1
|
Hassanein F, Fadel HH, Shehata AI, Hamdy NA, Masoud IM. In silico study to explore the mechanism of Toxoplasma-induced inflammation and target therapy based on sero and salivary Toxoplasma. Sci Rep 2024; 14:13600. [PMID: 38866852 PMCID: PMC11169245 DOI: 10.1038/s41598-024-63735-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/31/2024] [Indexed: 06/14/2024] Open
Abstract
We aimed to assess salivary and seroprevalence of Toxoplasma immunoglobulins in risky populations and evaluate drug docking targeting TgERP. A cross-sectional study was conducted in Alexandria University hospitals' outpatient clinics. 192 participants were enrolled from September 2022 to November 2023. Anti-Toxoplasma IgG and IgM were determined in serum and saliva by ELISA. An in-Silico study examined TgERP's protein-protein interactions (PPIs) with pro-inflammatory cytokine receptors, anti-inflammatory cytokine, cell cycle progression regulatory proteins, a proliferation marker, and nuclear envelope integrity-related protein Lamin B1. Our findings revealed that anti-T. gondii IgG were detected in serum (66.1%) and saliva (54.7%), with 2.1% of both samples were positive for IgM. Salivary IgG had 75.59% sensitivity, 86.15% specificity, 91.40% PPV, 64.40% NPP, 79.17% accuracy and fair agreement with serum IgG. On the other hand, the sensitivity, specificity, PPV, NPV, and accuracy in detecting salivary IgM were 75.0%, 99.47%, 75.0%, 99.47%, and 98.96%. AUC 0.859 indicates good discriminatory power. Examined synthetic drugs and natural products can target specific amino acids residues of TgERP that lie at the same binding interface with LB1 and Ki67, subsequently, hindering their interaction. Hence, salivary samples can be a promising diagnostic approach. The studied drugs can counteract the pro-inflammatory action of TgERP.
Collapse
Affiliation(s)
- Faika Hassanein
- Department of Microbiology & Immunology, Faculty of Dentistry, Pharos University in Alexandria, Alexandria, Egypt.
| | - Hewida H Fadel
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria, Egypt
| | - Amany I Shehata
- Department of Tropical Health, High Institute of Public Health, Alexandria University, Alexandria, Egypt
| | - Noha Alaa Hamdy
- Department of Clinical Pharmacy & Pharmacy Practice, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Inas M Masoud
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| |
Collapse
|
2
|
Phosphorylated Peptide Derived from the Myosin Phosphatase Target Subunit Is a Novel Inhibitor of Protein Phosphatase-1. Int J Mol Sci 2023; 24:ijms24054789. [PMID: 36902219 PMCID: PMC10003451 DOI: 10.3390/ijms24054789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/20/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Identification of specific protein phosphatase-1 (PP1) inhibitors is of special importance regarding the study of its cellular functions and may have therapeutic values in diseases coupled to signaling processes. In this study, we prove that a phosphorylated peptide of the inhibitory region of myosin phosphatase (MP) target subunit (MYPT1), R690QSRRS(pT696)QGVTL701 (P-Thr696-MYPT1690-701), interacts with and inhibits the PP1 catalytic subunit (PP1c, IC50 = 3.84 µM) and the MP holoenzyme (Flag-MYPT1-PP1c, IC50 = 3.84 µM). Saturation transfer difference NMR measurements established binding of hydrophobic and basic regions of P-Thr696-MYPT1690-701 to PP1c, suggesting interactions with the hydrophobic and acidic substrate binding grooves. P-Thr696-MYPT1690-701 was dephosphorylated by PP1c slowly (t1/2 = 81.6-87.9 min), which was further impeded (t1/2 = 103 min) in the presence of the phosphorylated 20 kDa myosin light chain (P-MLC20). In contrast, P-Thr696-MYPT1690-701 (10-500 µM) slowed down the dephosphorylation of P-MLC20 (t1/2 = 1.69 min) significantly (t1/2 = 2.49-10.06 min). These data are compatible with an unfair competition mechanism between the inhibitory phosphopeptide and the phosphosubstrate. Docking simulations of the PP1c-P-MYPT1690-701 complexes with phosphothreonine (PP1c-P-Thr696-MYPT1690-701) or phosphoserine (PP1c-P-Ser696-MYPT1690-701) suggested their distinct poses on the surface of PP1c. In addition, the arrangements and distances of the surrounding coordinating residues of PP1c around the phosphothreonine or phosphoserine at the active site were distinct, which may account for their different hydrolysis rate. It is presumed that P-Thr696-MYPT1690-701 binds tightly at the active center but the phosphoester hydrolysis is less preferable compared to P-Ser696-MYPT1690-701 or phosphoserine substrates. Moreover, the inhibitory phosphopeptide may serve as a template to synthesize cell permeable PP1-specific peptide inhibitors.
Collapse
|
3
|
Kokot T, Köhn M. Emerging insights into serine/threonine-specific phosphoprotein phosphatase function and selectivity. J Cell Sci 2022; 135:277104. [DOI: 10.1242/jcs.259618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
ABSTRACT
Protein phosphorylation on serine and threonine residues is a widely distributed post-translational modification on proteins that acts to regulate their function. Phosphoprotein phosphatases (PPPs) contribute significantly to a plethora of cellular functions through the accurate dephosphorylation of phosphorylated residues. Most PPPs accomplish their purpose through the formation of complex holoenzymes composed of a catalytic subunit with various regulatory subunits. PPP holoenzymes then bind and dephosphorylate substrates in a highly specific manner. Despite the high prevalence of PPPs and their important role for cellular function, their mechanisms of action in the cell are still not well understood. Nevertheless, substantial experimental advancements in (phospho-)proteomics, structural and computational biology have contributed significantly to a better understanding of PPP biology in recent years. This Review focuses on recent approaches and provides an overview of substantial new insights into the complex mechanism of PPP holoenzyme regulation and substrate selectivity.
Collapse
Affiliation(s)
- Thomas Kokot
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg 1 , Freiburg 79104 , Germany
- University of Freiburg, 2 Faculty of Biology , Freiburg 79104 , Germany
| | - Maja Köhn
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg 1 , Freiburg 79104 , Germany
- University of Freiburg, 2 Faculty of Biology , Freiburg 79104 , Germany
| |
Collapse
|
4
|
Louzao MC, Vilariño N, Vale C, Costas C, Cao A, Raposo-Garcia S, Vieytes MR, Botana LM. Current Trends and New Challenges in Marine Phycotoxins. Mar Drugs 2022; 20:md20030198. [PMID: 35323497 PMCID: PMC8950113 DOI: 10.3390/md20030198] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 02/04/2023] Open
Abstract
Marine phycotoxins are a multiplicity of bioactive compounds which are produced by microalgae and bioaccumulate in the marine food web. Phycotoxins affect the ecosystem, pose a threat to human health, and have important economic effects on aquaculture and tourism worldwide. However, human health and food safety have been the primary concerns when considering the impacts of phycotoxins. Phycotoxins toxicity information, often used to set regulatory limits for these toxins in shellfish, lacks traceability of toxicity values highlighting the need for predefined toxicological criteria. Toxicity data together with adequate detection methods for monitoring procedures are crucial to protect human health. However, despite technological advances, there are still methodological uncertainties and high demand for universal phycotoxin detectors. This review focuses on these topics, including uncertainties of climate change, providing an overview of the current information as well as future perspectives.
Collapse
Affiliation(s)
- Maria Carmen Louzao
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
- Correspondence: (M.C.L.); (L.M.B.)
| | - Natalia Vilariño
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Carmen Vale
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Celia Costas
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Alejandro Cao
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Sandra Raposo-Garcia
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Mercedes R. Vieytes
- Departamento de Fisiologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain;
| | - Luis M. Botana
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
- Correspondence: (M.C.L.); (L.M.B.)
| |
Collapse
|
5
|
Synthesis, functional proteomics and biological evaluation of new 5-pyrazolyl ureas as potential anti-angiogenic compounds. Eur J Med Chem 2021; 226:113872. [PMID: 34600191 DOI: 10.1016/j.ejmech.2021.113872] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/09/2021] [Accepted: 09/24/2021] [Indexed: 11/21/2022]
Abstract
Based on biological results of previous synthesized pyrazolyl ureas able to interfere with angiogenesis process, we planned and synthesized the new benzyl-urea derivatives 2-4; some of them showed an interesting anti-proliferative profile and particularly 4e potently inhibited HUVEC proliferation. To shed light on the mechanism of action of 4e, its interactome has been deeply inspected to identify the most prominent protein partners, mainly taking into account kinome and phosphatome, through drug affinity responsive target stability experiments, followed by targeted limited proteolysis analysis. From these studies, PP1γ emerged as the most reliable 4e potential target in HUVEC. Molecular docking simulations on PP1γ were carried out to predict 4e binding mode. To assess its potential anti-angiogenic effect, 4e was tested in vitro to verify interference on kinase and phosphate activities. Overall, our results evidenced for 4e an interesting anti-angiogenic action, probably due to its action at intracellular level on PP1γ signalling pathways.
Collapse
|
6
|
Hu Z, Wang X, Zhang S, Zong W. Research on the discrepant inhibition mechanism of microcystin-LR disinfectant by-products target to protein phosphatase 1. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:45586-45595. [PMID: 33871773 DOI: 10.1007/s11356-021-12472-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/11/2021] [Indexed: 06/12/2023]
Abstract
The secondary contamination for microcystin disinfection by-products (MC-DBPs) is of concern due to the residual toxic structure similar to their original toxins. To evaluate the toxicity of MC-DBPs, the discrepant inhibition mechanisms target to protein phosphatase 1 (PP1) were evaluated. Five typical MCLR-DBPs related to the oxidation of Adda5 were identified as C49H75N10O13Cl (+1Cl1OH, P1/P2), C34H54N10O12 (+2OH, P3/P4), and C49H76N10O14 (P5). Toxicity inhibition experiment on PP1 showed that the toxicity was in the sequence of MCLR > P3 > P1 > P4 > P2 > P5. Base on MOE molecular simulation, the discrepant inhibition mechanisms for MCLR and MCLR-DBPs target to PP1 were further clarified. The combination of MCLR/MCLR-DBPs to PP1 was mainly restrained by residues Adda5 and Arg4. Above key sites promoted the binding of MCLR/MCLR-DBPs to PP1 through the hydrogen bonds (H2O ← Adda5, Tyr134 → Adda5, H2O ← Arg4, Tyr134 → Arg4, Glu275 ← Arg4), ionic bonds (Asp197-Adda5, Glu275-Arg4, Asp220 → Arg4), and H-pi bonds (Trp206 ↔ Adda5, Ser129 ↔ Adda5). The oxidation of Adda5 also affected Mdha7 participated ionic bond Glu275-Mdha7 and Glu6 participated hydrogen bond H2O → Glu6. Besides, the "integral hydrogen bonds and ionic bonds" between toxin and PP1 also had important effects on the toxin toxicity. In this way, the inhibition of "Adda5 destroyed" MC-DBPs target to PP1 was regulated.
Collapse
Affiliation(s)
- Zhengxin Hu
- College of Geography and Environment, Shandong Normal University, 1# Daxue Road, Jinan, 250358, Shandong, China
| | - Xiaoning Wang
- College of Geography and Environment, Shandong Normal University, 1# Daxue Road, Jinan, 250358, Shandong, China
| | - Shuhan Zhang
- College of Geography and Environment, Shandong Normal University, 1# Daxue Road, Jinan, 250358, Shandong, China
| | - Wansong Zong
- College of Geography and Environment, Shandong Normal University, 1# Daxue Road, Jinan, 250358, Shandong, China.
| |
Collapse
|
7
|
Arman T, Clarke JD. Microcystin Toxicokinetics, Molecular Toxicology, and Pathophysiology in Preclinical Rodent Models and Humans. Toxins (Basel) 2021; 13:toxins13080537. [PMID: 34437407 PMCID: PMC8402503 DOI: 10.3390/toxins13080537] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 02/06/2023] Open
Abstract
Microcystins are ubiquitous toxins produced by photoautotrophic cyanobacteria. Human exposures to microcystins occur through the consumption of contaminated drinking water, fish and shellfish, vegetables, and algal dietary supplements and through recreational activities. Microcystin-leucine-arginine (MCLR) is the prototypical microcystin because it is reported to be the most common and toxic variant and is the only microcystin with an established tolerable daily intake of 0.04 µg/kg. Microcystin toxicokinetics is characterized by low intestinal absorption, rapid and specific distribution to the liver, moderate metabolism to glutathione and cysteinyl conjugates, and low urinary and fecal excretion. Molecular toxicology involves covalent binding to and inhibition of protein phosphatases, oxidative stress, cell death (autophagy, apoptosis, necrosis), and cytoskeleton disruption. These molecular and cellular effects are interconnected and are commonly observed together. The main target organs for microcystin toxicity are the intestine, liver, and kidney. Preclinical data indicate microcystins may also have nervous, pulmonary, cardiac, and reproductive system toxicities. Recent evidence suggests that exposure to other hepatotoxic insults could potentiate microcystin toxicity and increase the risk for chronic diseases. This review summarizes the current knowledge for microcystin toxicokinetics, molecular toxicology, and pathophysiology in preclinical rodent models and humans. More research is needed to better understand human toxicokinetics and how multifactorial exposures contribute to disease pathogenesis and progression.
Collapse
|
8
|
Wilkins AL, Rundberget T, Sandvik M, Rise F, Knudsen BK, Kilcoyne J, Reguera B, Rial P, Wright EJ, Giddings SD, Boundy MJ, Rafuse C, Miles CO. Identification of 24- O-β-d-Glycosides and 7-Deoxy-Analogues of Okadaic Acid and Dinophysistoxin-1 and -2 in Extracts from Dinophysis Blooms, Dinophysis and Prorocentrum Cultures, and Shellfish in Europe, North America and Australasia. Toxins (Basel) 2021; 13:toxins13080510. [PMID: 34437381 PMCID: PMC8402559 DOI: 10.3390/toxins13080510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 11/25/2022] Open
Abstract
Two high-mass polar compounds were observed in aqueous side-fractions from the purification of okadaic acid (1) and dinophysistoxin-2 (2) from Dinophysis blooms in Spain and Norway. These were isolated and shown to be 24-O-β-d-glucosides of 1 and 2 (4 and 5, respectively) by nuclear magnetic resonance (NMR) spectroscopy, mass spectrometry, and enzymatic hydrolysis. These, together with standards of 1, 2, dinophysistoxin-1 (3), and a synthetic specimen of 7-deoxy-1 (7), combined with an understanding of their mass spectrometric fragmentation patterns, were then used to identify 1–5, the 24-O-β-d-glucoside of dinophysistoxin-1 (6), 7, 7-deoxy-2 (8), and 7-deoxy-3 (9) in a range of extracts from Dinophysis blooms, Dinophysis cultures, and contaminated shellfish from Spain, Norway, Ireland, Canada, and New Zealand. A range of Prorocentrum lima cultures was also examined by liquid chromatography–high resolution tandem mass spectrometry (LC–HRMS/MS) and was found to contain 1, 3, 7, and 9. However, although 4–6 were not detected in these cultures, low levels of putative glycosides with the same exact masses as 4 and 6 were present. The potential implications of these findings for the toxicology, metabolism, and biosynthesis of the okadaic acid group of marine biotoxins are briefly discussed.
Collapse
Affiliation(s)
- Alistair L. Wilkins
- Norwegian Veterinary Institute, P.O. Box 64, NO-1431 Ås, Norway; (A.L.W.); (T.R.); (M.S.)
- School of Science and Engineering, University of Waikato, Private Bag 3105, Hamilton 3240, New Zealand;
| | - Thomas Rundberget
- Norwegian Veterinary Institute, P.O. Box 64, NO-1431 Ås, Norway; (A.L.W.); (T.R.); (M.S.)
- Norwegian Institute for Water Research, Gaustadalléen 21, NO-0349 Oslo, Norway
| | - Morten Sandvik
- Norwegian Veterinary Institute, P.O. Box 64, NO-1431 Ås, Norway; (A.L.W.); (T.R.); (M.S.)
| | - Frode Rise
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern, NO-0315 Oslo, Norway;
| | - Brent K. Knudsen
- School of Science and Engineering, University of Waikato, Private Bag 3105, Hamilton 3240, New Zealand;
| | - Jane Kilcoyne
- Marine Institute, Rinville, Oranmore, County Galway H91 R673, Ireland;
| | - Beatriz Reguera
- Centro Oceanográfico de Vigo (IEO, CSIC), Subida a Radio Faro 50, 36390 Vigo, Spain; (B.R.); (P.R.)
| | - Pilar Rial
- Centro Oceanográfico de Vigo (IEO, CSIC), Subida a Radio Faro 50, 36390 Vigo, Spain; (B.R.); (P.R.)
| | - Elliott J. Wright
- Biotoxin Metrology, National Research Council, 1411 Oxford St., Halifax, NS B3H 3Z1, Canada; (E.J.W.); (S.D.G.); (C.R.)
| | - Sabrina D. Giddings
- Biotoxin Metrology, National Research Council, 1411 Oxford St., Halifax, NS B3H 3Z1, Canada; (E.J.W.); (S.D.G.); (C.R.)
| | - Michael J. Boundy
- Cawthron Institute, 98 Halifax Street East, Nelson 7010, New Zealand;
| | - Cheryl Rafuse
- Biotoxin Metrology, National Research Council, 1411 Oxford St., Halifax, NS B3H 3Z1, Canada; (E.J.W.); (S.D.G.); (C.R.)
| | - Christopher O. Miles
- Norwegian Veterinary Institute, P.O. Box 64, NO-1431 Ås, Norway; (A.L.W.); (T.R.); (M.S.)
- Biotoxin Metrology, National Research Council, 1411 Oxford St., Halifax, NS B3H 3Z1, Canada; (E.J.W.); (S.D.G.); (C.R.)
- Correspondence:
| |
Collapse
|
9
|
Zhang Q, Fan Z, Zhang L, You Q, Wang L. Strategies for Targeting Serine/Threonine Protein Phosphatases with Small Molecules in Cancer. J Med Chem 2021; 64:8916-8938. [PMID: 34156850 DOI: 10.1021/acs.jmedchem.1c00631] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Among numerous posttranslational regulation patterns, phosphorylation is reversibly controlled by the balance of kinases and phosphatases. The major form of cellular signaling involves the reversible phosphorylation of proteins on tyrosine, serine, or threonine residues. However, altered phosphorylation levels are found in diverse diseases, including cancer, making kinases and phosphatases ideal drug targets. In contrast to the success of prosperous kinase inhibitors, design of small molecules targeting phosphatase is struggling due to past bias and difficulty. This is especially true for serine/threonine phosphatases, one of the largest phosphatase families. From this perspective, we aim to provide insights into serine/threonine phosphatases and the small molecules targeting these proteins for drug development, especially in cancer. Through highlighting the modulation strategies, we aim to provide basic principles for the design of small molecules and future perspectives for the application of drugs targeting serine/threonine phosphatases.
Collapse
Affiliation(s)
- Qiuyue Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhongjiao Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lianshan Zhang
- Shanghai Hengrui Pharmaceutical Co., Ltd., Shanghai 200245, China
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
10
|
Regulation of Cardiac PKA Signaling by cAMP and Oxidants. Antioxidants (Basel) 2021; 10:antiox10050663. [PMID: 33923287 PMCID: PMC8146537 DOI: 10.3390/antiox10050663] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/31/2022] Open
Abstract
Pathologies, such as cancer, inflammatory and cardiac diseases are commonly associated with long-term increased production and release of reactive oxygen species referred to as oxidative stress. Thereby, protein oxidation conveys protein dysfunction and contributes to disease progression. Importantly, trials to scavenge oxidants by systemic antioxidant therapy failed. This observation supports the notion that oxidants are indispensable physiological signaling molecules that induce oxidative post-translational modifications in target proteins. In cardiac myocytes, the main driver of cardiac contractility is the activation of the β-adrenoceptor-signaling cascade leading to increased cellular cAMP production and activation of its main effector, the cAMP-dependent protein kinase (PKA). PKA-mediated phosphorylation of substrate proteins that are involved in excitation-contraction coupling are responsible for the observed positive inotropic and lusitropic effects. PKA-actions are counteracted by cellular protein phosphatases (PP) that dephosphorylate substrate proteins and thus allow the termination of PKA-signaling. Both, kinase and phosphatase are redox-sensitive and susceptible to oxidation on critical cysteine residues. Thereby, oxidation of the regulatory PKA and PP subunits is considered to regulate subcellular kinase and phosphatase localization, while intradisulfide formation of the catalytic subunits negatively impacts on catalytic activity with direct consequences on substrate (de)phosphorylation and cardiac contractile function. This review article attempts to incorporate the current perception of the functionally relevant regulation of cardiac contractility by classical cAMP-dependent signaling with the contribution of oxidant modification.
Collapse
|
11
|
Wang Y, Rao D, Wu X, Zhang Q, Wu S. Aptamer-based microcantilever-array biosensor for ultra-sensitive and rapid detection of okadaic acid. Microchem J 2021. [DOI: 10.1016/j.microc.2020.105644] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
12
|
Kerk D, Valdés-Tresanco ME, Toth R, Noskov SY, Ng KKS, Moorhead GB. Origin of the Phosphoprotein Phosphatase (PPP) sequence family in Bacteria: Critical ancestral sequence changes, radiation patterns and substrate binding features. BBA ADVANCES 2021; 1:100005. [PMID: 37082010 PMCID: PMC10074919 DOI: 10.1016/j.bbadva.2021.100005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Phosphoprotein phosphatases (PPP) belong to the PPP Sequence family, which in turn belongs to the broader metallophosphoesterase (MPE) superfamily. The relationship between the PPP Sequence family and other members of the MPE superfamily remains unresolved, in particular what transitions took place in an ancestral MPE to ultimately produce the phosphoprotein specific phosphatases (PPPs). Methods We use structural and sequence alignment data, phylogenetic tree analysis, sequence signature (Weblogo) analysis, in silico protein-peptide modeling data, and in silico mutagenesis to trace a likely route of evolution from MPEs to the PPP Sequence family. Hidden Markov Model (HMM) based iterative database search strategies were utilized to identify PPP Sequence Family members from numerous bacterial groups. Results Using Mre11 as proxy for an ancestral nuclease-like MPE we trace a possible evolutionary route that alters a single active site substrate binding His-residue to yield a new substrate binding accessory, the "2-Arg-Clamp". The 2-Arg-Clamp is not found in MPEs, but is present in all PPP Sequence family members, where the phosphomonesterase reaction predominates. Variation in position of the clamp arginines and a supplemental sequence loop likely provide substrate specificity for each PPP Sequence family group. Conclusions Loss of a key substrate binding His-in MPEs opened the path to bind novel substrates and evolution of the 2-Arg-Clamp, a sequence change seen in both bacterial and eukaryotic phosphoprotein phosphatases.General significance: We establish a likely evolutionary route from nuclease-like MPE to PPP Sequence family enzymes, that includes the phosphoprotein phosphatases.
Collapse
|
13
|
Khan R, Kulasiri D, Samarasinghe S. Functional repertoire of protein kinases and phosphatases in synaptic plasticity and associated neurological disorders. Neural Regen Res 2021; 16:1150-1157. [PMID: 33269764 PMCID: PMC8224123 DOI: 10.4103/1673-5374.300331] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Protein phosphorylation and dephosphorylation are two essential and vital cellular mechanisms that regulate many receptors and enzymes through kinases and phosphatases. Ca2+- dependent kinases and phosphatases are responsible for controlling neuronal processing; balance is achieved through opposition. During molecular mechanisms of learning and memory, kinases generally modulate positively while phosphatases modulate negatively. This review outlines some of the critical physiological and structural aspects of kinases and phosphatases involved in maintaining postsynaptic structural plasticity. It also explores the link between neuronal disorders and the deregulation of phosphatases and kinases.
Collapse
Affiliation(s)
- Raheel Khan
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University; Department of Molecular Biosciences, Lincoln University, Christchurch, New Zealand
| | - Don Kulasiri
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University; Department of Molecular Biosciences, Lincoln University, Christchurch, New Zealand
| | - Sandhya Samarasinghe
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University, Christchurch, New Zealand
| |
Collapse
|
14
|
Ren L, Hu Z, Wang Q, Du Y, Zong W. Regulation Efficacy and Mechanism of the Toxicity of Microcystin-LR Targeting Protein Phosphatase 1 via the Biodegradation Pathway. Toxins (Basel) 2020; 12:toxins12120790. [PMID: 33322407 PMCID: PMC7764552 DOI: 10.3390/toxins12120790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 11/16/2022] Open
Abstract
Biodegradation is important to regulate the toxicity and environmental risk of microcystins (MCs). To explore their regulation effectiveness and mechanism, typical biodegradation products originating from microcystin-LR (MCLR) were prepared and purified. The protein phosphatase 1 (PP1) inhibition experiment showed the biodegradation pathway was effective in regulating the toxicity of the biodegradation products by extending the biodegradation. With the assistance of molecular docking, the specific interaction between the toxins and PP1 was explored. The MCLR/MCLR biodegradation products combined with PP1 mainly by the aid of interactions related to the active sites Adda5, Glu6, Mdha7, and the ionic bonds/hydrogen bonds between the integral toxin and PP1. As a consequence, the interactions between Mn22+ and Asp64/Asp92 in the catalytic center were inhibited to varying degrees, resulting in the reduced toxicity of the biodegradation products. During the biodegradation process, the relevant key interactions might be weakened or even disappear, and thus the toxicity was regulated. It is worth noting that the secondary pollution of the partial products (especially for Adda5-Glu6-Mdha7-Ala1 and the linearized MCLR), which still possessed the major active sites, is of deep concern.
Collapse
|
15
|
Pan L, Chen J, He X, Zhan T, Shen H. Aqueous photodegradation of okadaic acid and dinophysistoxin-1: Persistence, kinetics, photoproducts, pathways, and toxicity evaluation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 743:140593. [PMID: 32673911 DOI: 10.1016/j.scitotenv.2020.140593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 06/11/2023]
Abstract
Diarrhetic shellfish poisoning (DSP) toxins are a class of natural organic contaminants that pose a serious threat not only to marine ecosystems and fisheries but also to human health. They are widely distributed in coastal and offshore waters around the world. However, the persistence and photochemical degradation characteristics of DSP in an aqueous environment are still unclear. This study aimed to elucidate the photochemical fate of two representative DSP toxins, namely, okadaic acid (OA) and dinophysistoxin-1 (DTX1). Results showed that photo-mediated chemical reactions play a crucial role in eliminating DSP toxins in seawater. However, the degradation of OA and DTX1 was relatively slow under natural solar radiation, with a removal efficiency of 90.0% after exposure for more than 20 days. When the reaction solutions of OA and DTX1 were exposed to Hg lamp radiation, their degradation followed pseudo-first-order kinetics, and was remarkably influenced by seawater pH and metal-ion concentration. A total of 24 tentative transformation products (TPs) of OA and DTX1 were identified via liquid chromatography high-resolution mass spectrometry. C12 (C43H66O11) and C24 (C44H68O11) were the main TPs. The following possible photodegradation pathways were proposed: decarboxylation, photoinduced hydrolysis, chain scission, and photo-oxidation. Toxicity assays via protein phosphatase 2A inhibition proved that photochemical processes could significantly reduce the DSP toxicity of irradiated solutions by approximately 88%. This work provides an enhanced understanding of the fate of DSP toxins in the aqueous environment, allowing for an improved assessment of their environmental impacts.
Collapse
Affiliation(s)
- Lei Pan
- Marine Bioresource and Environment Research Center, Key Laboratory of Marine Eco-Environmental Science and Technology, The First Institute of Oceanography, Ministry of Natural Resources, Qingdao 266061, China; College of Chemistry and Molecular Engineering, Qingdao Technology University of Shandong, Qingdao 266042, China
| | - Junhui Chen
- Marine Bioresource and Environment Research Center, Key Laboratory of Marine Eco-Environmental Science and Technology, The First Institute of Oceanography, Ministry of Natural Resources, Qingdao 266061, China; Laboratory for Marine Ecology and Environmental Science, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, China.
| | - Xiuping He
- Marine Bioresource and Environment Research Center, Key Laboratory of Marine Eco-Environmental Science and Technology, The First Institute of Oceanography, Ministry of Natural Resources, Qingdao 266061, China; Laboratory for Marine Ecology and Environmental Science, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, China
| | - Tianrong Zhan
- College of Chemistry and Molecular Engineering, Qingdao Technology University of Shandong, Qingdao 266042, China
| | - Huihui Shen
- Marine Bioresource and Environment Research Center, Key Laboratory of Marine Eco-Environmental Science and Technology, The First Institute of Oceanography, Ministry of Natural Resources, Qingdao 266061, China; Laboratory for Marine Ecology and Environmental Science, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, China
| |
Collapse
|
16
|
Manda JN, Butler BB, Aponick A. Synthesis and Biological Evaluation of the Southern Hemisphere of Spirastrellolide A and Analogues. J Org Chem 2020; 85:13694-13709. [PMID: 33111529 DOI: 10.1021/acs.joc.0c01867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The synthesis and biological evaluation of truncated spirastrellolide A analogues comprised of the southern hemisphere against protein phosphatase 2A are described. A convergent synthesis was designed featuring two gold-catalyzed cyclization reactions, specifically, a dehydrative cyclization of monoallylic diols for the synthesis of the tetrahydropyran (A-ring) and a regioselective spiroketalization for the efficient generation of the [6,6]-spiroketal (B, C-ring system). The synthesis of the southern hemisphere of spirastrellolide A was achieved involving the longest linear sequence of 19 steps. A total of eight spirastrellolide A analogues were synthesized, and preliminary PP2A enzyme assay inhibition studies were performed for the first time on analogues of the southern hemisphere. Several analogues showed inhibition, which is a positive indication and perhaps suggests that the unsaturated spiroketal fragment might be crucial to induce PP2A inhibition.
Collapse
Affiliation(s)
- Jagadeesh Nagendra Manda
- Florida Center for Heterocyclic Compounds and Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - Barry B Butler
- Florida Center for Heterocyclic Compounds and Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - Aaron Aponick
- Florida Center for Heterocyclic Compounds and Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
17
|
Campos A, Freitas M, de Almeida AM, Martins JC, Domínguez-Pérez D, Osório H, Vasconcelos V, Reis Costa P. OMICs Approaches in Diarrhetic Shellfish Toxins Research. Toxins (Basel) 2020; 12:E493. [PMID: 32752012 PMCID: PMC7472309 DOI: 10.3390/toxins12080493] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/22/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022] Open
Abstract
Diarrhetic shellfish toxins (DSTs) are among the most prevalent marine toxins in Europe's and in other temperate coastal regions. These toxins are produced by several dinoflagellate species; however, the contamination of the marine trophic chain is often attributed to species of the genus Dinophysis. This group of toxins, constituted by okadaic acid (OA) and analogous molecules (dinophysistoxins, DTXs), are highly harmful to humans, causing severe poisoning symptoms caused by the ingestion of contaminated seafood. Knowledge on the mode of action and toxicology of OA and the chemical characterization and accumulation of DSTs in seafood species (bivalves, gastropods and crustaceans) has significantly contributed to understand the impacts of these toxins in humans. Considerable information is however missing, particularly at the molecular and metabolic levels involving toxin uptake, distribution, compartmentalization and biotransformation and the interaction of DSTs with aquatic organisms. Recent contributions to the knowledge of DSTs arise from transcriptomics and proteomics research. Indeed, OMICs constitute a research field dedicated to the systematic analysis on the organisms' metabolisms. The methodologies used in OMICs are also highly effective to identify critical metabolic pathways affecting the physiology of the organisms. In this review, we analyze the main contributions provided so far by OMICs to DSTs research and discuss the prospects of OMICs with regard to the DSTs toxicology and the significance of these toxins to public health, food safety and aquaculture.
Collapse
Affiliation(s)
- Alexandre Campos
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, 4450–208 Porto, Portugal; (M.F.); (J.C.M.); (D.D.-P.); (V.V.)
| | - Marisa Freitas
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, 4450–208 Porto, Portugal; (M.F.); (J.C.M.); (D.D.-P.); (V.V.)
- ESS-P.Porto, School of Health, Polytechnic Institute of Porto. Rua Dr. António Bernardino de Almeida, 400, 4200-072 Porto, Portugal
| | - André M. de Almeida
- LEAF-Instituto Superior de Agronomia, Universidade de Lisboa, 1349-017 Lisboa, Portugal;
| | - José Carlos Martins
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, 4450–208 Porto, Portugal; (M.F.); (J.C.M.); (D.D.-P.); (V.V.)
| | - Dany Domínguez-Pérez
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, 4450–208 Porto, Portugal; (M.F.); (J.C.M.); (D.D.-P.); (V.V.)
| | - Hugo Osório
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Ipatimup—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, 4200-135 Porto, Portugal
- Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| | - Vitor Vasconcelos
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, 4450–208 Porto, Portugal; (M.F.); (J.C.M.); (D.D.-P.); (V.V.)
- Biology Department, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Pedro Reis Costa
- IPMA—Instituto Português do Mar da Atmosfera, Rua Alfredo Magalhães Ramalho, 6, 1495-006 Lisbon, Portugal;
| |
Collapse
|
18
|
Measurement of Microcystin and Nodularin Activity in Human Urine by Immunocapture-Protein Phosphatase 2A Assay. Toxins (Basel) 2019; 11:toxins11120729. [PMID: 31847123 PMCID: PMC6950260 DOI: 10.3390/toxins11120729] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 12/14/2022] Open
Abstract
Microcystins (MC) and nodularin (NOD) are toxins released by cyanobacteria during harmful algal blooms. They are potent inhibitors of protein phosphatases 1 and 2A (PP1 and PP2A) and cause a variety of adverse symptoms in humans and animals if ingested. More than 250 chemically diverse congeners of MCs have been identified, but certified reference materials are only available for a few. A diagnostic test that does not require each reference material for detection is necessary to identify human exposures. To address this need, our lab has developed a method that uses an antibody to specifically isolate MCs and NOD from urine prior to detection via a commercially available PP2A kit. This assay quantitates the summed inhibitory activity of nearly all MCs and NOD on PP2A relative to a common MC congener, microcystin-LR (MC-LR). The quantitation range for MC-LR using this method is from 0.050–0.500 ng/mL. No background responses were detected in a convenience set of 50 individual urines. Interday and intraday % accuracies ranged from 94%–118% and relative standard deviations were 15% or less, meeting FDA guidelines for receptor binding assays. The assay detected low levels of MCs in urines from three individuals living in close proximity to harmful algal blooms (HABs) in Florida.
Collapse
|
19
|
Protein phosphatase 1 of Leishmania donovani exhibits conserved catalytic residues and pro-inflammatory response. Biochem Biophys Res Commun 2019; 516:770-776. [DOI: 10.1016/j.bbrc.2019.06.085] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 06/16/2019] [Indexed: 12/13/2022]
|
20
|
Wang X, Obeidat M, Li L, Pasarj P, Aburahess S, Holmes CFB, Ballermann BJ. TIMAP inhibits endothelial myosin light chain phosphatase by competing with MYPT1 for the catalytic protein phosphatase 1 subunit PP1cβ. J Biol Chem 2019; 294:13280-13291. [PMID: 31315927 PMCID: PMC6737228 DOI: 10.1074/jbc.ra118.006075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 06/17/2019] [Indexed: 11/06/2022] Open
Abstract
Transforming growth factor-β membrane associated protein (TIMAP) is an endothelial cell (EC)-predominant PP1 regulatory subunit and a member of the myosin phosphatase target (MYPT) protein family. The MYPTs preferentially bind the catalytic protein phosphatase 1 subunit PP1cβ, forming myosin phosphatase holoenzymes. We investigated whether TIMAP/PP1cβ could also function as a myosin phosphatase. Endogenous PP1cβ, myosin light chain 2 (MLC2), and myosin IIA heavy chain coimmunoprecipitated from EC lysates with endogenous TIMAP, and endogenous MLC2 colocalized with TIMAP in EC projections. Purified recombinant GST-TIMAP interacted directly with purified recombinant His-MLC2. However, TIMAP overexpression in EC enhanced MLC2 phosphorylation, an effect not observed with a TIMAP mutant that does not bind PP1cβ. Conversely, MLC2 phosphorylation was reduced in lung lysates from TIMAP-deficient mice and upon silencing of endogenous TIMAP expression in ECs. Ectopically expressed TIMAP slowed the rate of MLC2 dephosphorylation, an effect requiring TIMAP-PP1cβ interaction. The association of MYPT1 with PP1cβ was profoundly reduced in the presence of excess TIMAP, leading to proteasomal MYPT1 degradation. In the absence of TIMAP, MYPT1-associated PP1cβ readily bound immobilized microcystin-LR, an active-site inhibitor of PP1c. By contrast, TIMAP-associated PP1cβ did not interact with microcystin-LR, indicating that the active site of PP1cβ is blocked when it is bound to TIMAP. Thus, TIMAP inhibits myosin phosphatase activity in ECs by competing with MYPT1 for PP1cβ and blocking the PP1cβ active site.
Collapse
Affiliation(s)
- Xin Wang
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Marya Obeidat
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Laiji Li
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Phuwadet Pasarj
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Salah Aburahess
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Charles F B Holmes
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Barbara J Ballermann
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2G3, Canada.
| |
Collapse
|
21
|
Wang Q, Wang X, Zhang S, Zong W. Molecular mechanism for the discrepant inhibition of microcystins on protein phosphatase 1. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:21774-21783. [PMID: 31134550 DOI: 10.1007/s11356-019-05520-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 05/17/2019] [Indexed: 06/09/2023]
Abstract
Due to variable amino acid residues at positions 2 and 4, microcystins (MCs) had diversified variants with different toxicities. To evaluate the discrepant toxicity, the inhibition effects of five typical MC variants (with the changed amino acid residues at position 4) target to PP1 were evaluated. The inhibition sequence was verified as follows: MCLR (IC50 = 2.6 μg/L) > MCLF (IC50 = 4.4 μg/L) > MCLA (IC50 = 5.5 μg/L) > MCLY (IC50 = 7.9 μg/L) > MCLW (IC50 = 13.6 μg/L). To further clarify the inhibition mechanism for variant toxicity, the interactions between MCs and PP1 were evaluated with the assistance of MOE molecule simulation. Results show the hydrophobic interaction (Adda5 with PP1) and the hydrogen bonds (especially for Z4 → Glu275) were positively correlated with MC toxicity, while the hydrogen bonds (Leu2 ← Arg96, IsoAsp3 ← Arg96, and IsoAsp3 ← Tyr134) and the ion bonds (between Mn2+ and His173/Asn124/Asp92) were negatively correlated with toxicity. However, the hydrogen bonds (Ala1 → Glu275, Mdha7 ← Gly274, Z4 ← Arg221, and Adda5 ← His125), the covalent combination (between Mdha7 and Cys273), and the ion bonds (between Mn2+ and His248/Asp64/His66) were weakly correlated with toxicity. By further analysis, the steric hindrance and hydrophobicity introduced by different Z4 residues affected the changes for combination area and energy of MC-PP1 complexes, leading to the discrepancies in MC toxicity.
Collapse
Affiliation(s)
- Qian Wang
- College of Geography and Environment, Shandong Normal University, 88# East Wenhua Road, Jinan, 250014, Shandong, China
| | - Xiaoning Wang
- College of Geography and Environment, Shandong Normal University, 88# East Wenhua Road, Jinan, 250014, Shandong, China
| | - Shuhan Zhang
- College of Geography and Environment, Shandong Normal University, 88# East Wenhua Road, Jinan, 250014, Shandong, China
| | - Wansong Zong
- College of Geography and Environment, Shandong Normal University, 88# East Wenhua Road, Jinan, 250014, Shandong, China.
| |
Collapse
|
22
|
Brautigan DL, Shenolikar S. Protein Serine/Threonine Phosphatases: Keys to Unlocking Regulators and Substrates. Annu Rev Biochem 2019; 87:921-964. [PMID: 29925267 DOI: 10.1146/annurev-biochem-062917-012332] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protein serine/threonine phosphatases (PPPs) are ancient enzymes, with distinct types conserved across eukaryotic evolution. PPPs are segregated into types primarily on the basis of the unique interactions of PPP catalytic subunits with regulatory proteins. The resulting holoenzymes dock substrates distal to the active site to enhance specificity. This review focuses on the subunit and substrate interactions for PPP that depend on short linear motifs. Insights about these motifs from structures of holoenzymes open new opportunities for computational biology approaches to elucidate PPP networks. There is an expanding knowledge base of posttranslational modifications of PPP catalytic and regulatory subunits, as well as of their substrates, including phosphorylation, acetylation, and ubiquitination. Cross talk between these posttranslational modifications creates PPP-based signaling. Knowledge of PPP complexes, signaling clusters, as well as how PPPs communicate with each other in response to cellular signals should unlock the doors to PPP networks and signaling "clouds" that orchestrate and coordinate different aspects of cell physiology.
Collapse
Affiliation(s)
- David L Brautigan
- Center for Cell Signaling and Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA;
| | - Shirish Shenolikar
- Signature Research Programs in Cardiovascular and Metabolic Disorders and Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore 169857
| |
Collapse
|
23
|
Swingle MR, Honkanen RE. Inhibitors of Serine/Threonine Protein Phosphatases: Biochemical and Structural Studies Provide Insight for Further Development. Curr Med Chem 2019; 26:2634-2660. [PMID: 29737249 PMCID: PMC10013172 DOI: 10.2174/0929867325666180508095242] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/05/2018] [Accepted: 03/29/2018] [Indexed: 11/22/2022]
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.
Collapse
Affiliation(s)
- Mark R Swingle
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile AL 36688, United States
| | - Richard E Honkanen
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile AL 36688, United States
| |
Collapse
|
24
|
Yu J, Deng T, Xiang S. Structural basis for protein phosphatase 1 recruitment by glycogen‐targeting subunits. FEBS J 2018; 285:4646-4659. [DOI: 10.1111/febs.14699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/15/2018] [Accepted: 11/09/2018] [Indexed: 10/27/2022]
Affiliation(s)
- Jun Yu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety Shanghai Institute of Nutrition and Health Shanghai Institutes for Biological Sciences University of Chinese Academy of Sciences Chinese Academy of Sciences Shanghai China
| | - Tingting Deng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety Shanghai Institute of Nutrition and Health Shanghai Institutes for Biological Sciences University of Chinese Academy of Sciences Chinese Academy of Sciences Shanghai China
| | - Song Xiang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety Shanghai Institute of Nutrition and Health Shanghai Institutes for Biological Sciences University of Chinese Academy of Sciences Chinese Academy of Sciences Shanghai China
- Key laboratory of Immune Microenvironment and Disease (Ministry of Education) Tianjin Medical University China
| |
Collapse
|
25
|
Salvi F, Trebacz M, Kokot T, Hoermann B, Rios P, Barabas O, Kӧhn M. Effects of stably incorporated iron on protein phosphatase-1 structure and activity. FEBS Lett 2018; 592:4028-4038. [PMID: 30403291 PMCID: PMC6587554 DOI: 10.1002/1873-3468.13284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/04/2018] [Accepted: 10/30/2018] [Indexed: 01/21/2023]
Abstract
Protein phosphatase‐1 (PP1) drives a large amount of phosphoSer/Thr protein dephosphorylations in eukaryotes to counteract multiple kinases in signaling pathways. The phosphatase requires divalent metal cations for catalytic activity and contains iron naturally. Iron has been suggested to have an influence on PP1 activity through Fe2+ and Fe3+ oxidation states. However, much biochemical and all structural data have been obtained with recombinant PP1 containing Mn2+ ions. Purifying iron‐containing PP1 from Escherichia coli has thus far not been possible. Here, we present the preparation, characterization, and structure of iron‐bound PP1α in inactive and active states. We establish a key role for the electronic/redox properties of iron in PP1 activity and shed light on the difference in substrate specificity between iron‐ and manganese‐containing PP1.
Collapse
Affiliation(s)
- Francesca Salvi
- Genome Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Malgorzata Trebacz
- Faculty of Biology and Centre for Biological Signalling Studies (BIOSS)University of FreiburgGermany
| | - Thomas Kokot
- Faculty of Biology and Centre for Biological Signalling Studies (BIOSS)University of FreiburgGermany
| | - Bernhard Hoermann
- Faculty of Biology and Centre for Biological Signalling Studies (BIOSS)University of FreiburgGermany
- Faculty of BiosciencesCollaboration for Joint PhD Degree between EMBL and Heidelberg UniversityGermany
| | - Pablo Rios
- Faculty of Biology and Centre for Biological Signalling Studies (BIOSS)University of FreiburgGermany
| | - Orsolya Barabas
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Maja Kӧhn
- Genome Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
- Faculty of Biology and Centre for Biological Signalling Studies (BIOSS)University of FreiburgGermany
| |
Collapse
|
26
|
Zong W, Wang Q, Zhang S, Teng Y, Du Y. Regulation on the toxicity of microcystin-LR target to protein phosphatase 1 by biotransformation pathway: effectiveness and mechanism. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:26020-26029. [PMID: 29968216 DOI: 10.1007/s11356-018-2676-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 06/26/2018] [Indexed: 06/08/2023]
Abstract
Biotransformation was an important pathway to regulate the toxicity of microcystins (MCs) targeted to protein phosphatases (PPs). To explore the regulation effectiveness and mechanism, several typical biothiol transformation products originated from MCLR were prepared by nucleophilic addition reaction. The reduced inhibition effect of MCLR transformation products on PP1 was evaluated and compared with their original toxin. Though molecular simulation showed the introduced biothiols enhanced the total combination areas and energies for target complexes, the steric hindrance of introduced biothiols inhibited the combination between the key action sites (Mdha7 and Adda5 residues) and PP1. Furthermore, the introduced biothiols also weakened the hydrogen bonds for some key interaction sites and altered the ion bonds between PP1 and the two Mn2+ ions in the catalytic center. The discrepant regulation effect for biothiols on the toxicity of MCLR was closely related to above indexes and influenced by molecular sides.
Collapse
Affiliation(s)
- Wansong Zong
- College of Geography and Environment, Shandong Normal University, 88# East Wenhua Road, Jinan, 250014, Shandong, China.
| | - Qian Wang
- College of Geography and Environment, Shandong Normal University, 88# East Wenhua Road, Jinan, 250014, Shandong, China
| | - Shuhan Zhang
- College of Geography and Environment, Shandong Normal University, 88# East Wenhua Road, Jinan, 250014, Shandong, China
| | - Yue Teng
- School of Environmental and Civil Engineering, Jiangnan University, 1800# Lihu Avenue, Wuxi, 214122, Jiangsu, China
| | - Yonggang Du
- College of Geography and Environment, Shandong Normal University, 88# East Wenhua Road, Jinan, 250014, Shandong, China
| |
Collapse
|
27
|
Matsui T, Yamamoto K, Fujita T, Morihashi K. Molecular Dynamics and Quantum Chemical Approach for the Estimation of an Intramolecular Hydrogen Bond Strength in Okadaic Acid. J Phys Chem B 2018; 122:7233-7242. [DOI: 10.1021/acs.jpcb.8b03272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
28
|
Cruz P, Martínez Leal JF, Daranas AH, Pérez M, Cuevas C. On the Mechanism of Action of Dragmacidins I and J, Two New Representatives of a New Class of Protein Phosphatase 1 and 2A Inhibitors. ACS OMEGA 2018; 3:3760-3767. [PMID: 30023878 PMCID: PMC6045348 DOI: 10.1021/acsomega.7b01786] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/21/2018] [Indexed: 06/08/2023]
Abstract
Two new brominated bis(indole) alkaloids, dragmacidins I (1) and J (2), showing low micromolar cytostatic activity, along with three known congeners were isolated from the Tanzanian sponge Dragmacidon sp. and their structures determined by the analysis of their NMR and MS data. From the study of their mechanism of action, it can be concluded that the mitotic arrest at metaphase in treated tumor cells, mediated by inhibition of PP1 and/or PP2A phosphatases is involved in the observed antiproliferative activity. Differences in their bioactivities were rationalized, and a plausible binding mode is proposed on the basis of computational simulations.
Collapse
Affiliation(s)
- Patricia
G. Cruz
- R&D
Area, PharmaMar, S.A., Avda. de los Reyes 1, Pol. Ind. La Mina, Colmenar Viejo, Madrid 28770, Spain
| | | | - Antonio Hernández Daranas
- Instituto
de Productos Naturales y Agrobiología, Consejo
Superior de Investigaciones Científicas (CSIC), La Laguna, Santa Cruz de Tenerife 38206, Spain
| | - Marta Pérez
- R&D
Area, PharmaMar, S.A., Avda. de los Reyes 1, Pol. Ind. La Mina, Colmenar Viejo, Madrid 28770, Spain
| | - Carmen Cuevas
- R&D
Area, PharmaMar, S.A., Avda. de los Reyes 1, Pol. Ind. La Mina, Colmenar Viejo, Madrid 28770, Spain
| |
Collapse
|
29
|
Microcystins: Synthesis and structure–activity relationship studies toward PP1 and PP2A. Bioorg Med Chem 2018; 26:1118-1126. [DOI: 10.1016/j.bmc.2017.08.040] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/19/2017] [Accepted: 08/23/2017] [Indexed: 11/19/2022]
|
30
|
Protein Phosphatase 2A in the Regulation of Wnt Signaling, Stem Cells, and Cancer. Genes (Basel) 2018; 9:genes9030121. [PMID: 29495399 PMCID: PMC5867842 DOI: 10.3390/genes9030121] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/16/2018] [Accepted: 02/21/2018] [Indexed: 12/21/2022] Open
Abstract
Protein phosphorylation is a ubiquitous cellular process that allows for the nuanced and reversible regulation of protein activity. Protein phosphatase 2A (PP2A) is a heterotrimeric serine-threonine phosphatase—composed of a structural, regulatory, and catalytic subunit—that controls a variety of cellular events via protein dephosphorylation. While much is known about PP2A and its basic biochemistry, the diversity of its components—especially the multitude of regulatory subunits—has impeded the determination of PP2A function. As a consequence of this complexity, PP2A has been shown to both positively and negatively regulate signaling networks such as the Wnt pathway. Wnt signaling modulates major developmental processes, and is a dominant mediator of stem cell self-renewal, cell fate, and cancer stem cells. Because PP2A affects Wnt signaling both positively and negatively and at multiple levels, further understanding of this complex dynamic may ultimately provide insight into stem cell biology and how to better treat cancers that result from alterations in Wnt signaling. This review will summarize literature that implicates PP2A as a tumor suppressor, explore PP2A mutations identified in human malignancy, and focus on PP2A in the regulation of Wnt signaling and stem cells so as to better understand how aberrancy in this pathway can contribute to tumorigenesis.
Collapse
|
31
|
Hu M, Qu X, Pan L, Fu C, Jia P, Liu Q, Wang Y. Effects of toxic Microcystis aeruginosa on the silver carp Hypophthalmichtys molitrix revealed by hepatic RNA-seq and miRNA-seq. Sci Rep 2017; 7:10456. [PMID: 28874710 PMCID: PMC5585339 DOI: 10.1038/s41598-017-10335-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/02/2017] [Indexed: 12/27/2022] Open
Abstract
High-throughput sequencing was applied to analyze the effects of toxic Microcystis aeruginosa on the silver carp Hypophthalmichthys molitrix. Silver carps were exposed to two cyanobacteria species (toxic and non-toxic) for RNA-seq and miRNA-seq analysis. RNA-seq revealed that the liver tissue contained 105,379 unigenes. Of these genes, 143 were significantly differentiated, 82 were markedly up-regulated, and 61 were remarkably down-regulated. GO term enrichment analysis indicated that 35 of the 154 enriched GO terms were significantly enriched. KEGG pathway enrichment analysis demonstrated that 17 of the 118 enriched KEGG pathways were significantly enriched. A considerable number of disease/immune-associated GO terms and significantly enriched KEGG pathways were also observed. The sequence length determined by miRNA-seq was mainly distributed in 20-23 bp and composed of 882,620 unique small RNAs, and 53% of these RNAs were annotated to miRNAs. As confirmed, 272 known miRNAs were differentially expressed, 453 novel miRNAs were predicted, 112 miRNAs were well matched with 7,623 target genes, and 203 novel miRNAs were matched with 15,453 target genes. qPCR also indicated that Steap4, Cyp7a1, CABZ01088134.1, and PPP1R3G were significantly differentially expressed and might play major roles in the toxic, detoxifying, and antitoxic mechanisms of microcystin in fish.
Collapse
Affiliation(s)
- Menghong Hu
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
- The Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai, 201306, China
- Centre for Research on Environmental Ecology and Fish Nutrion (CREEFN) of the Ministry Agriculture, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture Ministry, Ocean University, Shanghai, China
| | - Xiancheng Qu
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
- The Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai, 201306, China
- Centre for Research on Environmental Ecology and Fish Nutrion (CREEFN) of the Ministry Agriculture, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture Ministry, Ocean University, Shanghai, China
| | - Lisha Pan
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Chunxue Fu
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Peixuan Jia
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Qigen Liu
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China.
- The Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai, 201306, China.
- Centre for Research on Environmental Ecology and Fish Nutrion (CREEFN) of the Ministry Agriculture, Shanghai Ocean University, Shanghai, China.
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture Ministry, Ocean University, Shanghai, China.
| | - Youji Wang
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China.
- The Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai, 201306, China.
- International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, Shanghai, China.
| |
Collapse
|
32
|
Wakimoto T, Egami Y, Abe I. Calyculin: Nature's way of making the sponge-derived cytotoxin. Nat Prod Rep 2017; 33:751-60. [PMID: 26923942 DOI: 10.1039/c5np00123d] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Covering: up to 2015.Calyculin A is a major cytotoxic compound isolated from the Japanese marine sponge Discodermia calyx. Its potent cytotoxicity is attributable to the specific inhibition of protein phosphatases 1 and 2A, as in the case of okadaic acid and the microcystins. Its chemical structure is well-designed not only for enzyme inhibition but also for higher membrane permeability in order to impart its potent cytotoxicity. The biosynthetic gene cluster of this densely functionalized polyketide and nonribosomal peptide hybrid molecule was recently identified from the sponge-microbe association. The producer organism and the dynamic bioconversion process were also revealed. In this highlight, we focus on the recent studies addressing nature's design and biogenesis of the sponge-derived cytotoxin, calyculin A.
Collapse
Affiliation(s)
- Toshiyuki Wakimoto
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo 060-0812, Japan.
| | - Yoko Egami
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo 060-0812, Japan.
| | - Ikuro Abe
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
33
|
Molecular Mechanism for the Regulation of Microcystin Toxicity to Protein Phosphatase 1 by Glutathione Conjugation Pathway. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9676504. [PMID: 28337461 PMCID: PMC5350311 DOI: 10.1155/2017/9676504] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/08/2016] [Accepted: 11/15/2016] [Indexed: 11/21/2022]
Abstract
Glutathione (GSH) conjugation was an important pathway to regulate the toxicity of microcystins (MCs) targeted to protein phosphatases. To explore the specific molecular mechanism for GSH detoxification, two typical MC-GSHs (derived from MCLR and MCRR) were synthesized, prepared, and purified according to previous research. Then, the reduced inhibition effect for MC-GSHs on protein phosphatase 1 was verified by comparing with their original toxins. To further clarify the molecular mechanism for MC-GSHs detoxification, we evaluated the interactions between MCs/MC-GSHs and PP1 with the assistance of MOE molecule simulation. When GSH was introduced to MCs, the covalent binding (Mdha7 to Cys273), the hydrophobic interaction (Adda5 with PP1), the hydrogen bonds (especially for Lys2-Arg96 and Glu6-Tyr272), the covalent combination (between Mdha7 and Cys273), and the ion bonds (between Mn2+ and Asn124/His248/Asp64/His66) of MCLR/MCRR-PP1 complexes weakened to a certain extent, while the ion bonds between Mn2+ and His173/Asp92 residues increased. It was not difficult to find that the toxicity of MCs was closely related to the above sites/interactions and the above key information for MCs-PP1; MC-GSHs-PP1 complexes were important for clarifying the detoxification mechanism of MC-GSHs pathway. This study offers a comprehensive cognition on MCs toxicity regulation and provides valid theoretical support to control their potential risk.
Collapse
|
34
|
Twiner MJ, Doucette GJ, Pang Y, Fang C, Forsyth CJ, Miles CO. Structure-Activity Relationship Studies Using Natural and Synthetic Okadaic Acid/Dinophysistoxin Toxins. Mar Drugs 2016; 14:md14110207. [PMID: 27827901 PMCID: PMC5128750 DOI: 10.3390/md14110207] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 08/07/2016] [Accepted: 10/31/2016] [Indexed: 12/25/2022] Open
Abstract
Okadaic acid (OA) and the closely related dinophysistoxins (DTXs) are algal toxins that accumulate in shellfish and are known serine/threonine protein phosphatase (ser/thr PP) inhibitors. Phosphatases are important modulators of enzyme activity and cell signaling pathways. However, the interactions between the OA/DTX toxins and phosphatases are not fully understood. This study sought to identify phosphatase targets and characterize their structure–activity relationships (SAR) with these algal toxins using a combination of phosphatase activity and cytotoxicity assays. Preliminary screening of 21 human and yeast phosphatases indicated that only three ser/thr PPs (PP2a, PP1, PP5) were inhibited by physiologically saturating concentrations of DTX2 (200 nM). SAR studies employed naturally-isolated OA, DTX1, and DTX2, which vary in degree and/or position of methylation, in addition to synthetic 2-epi-DTX2. OA/DTX analogs induced cytotoxicity and inhibited PP activity with a relatively conserved order of potency: OA = DTX1 ≥ DTX2 >> 2-epi-DTX. The PPs were also differentially inhibited with sensitivities of PP2a > PP5 > PP1. These findings demonstrate that small variations in OA/DTX toxin structures, particularly at the head region (i.e., C1/C2), result in significant changes in toxicological potency, whereas changes in methylation at C31 and C35 (tail region) only mildly affect potency. In addition to this being the first study to extensively test OA/DTX analogs’ activities towards PP5, these data will be helpful for accurately determining toxic equivalence factors (TEFs), facilitating molecular modeling efforts, and developing highly selective phosphatase inhibitors.
Collapse
Affiliation(s)
- Michael J Twiner
- School of Medicine, Wayne State University, Detroit, MI 48201, USA.
- Department of Natural Sciences, University of Michigan, Dearborn, MI 48128, USA.
| | - Gregory J Doucette
- Marine Biotoxins Program, Center for Coastal Environmental Health and Biomolecular Research, NOAA/National Ocean Service, Charleston, SC 29412, USA.
| | - Yucheng Pang
- Department of Chemistry, The Ohio State University, Columbus, OH 43220, USA.
| | - Chao Fang
- Department of Chemistry, The Ohio State University, Columbus, OH 43220, USA.
| | - Craig J Forsyth
- Department of Chemistry, The Ohio State University, Columbus, OH 43220, USA.
| | - Christopher O Miles
- Section for Chemistry and Toxicology, Norwegian Veterinary Institute, Oslo 0454, Norway.
| |
Collapse
|
35
|
Fontanillo M, Zemskov I, Häfner M, Uhrig U, Salvi F, Simon B, Wittmann V, Köhn M. Synthesis of Highly Selective Submicromolar Microcystin-Based Inhibitors of Protein Phosphatase (PP)2A over PP1. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201606449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Miriam Fontanillo
- Genome Biology Unit; European Molecular Biology Laboratory; Meyerhofstrasse 1 69117 Heidelberg Germany
| | - Ivan Zemskov
- Department of Chemistry and Konstanz Research School Chemical Biology (KoRS-CB); University of Konstanz; Fach 709 78457 Konstanz Germany
| | - Maximilian Häfner
- Department of Chemistry and Konstanz Research School Chemical Biology (KoRS-CB); University of Konstanz; Fach 709 78457 Konstanz Germany
| | - Ulrike Uhrig
- Chemical Biology Core Facility; European Molecular Biology Laboratory; Meyerhofstrasse 1 69117 Heidelberg Germany
| | - Francesca Salvi
- Genome Biology Unit; European Molecular Biology Laboratory; Meyerhofstrasse 1 69117 Heidelberg Germany
| | - Bernd Simon
- Structural and Computational Biology Unit; European Molecular Biology Laboratory; Meyerhofstrasse 1 69117 Heidelberg Germany
| | - Valentin Wittmann
- Department of Chemistry and Konstanz Research School Chemical Biology (KoRS-CB); University of Konstanz; Fach 709 78457 Konstanz Germany
| | - Maja Köhn
- Genome Biology Unit; European Molecular Biology Laboratory; Meyerhofstrasse 1 69117 Heidelberg Germany
| |
Collapse
|
36
|
Fontanillo M, Zemskov I, Häfner M, Uhrig U, Salvi F, Simon B, Wittmann V, Köhn M. Synthesis of Highly Selective Submicromolar Microcystin-Based Inhibitors of Protein Phosphatase (PP)2A over PP1. Angew Chem Int Ed Engl 2016; 55:13985-13989. [PMID: 27723199 PMCID: PMC5113787 DOI: 10.1002/anie.201606449] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 08/19/2016] [Indexed: 12/20/2022]
Abstract
Research and therapeutic targeting of the phosphoserine/threonine phosphatases PP1 and PP2A is hindered by the lack of selective inhibitors. The microcystin (MC) natural toxins target both phosphatases with equal potency, and their complex synthesis has complicated structure–activity relationship studies in the past. We report herein the synthesis and biochemical evaluation of 11 MC analogues, which was accomplished through an efficient strategy combining solid‐ and solution‐phase approaches. Our approach led to the first MC analogue with submicromolar inhibitory potency that is strongly selective for PP2A over PP1 and does not require the complex lipophilic Adda group. Through mutational and structural analyses, we identified a new key element for binding, as well as reasons for the selectivity. This work gives unprecedented insight into how selectivity between these phosphatases can be achieved with MC analogues.
Collapse
Affiliation(s)
- Miriam Fontanillo
- Genome Biology Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Ivan Zemskov
- Department of Chemistry and Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Fach 709, 78457, Konstanz, Germany
| | - Maximilian Häfner
- Department of Chemistry and Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Fach 709, 78457, Konstanz, Germany
| | - Ulrike Uhrig
- Chemical Biology Core Facility, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Francesca Salvi
- Genome Biology Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Bernd Simon
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Valentin Wittmann
- Department of Chemistry and Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Fach 709, 78457, Konstanz, Germany
| | - Maja Köhn
- Genome Biology Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117, Heidelberg, Germany.
| |
Collapse
|
37
|
Ma L, Bayram Y, McLaughlin HM, Cho MT, Krokosky A, Turner CE, Lindstrom K, Bupp CP, Mayberry K, Mu W, Bodurtha J, Weinstein V, Zadeh N, Alcaraz W, Powis Z, Shao Y, Scott DA, Lewis AM, White JJ, Jhangiani SN, Gulec EY, Lalani SR, Lupski JR, Retterer K, Schnur RE, Wentzensen IM, Bale S, Chung WK. De novo missense variants in PPP1CB are associated with intellectual disability and congenital heart disease. Hum Genet 2016; 135:1399-1409. [PMID: 27681385 DOI: 10.1007/s00439-016-1731-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 09/19/2016] [Indexed: 10/20/2022]
Abstract
Intellectual disabilities are genetically heterogeneous and can be associated with congenital anomalies. Using whole-exome sequencing (WES), we identified five different de novo missense variants in the protein phosphatase-1 catalytic subunit beta (PPP1CB) gene in eight unrelated individuals who share an overlapping phenotype of dysmorphic features, macrocephaly, developmental delay or intellectual disability (ID), congenital heart disease, short stature, and skeletal and connective tissue abnormalities. Protein phosphatase-1 (PP1) is a serine/threonine-specific protein phosphatase involved in the dephosphorylation of a variety of proteins. The PPP1CB gene encodes a PP1 subunit that regulates the level of protein phosphorylation. All five altered amino acids we observed are highly conserved among the PP1 subunit family, and all are predicted to disrupt PP1 subunit binding and impair dephosphorylation. Our data suggest that our heterozygous de novo PPP1CB pathogenic variants are associated with syndromic intellectual disability.
Collapse
Affiliation(s)
- Lijiang Ma
- Department of Pediatrics, Columbia University Medical Center, 1150 St. Nicholas Avenue, New York, NY, 10032, USA
| | - Yavuz Bayram
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Alyson Krokosky
- Walter Reed National Military Medical Center, Bethesda, MD, USA
| | | | - Kristin Lindstrom
- Division of Genetics and Metabolism, Phoenix Children's Hospital, Phoenix, AZ, USA
| | | | | | - Weiyi Mu
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Joann Bodurtha
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Veronique Weinstein
- Division of Genetics and Metabolism, Children's National Medical Center, Washington, DC, USA
| | | | | | - Zöe Powis
- Ambry Genetics, Aliso Viejo, CA, USA
| | - Yunru Shao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Daryl A Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Andrea M Lewis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Janson J White
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Shalani N Jhangiani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Elif Yilmaz Gulec
- Medical Genetics Section, Kanuni Sultan Suleyman Training and Research Hospital, Istanbul, Turkey
| | - Seema R Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,Texas Children's Hospital, Houston, TX, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | | | | | | | | | - Wendy K Chung
- Department of Pediatrics, Columbia University Medical Center, 1150 St. Nicholas Avenue, New York, NY, 10032, USA.
| |
Collapse
|
38
|
Kumar GS, Gokhan E, De Munter S, Bollen M, Vagnarelli P, Peti W, Page R. The Ki-67 and RepoMan mitotic phosphatases assemble via an identical, yet novel mechanism. eLife 2016; 5. [PMID: 27572260 PMCID: PMC5005033 DOI: 10.7554/elife.16539] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/16/2016] [Indexed: 12/26/2022] Open
Abstract
Ki-67 and RepoMan have key roles during mitotic exit. Previously, we showed that Ki-67 organizes the mitotic chromosome periphery and recruits protein phosphatase 1 (PP1) to chromatin at anaphase onset, in a similar manner as RepoMan (Booth et al., 2014). Here we show how Ki-67 and RepoMan form mitotic exit phosphatases by recruiting PP1, how they distinguish between distinct PP1 isoforms and how the assembly of these two holoenzymes are dynamically regulated by Aurora B kinase during mitosis. Unexpectedly, our data also reveal that Ki-67 and RepoMan bind PP1 using an identical, yet novel mechanism, interacting with a PP1 pocket that is engaged only by these two PP1 regulators. These findings not only show how two distinct mitotic exit phosphatases are recruited to their substrates, but also provide immediate opportunities for the design of novel cancer therapeutics that selectively target the Ki-67:PP1 and RepoMan:PP1 holoenzymes.
Collapse
Affiliation(s)
- Ganesan Senthil Kumar
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, United States
| | - Ezgi Gokhan
- College of Health and Life Science, Research Institute for Environment, Health and Society, Brunel University London, Uxbridge, United Kingdom
| | - Sofie De Munter
- Laboratory of Biosignaling and Therapeutics, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Mathieu Bollen
- Laboratory of Biosignaling and Therapeutics, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Paola Vagnarelli
- College of Health and Life Science, Research Institute for Environment, Health and Society, Brunel University London, Uxbridge, United Kingdom
| | - Wolfgang Peti
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, United States
| | - Rebecca Page
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, United States
| |
Collapse
|
39
|
Structural and functional basis of protein phosphatase 5 substrate specificity. Proc Natl Acad Sci U S A 2016; 113:9009-14. [PMID: 27466404 DOI: 10.1073/pnas.1603059113] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The serine/threonine phosphatase protein phosphatase 5 (PP5) regulates hormone- and stress-induced cellular signaling by association with the molecular chaperone heat shock protein 90 (Hsp90). PP5-mediated dephosphorylation of the cochaperone Cdc37 is essential for activation of Hsp90-dependent kinases. However, the details of this mechanism remain unknown. We determined the crystal structure of a Cdc37 phosphomimetic peptide bound to the catalytic domain of PP5. The structure reveals PP5 utilization of conserved elements of phosphoprotein phosphatase (PPP) structure to bind substrate and provides a template for many PPP-substrate interactions. Our data show that, despite a highly conserved structure, elements of substrate specificity are determined within the phosphatase catalytic domain itself. Structure-based mutations in vivo reveal that PP5-mediated dephosphorylation is required for kinase and steroid hormone receptor release from the chaperone complex. Finally, our data show that hyper- or hypoactivity of PP5 mutants increases Hsp90 binding to its inhibitor, suggesting a mechanism to enhance the efficacy of Hsp90 inhibitors by regulation of PP5 activity in tumors.
Collapse
|
40
|
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, Honkanen RE. Crystal structures and mutagenesis of PPP-family ser/thr protein phosphatases elucidate the selectivity of cantharidin and novel norcantharidin-based inhibitors of PP5C. Biochem Pharmacol 2016; 109:14-26. [PMID: 27002182 DOI: 10.1016/j.bcp.2016.03.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 03/15/2016] [Indexed: 12/24/2022]
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.
Collapse
Affiliation(s)
| | - Mark R Swingle
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Edward A Salter
- Department of Chemistry, University of South Alabama, Mobile, AL 36688, USA
| | - Eric Wood
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA; Department of Chemistry, University of South Alabama, Mobile, AL 36688, USA
| | - Brandon D'Arcy
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Catherine Zivanov
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA; Department of Chemistry, University of South Alabama, Mobile, AL 36688, USA
| | - Kevin Abney
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Alla Musiyenko
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Scott F Rusin
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Arminja Kettenbach
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA; Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Larry Yet
- Department of Chemistry, University of South Alabama, Mobile, AL 36688, USA
| | - Chad E Schroeder
- Department of Medicinal Chemistry, University of Kansas Specialized Chemistry Center, Lawrence, KS 66047, USA
| | - Jennifer E Golden
- Department of Medicinal Chemistry, University of Kansas Specialized Chemistry Center, Lawrence, KS 66047, USA
| | - Wade H Dunham
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Anne-Claude Gingras
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Surajit Banerjee
- Northeastern Collaborative Access Team (NE-CAT) Cornell University, Lemont, IL, USA
| | - David Forbes
- Department of Chemistry, University of South Alabama, Mobile, AL 36688, USA
| | - Andrzej Wierzbicki
- Department of Chemistry, University of South Alabama, Mobile, AL 36688, USA
| | - Richard E Honkanen
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA.
| |
Collapse
|
41
|
Cell Death Inducing Microbial Protein Phosphatase Inhibitors--Mechanisms of Action. Mar Drugs 2015; 13:6505-20. [PMID: 26506362 PMCID: PMC4626703 DOI: 10.3390/md13106505] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 10/12/2015] [Accepted: 10/15/2015] [Indexed: 02/07/2023] Open
Abstract
Okadaic acid (OA) and microcystin (MC) as well as several other microbial toxins like nodularin and calyculinA are known as tumor promoters as well as inducers of apoptotic cell death. Their intracellular targets are the major serine/threonine protein phosphatases. This review summarizes mechanisms believed to be responsible for the death induction and tumor promotion with focus on the interdependent production of reactive oxygen species (ROS) and activation of Ca2+/calmodulin kinase II (CaM-KII). New data are presented using inhibitors of specific ROS producing enzymes to curb nodularin/MC-induced liver cell (hepatocyte) death. They indicate that enzymes of the arachidonic acid pathway, notably phospholipase A2, 5-lipoxygenase, and cyclooxygenases, may be required for nodularin/MC-induced (and presumably OA-induced) cell death, suggesting new ways to overcome at least some aspects of OA and MC toxicity.
Collapse
|
42
|
Fuwa H, Sakamoto K, Muto T, Sasaki M. Concise synthesis of the C15–C38 fragment of okadaic acid, a specific inhibitor of protein phosphatases 1 and 2A. Tetrahedron 2015. [DOI: 10.1016/j.tet.2015.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
43
|
Ehara H, Makino M, Kodama K, Konoki K, Ito T, Sekine SI, Fukuzawa S, Yokoyama S, Tachibana K. Crystal Structure of Okadaic Acid Binding Protein 2.1: A Sponge Protein Implicated in Cytotoxin Accumulation. Chembiochem 2015; 16:1435-9. [PMID: 25965326 DOI: 10.1002/cbic.201500141] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Indexed: 12/18/2022]
Abstract
Okadaic acid (OA) is a marine polyether cytotoxin that was first isolated from the marine sponge Halichondria okadai. OA is a potent inhibitor of protein serine/threonine phosphatases (PP) 1 and 2A, and the structural basis of phosphatase inhibition has been well investigated. However, the role and mechanism of OA retention in the marine sponge have remained elusive. We have solved the crystal structure of okadaic acid binding protein 2.1 (OABP2.1) isolated from H. okadai; it has strong affinity for OA and limited sequence homology to other proteins. The structure revealed that OABP2.1 consists of two α-helical domains, with the OA molecule deeply buried inside the protein. In addition, the global fold of OABP2.1 was unexpectedly similar to that of aequorin, a jellyfish photoprotein. The presence of structural homologues suggested that, by using similar protein scaffolds, marine invertebrates have developed diverse survival systems adapted to their living environments.
Collapse
Affiliation(s)
- Haruhiko Ehara
- Department of Biophysics and Biochemistry and Laboratory of Structural Biology, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033 (Japan).,RIKEN Systems and Structural Biology Center, 1-7-22, Suehiro-cho, Tsurumi, Yokohama 230-0045 (Japan).,RIKEN Center for Life Science Technologies, 1-7-22, Suehiro-cho, Tsurumi, Yokohama 230-0045 (Japan)
| | - Marie Makino
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033 (Japan)
| | - Koichiro Kodama
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033 (Japan)
| | - Keiichi Konoki
- Graduate School of Agricultural Science, Tohoku University, 1-1 Tsutsumidori-Amamiyamachi, Aoba-ku, Sendai, Miyagi 981-8555 (Japan)
| | - Takuhiro Ito
- Department of Biophysics and Biochemistry and Laboratory of Structural Biology, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033 (Japan).,RIKEN Systems and Structural Biology Center, 1-7-22, Suehiro-cho, Tsurumi, Yokohama 230-0045 (Japan).,RIKEN Center for Life Science Technologies, 1-7-22, Suehiro-cho, Tsurumi, Yokohama 230-0045 (Japan)
| | - Shun-ichi Sekine
- Department of Biophysics and Biochemistry and Laboratory of Structural Biology, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033 (Japan).,RIKEN Systems and Structural Biology Center, 1-7-22, Suehiro-cho, Tsurumi, Yokohama 230-0045 (Japan).,RIKEN Center for Life Science Technologies, 1-7-22, Suehiro-cho, Tsurumi, Yokohama 230-0045 (Japan)
| | - Seketsu Fukuzawa
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033 (Japan).
| | - Shigeyuki Yokoyama
- Department of Biophysics and Biochemistry and Laboratory of Structural Biology, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033 (Japan). .,RIKEN Systems and Structural Biology Center, 1-7-22, Suehiro-cho, Tsurumi, Yokohama 230-0045 (Japan). .,RIKEN Structural Biology Laboratory, 1-7-22, Suehiro-cho, Tsurumi, Yokohama 230-0045 (Japan).
| | - Kazuo Tachibana
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033 (Japan).
| |
Collapse
|
44
|
Bastan R, Eskandari N, Sabzghabaee AM, Manian M. Serine/Threonine phosphatases: classification, roles and pharmacological regulation. Int J Immunopathol Pharmacol 2015; 27:473-84. [PMID: 25572726 DOI: 10.1177/039463201402700402] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Phosphatases are important enzymes in a variety of biochemical pathways in different cells which they catalyze opposing reactions of phosphorylation and dephosphorylation, which may modulate the function of crucial signaling proteins in different cells. This is an important mechanism in the regulation of intracellular signal transduction pathways in many cells. Phosphatases play a key role in regulating signal transduction. It is known that phosphatases are specific for cleavage of either serine-threonine or tyrosine phosphate groups. To date, numerous compounds have been identified. This paper reviews the classification, roles and pharmacological of protein serine/threonine phosphates.
Collapse
Affiliation(s)
- R Bastan
- Department of Human Vaccine, Razi-Karaj Institute, Karaj, Iran
| | - N Eskandari
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - A M Sabzghabaee
- Isfahan Clinical Toxicology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - M Manian
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
45
|
Strategies to make protein serine/threonine (PP1, calcineurin) and tyrosine phosphatases (PTP1B) druggable: achieving specificity by targeting substrate and regulatory protein interaction sites. Bioorg Med Chem 2015; 23:2781-5. [PMID: 25771485 DOI: 10.1016/j.bmc.2015.02.040] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/09/2015] [Accepted: 02/19/2015] [Indexed: 12/18/2022]
Abstract
The established dogma is that protein serine/threonine (PSPs) and tyrosine (PTPs) phosphatases are unattainable drug targets. This is because natural product inhibitors of PSP active sites are lethal, while the active sites of PTPs are exceptionally conserved and charged, making it nearly impossible to develop PTP inhibitors that are selective. However, due to a series of recent structural and functional studies, this view of phosphatases is about to undergo a radical change. Rather than target active sites, these studies have demonstrated that targeting PSP/PTP protein (substrate/regulatory) interaction sites, which are distal from the active sites, are highly viable and suitable drugs targets. This is especially true for calcineurin (CN), in which the blockbuster immunosuppressant drugs FK506 and cyclosporin A were recently demonstrated to bind and block one of the key CN substrate interaction sites, the LxVP site. Additional studies show that this approach-targeting substrate and/or regulatory protein interaction sites-also holds incredible promise for protein phosphatase 1 (PP1)-related diseases. Finally, domains outside PTP catalytic domains have also recently been demonstrated to directly alter PTP activity. Collectively, these novel insights offer new, transformative perspectives for the therapeutic targeting of PSPs by interfering with the binding of PIPs or substrates and PTPs by targeting allosteric sites outside their catalytic domains.
Collapse
|
46
|
Fuwa H, Sakamoto K, Muto T, Sasaki M. Concise Synthesis of the C15–C38 Fragment of Okadaic Acid: Application of the Suzuki–Miyaura Reaction to Spiroacetal Synthesis. Org Lett 2014; 17:366-9. [DOI: 10.1021/ol503491t] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Haruhiko Fuwa
- Graduate
School of Life Sciences, Tohoku University, 2-1-1 Katahira,
Aoba-ku, Sendai 980-8577, Japan
| | - Keita Sakamoto
- Graduate
School of Life Sciences, Tohoku University, 2-1-1 Katahira,
Aoba-ku, Sendai 980-8577, Japan
| | - Takashi Muto
- Graduate
School of Life Sciences, Tohoku University, 2-1-1 Katahira,
Aoba-ku, Sendai 980-8577, Japan
| | - Makoto Sasaki
- Graduate
School of Life Sciences, Tohoku University, 2-1-1 Katahira,
Aoba-ku, Sendai 980-8577, Japan
| |
Collapse
|
47
|
Korrodi-Gregório L, Esteves SLC, Fardilha M. Protein phosphatase 1 catalytic isoforms: specificity toward interacting proteins. Transl Res 2014; 164:366-91. [PMID: 25090308 DOI: 10.1016/j.trsl.2014.07.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/26/2014] [Accepted: 07/01/2014] [Indexed: 01/21/2023]
Abstract
The coordinated and reciprocal action of serine-threonine protein kinases and protein phosphatases produces transitory phosphorylation, a fundamental regulatory mechanism for many biological processes. Phosphoprotein phosphatase 1 (PPP1), a major serine-threonine phosphatase, in particular, is ubiquitously distributed and regulates a broad range of cellular functions, including glycogen metabolism, cell cycle progression, and muscle relaxation. PPP1 has evolved effective catalytic machinery but in vitro lacks substrate specificity. In vivo, its specificity is achieved not only by the existence of different PPP1 catalytic isoforms, but also by binding of the catalytic moiety to a large number of regulatory or targeting subunits. Here, we will address exhaustively the existence of diverse PPP1 catalytic isoforms and the relevance of their specific partners and consequent functions.
Collapse
Affiliation(s)
- Luís Korrodi-Gregório
- Laboratório de Transdução de Sinais, Departamento de Biologia, Secção Autónoma de Ciências de Saúde, Centro de Biologia Celular, Universidade de Aveiro, Aveiro, Portugal
| | - Sara L C Esteves
- Laboratório de Transdução de Sinais, Departamento de Biologia, Secção Autónoma de Ciências de Saúde, Centro de Biologia Celular, Universidade de Aveiro, Aveiro, Portugal
| | - Margarida Fardilha
- Laboratório de Transdução de Sinais, Departamento de Biologia, Secção Autónoma de Ciências de Saúde, Centro de Biologia Celular, Universidade de Aveiro, Aveiro, Portugal.
| |
Collapse
|
48
|
Jérôme M, Paudel HK. 14-3-3ζ regulates nuclear trafficking of protein phosphatase 1α (PP1α) in HEK-293 cells. Arch Biochem Biophys 2014; 558:28-35. [DOI: 10.1016/j.abb.2014.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 06/09/2014] [Accepted: 06/11/2014] [Indexed: 12/24/2022]
|
49
|
Kuo TF, Mao D, Hirata N, Khambu B, Kimura Y, Kawase E, Shimogawa H, Ojika M, Nakatsuji N, Ueda K, Uesugi M. Selective elimination of human pluripotent stem cells by a marine natural product derivative. J Am Chem Soc 2014; 136:9798-801. [PMID: 24992689 DOI: 10.1021/ja501795c] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
One of the current obstacles to stem cell therapy is the tumorigenic potential of residual undifferentiated stem cells. The present study reports rediscovery of a synthetic derivative of okadaic acid, a marine polyether toxin, as a reagent that selectively induces the death of human pluripotent stem cells. Cell-based screening of 333 cytotoxic compounds identified methyl 27-deoxy-27-oxookadaate (molecule 1) as a substrate of two ATP-binding cassette (ABC) transporters, ABCB1 (MDR1) and ABCG2 (BCRP), whose expression is repressed in human embryonic stem cells and induced pluripotent stem cells. The results demonstrate that selective elimination of human pluripotent stem cells can be achieved by designing cytotoxic small molecules with appropriate ABC-transporter selectivity.
Collapse
Affiliation(s)
- Ting-Fang Kuo
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University , Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kittler K, Fessard V, Maul R, Hurtaud-Pessel D. CYP3A4 activity reduces the cytotoxic effects of okadaic acid in HepaRG cells. Arch Toxicol 2014; 88:1519-26. [DOI: 10.1007/s00204-014-1206-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 01/20/2014] [Indexed: 11/29/2022]
|