nucleophilic addition. The synthetic route provided rapid access to the C4-C6 stereoisomers of the cytostatin lactone, which were prepared and used to define the C4-C6 relative stereochemistry of the natural product. In addition to the natural product, each of the C10-C11 diastereomers of cytostatin was divergently prepared (11 steps from key convergence step) by this route and used to unequivocally confirm the relative and absolute stereochemistry of cytostatin. Each of the cytostatin diastereomers exhibited a reduced activity toward inhibition of PP2A (>100-fold), demonstrating the importance of the presence and stereochemistry of the C10-methyl and C11-hydroxy groups for potent PP2A inhibition. Extensions of the studies provided dephosphocytostatin, sulfocytostatin (a key analogue related to the natural product sultriecin), 11-deshydroxycytostatin, and an analogue lacking the entire C12-C18 (Z,Z,E)-triene segment, which were used to define the magnitude of the C9-phosphate (>4000-fold), C11-alcohol (250-fold), and triene (220-fold) contribution to PP2A inhibition. A model of cytostatin bound to the active site of PP2A is presented, compared to that of fostriecin, which is also presented in detail for the first time, and used to provide insights into the role of the key substituents. Notably, the alpha,beta unsaturated lactone of cytostatin, like that of fostriecin, is projected to serve as a key electrophile, providing a covalent adduct with Cys269 unique to PP2A, contributing to its potency (> or =200-fold for fostriecin) and accounting for its selectivity.
- Published
- 2006
- Full Text
- View/download PDF
42. Cantharidin-induced mitotic arrest is associated with the formation of aberrant mitotic spindles and lagging chromosomes resulting, in part, from the suppression of PP2Aalpha.
- Author
-
Bonness K, Aragon IV, Rutland B, Ofori-Acquah S, Dean NM, and Honkanen RE
- Subjects
- Apoptosis drug effects, Cantharidin toxicity, Cell Cycle, Chromosomes, Human drug effects, Dose-Response Relationship, Drug, Fluorescent Antibody Technique, HeLa Cells, Humans, Oligodeoxyribonucleotides, Antisense metabolism, Oligonucleotides pharmacology, Phosphoprotein Phosphatases metabolism, Protein Phosphatase 2, RNA, Small Interfering metabolism, Time Factors, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Cantharidin pharmacology, Mitosis drug effects, Phosphoprotein Phosphatases antagonists & inhibitors, Spindle Apparatus drug effects
- Abstract
Cantharidin, a natural vesicant, inhibits the activity of several PPP family phosphatases, displays antitumor activity, and induces apoptosis in many types of tumor cells. However, the molecular mechanisms underlying the antitumor activity of cantharidin are not clear. Here, dose-response studies confirm a strong correlation between the suppression of phosphatase activity and cell death. Flow cytometry analysis indicates that before apoptosis, cantharidin delays cell cycle progression following DNA replication with no apparent effect on G(1)-S or S-G(2) phase progression. In contrast, studies with double thymidine-synchronized populations of cells indicate that cantharidin can rapidly arrest growth when added during G(2) or early M phase. Immunostaining indicates that cell cycle arrest occurs before the completion of mitosis and is associated with the appearance of aberrant mitotic spindles. Live cell imaging with time-lapse microscopy shows that cantharidin disrupts the metaphase alignment of chromosomes and produces a prolonged mitotic arrest, with the onset of apoptosis occurring before the onset of anaphase. To explore the contribution of individual phosphatases, antisense oligonucleotides and small interfering RNA were developed to suppress the expression of cantharidin-sensitive phosphatases. The suppression of PP2Aalpha, but not PP2Abeta, is sufficient to induce metaphase arrest, during which time lagging chromosomes are observed moving between the spindle poles and the metaphase plate. Immunostaining revealed slightly abnormal, yet predominately bipolar, mitotic spindles. Nonetheless, after a 10- to 15-hour delay, the cells enter anaphase, suggesting that an additional cantharidin-sensitive phosphatase is involved in the progression from metaphase into anaphase or to prevent the onset of apoptosis in cells arrested during mitosis.
- Published
- 2006
- Full Text
- View/download PDF
43. Glutamate inhibits protein phosphatases and promotes insulin exocytosis in pancreatic beta-cells.
- Author
-
Lehtihet M, Webb DL, Honkanen RE, and Sjöholm A
- Subjects
- Animals, Cell Line, Dose-Response Relationship, Drug, Exocytosis drug effects, Insulin Secretion, Islets of Langerhans drug effects, Okadaic Acid pharmacology, Rats, Calcium metabolism, Exocytosis physiology, Glucose pharmacology, Glutamic Acid pharmacology, Insulin metabolism, Islets of Langerhans metabolism, Phosphoprotein Phosphatases antagonists & inhibitors, Phosphoprotein Phosphatases metabolism
- Abstract
In human type 2 diabetes mellitus, loss of glucose-sensitive insulin secretion from the pancreatic beta-cell is an early pathogenetic event, but the mechanisms involved in glucose sensing are poorly understood. A messenger role has been postulated for L-glutamate in linking glucose stimulation to sustained insulin exocytosis in the beta-cell, but the precise nature by which L-glutamate controls insulin secretion remains elusive. Effects of L-glutamate on the activities of ser/thr protein phosphatases (PPase) and Ca(2+)-regulated insulin exocytosis in INS-1E cells were investigated. Glucose increases L-glutamate contents and promotes insulin secretion from INS-1E cells. L-glutamate also dose-dependently inhibits PPase enzyme activities analogous to the specific PPase inhibitor, okadaic acid. L-glutamate and okadaic acid directly and non-additively promote insulin exocytosis from permeabilized INS-1E cells in a Ca(2+)-independent manner. Thus, an increase in phosphorylation state, through inhibition of protein dephosphorylation by glucose-derived L-glutamate, may be a novel regulatory mechanism linking glucose sensing to sustained insulin exocytosis.
- Published
- 2005
- Full Text
- View/download PDF
44. Constitutive over expression of serine/threonine protein phosphatase 5 (PP5) augments estrogen-dependent tumor growth in mice.
- Author
-
Golden T, Aragon IV, Zhou G, Cooper SR, Dean NM, and Honkanen RE
- Subjects
- Animals, Cell Division drug effects, Humans, Male, Mice, Mice, Nude, Operator Regions, Genetic, Plasmids, Promoter Regions, Genetic, Protein Synthesis Inhibitors pharmacology, Tetracycline pharmacology, Trans-Activators, Transplantation, Heterologous, Tumor Cells, Cultured, Breast Neoplasms, Male enzymology, Breast Neoplasms, Male pathology, Estradiol pharmacology, Nuclear Proteins metabolism, Phosphoprotein Phosphatases metabolism
- Abstract
Serine/threonine protein phosphatase 5 (PP5) appears to play an underappreciated role in the regulation of cellular proliferation. In estrogen-responsive cells, PP5 expression is stimulated by 17 beta-estradiol, and in a variety of p53 wild-type tumor cells the suppression of PP5 expression with ISIS 15534 inhibits growth. To further explore the relationship between PP5 and the development of human cancer, here we tested the effect of elevated PP5 expression on tumor growth using a mouse xenograph model and a stable MCF-7 cell line in which the expression of wild-type PP5 was placed under the control of tetracycline-off regulated transactivator and operator plasmids. In the xenograph model a modest two fold increase in PP5 protein levels significantly enhanced the growth rate of estrogen-dependent tumors, suggesting PP5 plays a positive role in tumor development.
- Published
- 2004
- Full Text
- View/download PDF
45. Ser/Thr protein phosphatase 5 inactivates hypoxia-induced activation of an apoptosis signal-regulating kinase 1/MKK-4/JNK signaling cascade.
- Author
-
Zhou G, Golden T, Aragon IV, and Honkanen RE
- Subjects
- Base Sequence, Blotting, Western, Cell Line, Cell Line, Tumor, Enzyme Activation, Genes, Reporter, Humans, Hypoxia, Luciferases metabolism, Microcystins, Models, Biological, Molecular Sequence Data, Nuclear Proteins metabolism, Oxygen metabolism, Peptides, Cyclic pharmacology, Phosphoprotein Phosphatases metabolism, Phosphorylation, Promoter Regions, Genetic, Protein Binding, Protein Structure, Tertiary, RNA, Double-Stranded metabolism, RNA, Small Interfering metabolism, Reverse Transcriptase Polymerase Chain Reaction, Sepharose metabolism, Sequence Homology, Nucleic Acid, Signal Transduction, Threonine metabolism, Time Factors, Transcriptional Activation, Apoptosis, JNK Mitogen-Activated Protein Kinases metabolism, MAP Kinase Kinase 4 metabolism, MAP Kinase Kinase Kinase 5 metabolism, Mitogen-Activated Protein Kinase Kinases metabolism, Nuclear Proteins physiology, Phosphoprotein Phosphatases physiology
- Abstract
Mitogen-activated protein kinase (MAPK) signaling cascades are multifunctional signaling networks that influence cell growth, differentiation, apoptosis, and cellular responses to stress. Since the activation/propagation of MAPK signaling requires the sequential phosphorylation of many downstream proteins, the phosphatases that dephosphorylate MAPKs represent critical elements in the control of MAPK-signaling networks. Here we show that hypoxia induces a transient increase in the activity of apoptosis signal-regulating kinase 1 (ASK-1), a MAPKKK that responds to oxidative stress by triggering cascades leading to the phosphorylation/activation of c-Jun N-terminal kinases (JNK) and p38-MAPK. Hypoxia-induced ASK-1/MKK-4/JNK signaling is suppressed by serine/threonine protein phosphatase type 5 (PP5), which acts to turn off ASK-1/MKK-4/JNK signaling via two mechanisms. First, in a rapid response hypoxia facilitates the association of endogenous PP5 with ASK-1. PP5 binds to the C-terminal domain of ASK-1, and studies with siRNA targeting PP5 indicate that PP5 acts to suppress the phosphorylation of MKK4 (Thr-261), JNK (Thr-183/Tyr-185), and c-Jun (Ser-63) without affecting the activating phosphorylation of p38 MAPK (Thr-180/Tyr-182), p44/p42-MAPK/ERK1/2 (Thr-202/Tyr-204), or c-Jun protein levels. If hypoxia is prolonged, the expression of PP5 is increased due to the activation of a transcriptional activator, which was identified as hypoxia-inducible factor-1. Together, these studies indicate that PP5 plays an important role in the survival of cells in a low oxygen environment by suppressing a hypoxia-induced ASK-1/MKK4/JNK signaling cascade that promotes an apoptotic response.
- Published
- 2004
- Full Text
- View/download PDF
46. Structural basis for the catalytic activity of human serine/threonine protein phosphatase-5.
- Author
-
Swingle MR, Honkanen RE, and Ciszak EM
- Subjects
- Amino Acid Sequence, Binding Sites, Catalysis, Chemical Phenomena, Chemistry, Physical, Conserved Sequence, Crystallization, Crystallography, X-Ray, Gene Expression, Humans, Hydrolysis, Models, Molecular, Molecular Structure, Nuclear Proteins genetics, Peptide Fragments chemistry, Peptide Fragments genetics, Phosphoprotein Phosphatases genetics, Recombinant Fusion Proteins, Sequence Alignment, Static Electricity, Structure-Activity Relationship, Nuclear Proteins chemistry, Nuclear Proteins metabolism, Phosphoprotein Phosphatases chemistry, Phosphoprotein Phosphatases metabolism
- Abstract
Serine/threonine protein phosphatase-5 (PP5) affects many signaling networks that regulate cell growth and cellular responses to stress. Here we report the crystal structure of the PP5 catalytic domain (PP5c) at a resolution of 1.6 A. From this structure we propose a mechanism for PP5-mediated hydrolysis of phosphoprotein substrates, which requires the precise positioning of two metal ions within a conserved Asp271-M1:M2-W1-His427-His304-Asp274 catalytic motif (where M1 and M2 are metals and W1 is a water molecule). The structure of PP5c provides a structural basis for explaining the exceptional catalytic proficiency of protein phosphatases, which are among the most powerful known catalysts. Resolution of the entire C terminus revealed a novel subdomain, and the structure of the PP5c should also aid development of type-specific inhibitors.
- Published
- 2004
- Full Text
- View/download PDF
47. Inositol hexakisphosphate and sulfonylureas regulate beta-cell protein phosphatases.
- Author
-
Lehtihet M, Honkanen RE, and Sjöholm A
- Subjects
- Animals, Cell Line, Tumor, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacology, Glucose metabolism, Insulin metabolism, Insulin Secretion, Marine Toxins, Muscles metabolism, Okadaic Acid metabolism, Oxazoles metabolism, Phosphoprotein Phosphatases metabolism, Phosphorylation, Protein Phosphatase 1, Rabbits, Rats, Signal Transduction, Phytic Acid metabolism, Sulfonylurea Compounds metabolism
- Abstract
In human type 2 diabetes, loss of glucose-stimulated insulin exocytosis from the pancreatic beta-cell is an early pathogenetic event. Mechanisms controlling insulin exocytosis are, however, not fully understood. We show here that inositol hexakisphosphate (InsP(6)), whose concentration transiently increases upon glucose stimulation, dose-dependently and differentially inhibits enzyme activities of ser/thr protein phosphatases in physiologically relevant concentrations. None of the hypoglycemic sulfonylureas tested affected protein phosphatase-1 or -2A activity at clinically relevant concentrations in these cells. Thus, an increase in cellular phosphorylation state, through inhibition of protein dephosphorylation by InsP(6), may be a novel regulatory mechanism linking glucose-stimulated polyphosphoinositide formation to insulin exocytosis in insulin-secreting cells.
- Published
- 2004
- Full Text
- View/download PDF
48. Fundamental role of the fostriecin unsaturated lactone and implications for selective protein phosphatase inhibition.
- Author
-
Buck SB, Hardouin C, Ichikawa S, Soenen DR, Gauss CM, Hwang I, Swingle MR, Bonness KM, Honkanen RE, and Boger DL
- Subjects
- Alkenes chemistry, Amino Acid Sequence, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Lactones chemistry, Lactones pharmacology, Molecular Sequence Data, Polyenes, Pyrones, Structure-Activity Relationship, Alkenes pharmacology, Phosphoprotein Phosphatases antagonists & inhibitors
- Abstract
Key derivatives and analogues of fostriecin were prepared and examined that revealed a fundamental role for the unsaturated lactone and confirmed the essential nature of the phosphate monoester. Thus, an identical 200-fold reduction in protein phosphatase 2A (PP2A) inhibition is observed with either the saturated lactone (7) or with an analogue that lacks the entire lactone (15). This 200-fold increase in PP2A inhibition attributable to the unsaturated lactone potentially may be due to reversible C269 alkylation within the PP beta12-beta13 active site loop accounting for PP2A/4 potency and selectivity.
- Published
- 2003
- Full Text
- View/download PDF
49. Identification of a functional link for the p53 tumor suppressor protein in dexamethasone-induced growth suppression.
- Author
-
Urban G, Golden T, Aragon IV, Cowsert L, Cooper SR, Dean NM, and Honkanen RE
- Subjects
- Antineoplastic Agents, Hormonal pharmacology, Blotting, Northern, Blotting, Western, Cell Division, Cell Separation, Cyclin-Dependent Kinase Inhibitor p21, Cyclins metabolism, Dose-Response Relationship, Drug, Flow Cytometry, G1 Phase, Humans, Luciferases metabolism, Nuclear Proteins metabolism, Oligonucleotides, Antisense pharmacology, Phosphoprotein Phosphatases metabolism, Phosphorylation, RNA, Messenger metabolism, Resting Phase, Cell Cycle, Serine chemistry, Time Factors, Tumor Cells, Cultured, Tumor Suppressor Protein p53 metabolism, Dexamethasone pharmacology, Nuclear Proteins physiology, Phosphoprotein Phosphatases physiology, Tumor Suppressor Protein p53 physiology
- Abstract
Serine/threonine phosphatase 5 (PP5) can act as a suppresser of p53-dependent growth suppression and has been reported to associate with several proteins, including the glucocorticoid receptor/heat-shock protein-90 complex. Still, the physiological/pathological roles of PP5 are unclear. To characterize the relationship of PP5, glucocorticoid receptor activation and p53, here we describe the development of chimeric antisense oligonucleotides that potently inhibit human p53 expression. This allowed us to regulate the expression of either p53 (e.g. with ISIS 110332) or PP5 (e.g. with ISIS 15534) in genetically identical cells. Studies with ISIS 110332 revealed that the suppression of p53 expression is associated with a decrease in the basal expression of the cyclin-dependent kinase inhibitor protein, p21(WAF1/Cip1), and a concomitant increase in the rate of cell proliferation. Suppression of p53 also blocks dexamethasone-induced p21(WAF1/Cip1) expression and G(1)-growth arrest. Furthermore, treatment with ISIS 110332, but not the mismatched controls, ablates the suppression of growth produced by prior treatment with dexamethasone. Additional studies revealed that dexamethasone-dependent p21(WAF1/Cip1) expression occurs without an apparent change in p53 protein levels or the phosphorylation status of p53 at Ser-6, -37, or -392. However, dexamethasone treatment is associated with an increase in p53 phosphorylation at Ser-15. Suppression of PP5 expression with ISIS 15534 also results in the hyperphosphorylation of p53 at Ser-15. Together, these findings indicate that the basal expression of p53 plays a functional role in a glucocorticoid receptor-mediated response regulating the expression of p21(Waf1/Cip1) via a mechanism that is suppressed by PP5 and associated with the phosphorylation of p53 at Ser-15.
- Published
- 2003
- Full Text
- View/download PDF
50. Regulating the expression of protein phosphatase type 5.
- Author
-
Golden TA and Honkanen RE
- Subjects
- Animals, Base Sequence, Binding Sites, Conserved Sequence, DNA Primers, Isoenzymes genetics, Mammals, Marine Toxins, Microcystins, Nuclear Proteins metabolism, Okadaic Acid pharmacology, Oligonucleotides, Antisense, Oxazoles pharmacology, Peptides, Cyclic pharmacology, Phosphoprotein Phosphatases metabolism, RNA, Antisense genetics, RNA, Messenger genetics, Recombinant Proteins metabolism, Transfection methods, Gene Expression Regulation, Enzymologic genetics, Nuclear Proteins genetics, Phosphoprotein Phosphatases genetics
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.