Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.907
Filter
1.
Osteoarthritis Cartilage ; 32(6): 680-689, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38432607

ABSTRACT

OBJECTIVE: Phlpp1 inhibition is a potential therapeutic strategy for cartilage regeneration and prevention of post-traumatic osteoarthritis (PTOA). To understand how Phlpp1 loss affects cartilage structure, cartilage elastic modulus was measured with atomic force microscopy (AFM) in male and female mice after injury. METHODS: Osteoarthritis was induced in male and female Wildtype (WT) and Phlpp1-/- mice by destabilization of the medial meniscus (DMM). At various timepoints post-injury, activity was measured, and knee joints examined with AFM and histology. In another cohort of WT mice, the PHLPP inhibitor NSC117079 was intra-articularly injected 4 weeks after injury. RESULTS: Male WT mice showed decreased activity and histological signs of cartilage damage at 12 but not 6-weeks post-DMM. Female mice showed a less severe response to DMM by comparison, with no histological changes seen at any time point. In both sexes the elastic modulus of medial condylar cartilage was decreased in WT mice but not Phlpp1-/- mice after DMM as measured by AFM. By 6-weeks, cartilage modulus had decreased from 2 MPa to 1 MPa in WT mice. Phlpp1-/- mice showed no change in modulus at 6-weeks and only a 25% decrease at 12-weeks. The PHLPP inhibitor NSC117079 protected cartilage structure and prevented signs of OA 6-weeks post-injury. CONCLUSIONS: AFM is a sensitive method for detecting early changes in articular cartilage post-injury. Phlpp1 suppression, either through genetic deletion or pharmacological inhibition, protects cartilage degradation in a model of PTOA, validating Phlpp1 as a therapeutic target for PTOA.


Subject(s)
Cartilage, Articular , Phosphoprotein Phosphatases , Animals , Cartilage, Articular/pathology , Cartilage, Articular/drug effects , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/genetics , Male , Female , Mice , Disease Models, Animal , Nuclear Proteins/genetics , Nuclear Proteins/antagonists & inhibitors , Mice, Knockout , Microscopy, Atomic Force , Osteoarthritis/pathology , Elastic Modulus , Osteoarthritis, Knee/etiology , Osteoarthritis, Knee/pathology , Tibial Meniscus Injuries/complications
2.
Curr Drug Targets ; 25(3): 171-189, 2024.
Article in English | MEDLINE | ID: mdl-38213163

ABSTRACT

There exists a huge number of patients suffering from chronic liver disease worldwide. As a disease with high incidence and mortality worldwide, strengthening the research on the pathogenesis of chronic liver disease and the development of novel drugs is an important issue related to the health of all human beings. Phosphorylation modification of proteins plays a crucial role in cellular signal transduction, and phosphatases are involved in the development of liver diseases. Therefore, this article summarized the important role of protein phosphatases in chronic liver disease with the aim of facilitating the development of drugs targeting protein phosphatases for the treatment of chronic liver disease.


Subject(s)
Liver Diseases , Phosphoprotein Phosphatases , Humans , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Liver Diseases/drug therapy , Liver Diseases/enzymology , Liver Diseases/metabolism , Chronic Disease , Signal Transduction/drug effects , Animals , Molecular Targeted Therapy , Phosphorylation , Enzyme Inhibitors/therapeutic use , Enzyme Inhibitors/pharmacology
3.
Food Chem Toxicol ; 169: 113449, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36206954

ABSTRACT

Okadaic acid (OA) is an important marine lipophilic phycotoxin responsible for diarrhetic shellfish poisoning (DSP). This toxin inhibits protein phosphatases (PPs) like PP2A and PP1, though, this action does not explain OA-induced toxicity and symptoms. Intestinal epithelia comprise the defence barrier against external agents where transport of fluid and electrolytes from and to the lumen is a tightly regulated process. In some intoxications this balance becomes dysregulated appearing diarrhoea. Therefore, we evaluated diarrhoea in orally OA-treated mice as well as in mice pre-treated with several doses of cyproheptadine (CPH) and then treated with OA at different times. We assessed stools electrolytes and ultrastructural alteration of the intestine, particularly evaluating tight and adherens junctions. We detected increased chloride and sodium faecal concentrations in the OA-exposed group, suggesting a secretory diarrhoea. Pre-treatment with CPH maintains chloride concentration in values similar to control mice. Intestinal cytomorphological alterations were observed for OA mice, whereas CPH pre-treatment attenuated OA-induced damage in proximal colon and jejunum at 2 h. Conversely, tight junctions' distance was only affected by OA in jejunum at the moment diarrhoea occurred. In this study we found cellular mechanisms by which OA induced diarrhoea revealing the complex toxicity of this compound.


Subject(s)
Diarrhea , Okadaic Acid , Animals , Mice , Chlorides/analysis , Chlorides/metabolism , Cyproheptadine/pharmacology , Diarrhea/chemically induced , Okadaic Acid/toxicity , Phosphoprotein Phosphatases/antagonists & inhibitors , Sodium/analysis , Sodium/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , Jejunum/drug effects , Jejunum/metabolism
4.
Cell Chem Biol ; 29(6): 930-946.e9, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35443151

ABSTRACT

Phosphatase PPM1F is a regulator of cell adhesion by fine-tuning integrin activity and actin cytoskeleton structures. Elevated expression of this enzyme in human tumors is associated with high invasiveness, enhanced metastasis, and poor prognosis. Thus, PPM1F is a target for pharmacological intervention, yet inhibitors of this enzyme are lacking. Here, we use high-throughput screening to identify Lockdown, a reversible and non-competitive PPM1F inhibitor. Lockdown is selective for PPM1F, because this compound does not inhibit other protein phosphatases in vitro and does not induce additional phenotypes in PPM1F knockout cells. Importantly, Lockdown-treated glioblastoma cells fully re-capitulate the phenotype of PPM1F-deficient cells as assessed by increased phosphorylation of PPM1F substrates and corruption of integrin-dependent cellular processes. Ester modification yields LockdownPro with increased membrane permeability and prodrug-like properties. LockdownPro suppresses tissue invasion by PPM1F-overexpressing human cancer cells, validating PPM1F as a therapeutic target and providing an access point to control tumor cell dissemination.


Subject(s)
Glioblastoma , Integrins , Neoplasm Invasiveness , Phosphoprotein Phosphatases , Cell Line, Tumor , Glioblastoma/drug therapy , Humans , Integrins/metabolism , Neoplasm Invasiveness/prevention & control , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylation
5.
J Med Chem ; 65(1): 507-519, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34931516

ABSTRACT

The repressor element-1 silencing transcription factor (REST) represses neuronal gene expression, whose dysregulation is implicated in brain tumors and neurological diseases. A high level of REST protein drives the tumor growth in some glioblastoma cells. While transcription factors like REST are challenging targets for small-molecule inhibitors, the inactivation of a regulatory protein, small CTD phosphatase 1 (SCP1), promotes REST degradation and reduces transcriptional activity. This study rationally designed a series of α,ß-unsaturated sulfones to serve as potent and selective covalent inhibitors against SCP1. The compounds inactivate SCP1 via covalent modification of Cys181 located at the active site entrance. Cellular studies showed that the inhibitors inactivate SCP1 in a time- and dose-dependent manner with an EC50 ∼1.5 µM, reducing REST protein levels and activating specific REST-suppressed genes. These compounds represent a promising line of small-molecule inhibitors as a novel lead for glioblastoma whose growth is driven by REST transcription activity.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Repressor Proteins/drug effects , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Cell Line, Tumor , Dose-Response Relationship, Drug , Glioblastoma/drug therapy , High-Throughput Screening Assays , Humans , Models, Molecular , Molecular Docking Simulation , Repressor Proteins/metabolism
6.
Toxicol Appl Pharmacol ; 430: 115726, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34537213

ABSTRACT

Breast cancer is the leading cause of cancer-related death in women worldwide, and despite multiple chemotherapeutic approaches, effective treatment strategies for advanced metastatic breast cancer are still lacking. Metabolic reprogramming is essential for tumor cell growth and propagation, and most cancers, including breast cancer, are accompanied by abnormalities in energy metabolism. Here, we confirmed that sodium cantharidate inhibited cell viability using the Cell Counting Kit-8, clonogenic assay, and Transwell assay. The cell cycle and apoptosis assays indicated that sodium cantharidate induced apoptosis and cell cycle arrest in breast cancer cells. Additionally, proteomic assays, western blots, and metabolic assays revealed that sodium cantharidate converted the metabolic phenotype of breast cancer cells from glycolysis to oxidative phosphorylation. Furthermore, bioinformatics analysis identified possible roles for p53 with respect to the effects of sodium cantharidate on breast cancer cells. Western blot, docking, and phosphatase assays revealed that the regulation of p53 activity by sodium cantharidate was related to its inhibition of protein phosphatase 5 activity. Moreover, sodium cantharidate significantly inhibited tumor growth in tumor-bearing nude mice. In summary, our study provides evidence for the use of sodium cantharidate as an effective and new therapeutic candidate for the treatment of human breast cancer in clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cantharidin/analogs & derivatives , Energy Metabolism/drug effects , Enzyme Inhibitors/pharmacology , Nuclear Proteins/antagonists & inhibitors , Phosphoprotein Phosphatases/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cantharidin/pharmacology , Cell Cycle Checkpoints/drug effects , Female , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
7.
Oxid Med Cell Longev ; 2021: 5570731, 2021.
Article in English | MEDLINE | ID: mdl-34394827

ABSTRACT

BACKGROUND: Ulcerative colitis (UC) is a chronic inflammatory disease with increasing prevalence worldwide. Barrier defect in intestinal epithelial cells (IECs) is one of the main pathogeneses in UC. Pyroptosis is a programmed lytic cell death and is triggered by inflammatory caspases, while little is known about its role in UC. METHODS: Differentially expressed genes (DEGs) were identified by comparing UC patients with healthy controls from the GEO datasets. The candidate genes involved in pyroptosis were obtained, and the underlying molecular mechanism in the progression of UC was explored in vivo and in vitro. RESULTS: Pleckstrin homology domain leucine-rich repeat protein phosphatase 2 (PHLPP2), a protein phosphatase, was downregulated and involved in regulating inflammation-induced IEC pyroptosis by modulating the NF-κB signaling in UC through bioinformatics analysis. Moreover, we demonstrated that PHLPP2 was downregulated in UC patients and UC mice. Besides, we found that PHLPP2 depletion activated the NF-κB signaling and increased the expressions of caspase-1 P20, Gasdermin N, IL-18, and IL-1ß contributing to IEC pyroptosis and inflammation in UC mice. Furthermore, we found that PHLPP2-/- mice developed hypersensitivity to dextran sulfate sodium (DSS) treatment toward colitis showing activated NF-κB signaling and dramatically induced expressions of caspase-1 P20, Gasdermin N, IL-18, and IL-1ß. Mechanically, this inflammation-induced downregulation of PHLPP2 was alleviated by an NF-κB signaling inhibitor in intestinal organoids of PHLPP2-/- mice and fetal colonic cells. CONCLUSIONS: PHLPP2 downexpression activated the NF-κB signaling and promoted the IEC pyroptosis, leading to UC progression. Therefore, PHLPP2 might be an attractive candidate therapeutic target for UC.


Subject(s)
Colitis, Ulcerative/pathology , Phosphoprotein Phosphatases/genetics , Pyroptosis , Signal Transduction , Animals , Colitis, Ulcerative/chemically induced , Dextran Sulfate/toxicity , Disease Models, Animal , Down-Regulation , Humans , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestines/cytology , Intestines/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , RNA Interference , RNA, Small Interfering/metabolism
8.
J Med Chem ; 64(13): 8916-8938, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34156850

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.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Phosphoprotein Phosphatases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Neoplasms/metabolism , Phosphoprotein Phosphatases/metabolism , Small Molecule Libraries/chemistry
9.
Biochem J ; 478(2): 341-355, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33502516

ABSTRACT

Protein kinase signalling, which transduces external messages to mediate cellular growth and metabolism, is frequently deregulated in human disease, and specifically in cancer. As such, there are 77 kinase inhibitors currently approved for the treatment of human disease by the FDA. Due to their historical association as the receptors for the tumour-promoting phorbol esters, PKC isozymes were initially targeted as oncogenes in cancer. However, a meta-analysis of clinical trials with PKC inhibitors in combination with chemotherapy revealed that these treatments were not advantageous, and instead resulted in poorer outcomes and greater adverse effects. More recent studies suggest that instead of inhibiting PKC, therapies should aim to restore PKC function in cancer: cancer-associated PKC mutations are generally loss-of-function and high PKC protein is protective in many cancers, including most notably KRAS-driven cancers. These recent findings have reframed PKC as having a tumour suppressive function. This review focusses on a potential new mechanism of restoring PKC function in cancer - through targeting of its negative regulator, the Ser/Thr protein phosphatase PHLPP. This phosphatase regulates PKC steady-state levels by regulating the phosphorylation of a key site, the hydrophobic motif, whose phosphorylation is necessary for the stability of the enzyme. We also consider whether the phosphorylation of the potent oncogene KRAS provides a mechanism by which high PKC expression may be protective in KRAS-driven human cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/metabolism , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Kinase C/metabolism , Genes, Tumor Suppressor , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Nuclear Proteins/antagonists & inhibitors , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
10.
Bioorg Med Chem ; 32: 116012, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33454654

ABSTRACT

Cantharidin is a potent natural protein phosphatase monoterpene anhydride inhibitor secreted by several species of blister beetle, with its demethylated anhydride analogue, (S)-palasonin, occurring as a constituent of the higher plant Butea frondosa. Cantharidin shows both potent protein phosphatase inhibitory and cancer cell cytotoxic activities, but possible preclinical development of this anhydride has been limited thus far by its toxicity. Thus, several synthetic derivatives of cantharidin have been prepared, of which some compounds exhibit improved antitumor potential and may have use as lead compounds. In the present review, the potential antitumor activity, structure-activity relationships, and development of cantharidin-based anticancer drug conjugates are summarized, with protein phosphatase-related and other types of mechanisms of action discussed. Protein phosphatases play a key role in the tumor microenvironment, and thus described herein is also the potential for developing new tumor microenvironment-targeted cancer chemotherapeutic agents, based on cantharidin and its naturally occurring analogues and synthetic derivatives.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cantharidin/pharmacology , Enzyme Inhibitors/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/chemistry , Butea/chemistry , Cantharidin/chemistry , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Phosphoprotein Phosphatases/metabolism
11.
Hepatology ; 74(1): 397-410, 2021 07.
Article in English | MEDLINE | ID: mdl-33314176

ABSTRACT

BACKGROUND AND AIMS: Following mild liver injury, pre-existing hepatocytes replicate. However, if hepatocyte proliferation is compromised, such as in chronic liver diseases, biliary epithelial cells (BECs) contribute to hepatocytes through liver progenitor cells (LPCs), thereby restoring hepatic mass and function. Recently, augmenting innate BEC-driven liver regeneration has garnered attention as an alternative to liver transplantation, the only reliable treatment for patients with end-stage liver diseases. Despite this attention, the molecular basis of BEC-driven liver regeneration remains poorly understood. APPROACH AND RESULTS: By performing a chemical screen with the zebrafish hepatocyte ablation model, in which BECs robustly contribute to hepatocytes, we identified farnesoid X receptor (FXR) agonists as inhibitors of BEC-driven liver regeneration. Here we show that FXR activation blocks the process through the FXR-PTEN (phosphatase and tensin homolog)-PI3K (phosphoinositide 3-kinase)-AKT-mTOR (mammalian target of rapamycin) axis. We found that FXR activation blocked LPC-to-hepatocyte differentiation, but not BEC-to-LPC dedifferentiation. FXR activation also suppressed LPC proliferation and increased its death. These defects were rescued by suppressing PTEN activity with its chemical inhibitor and ptena/b mutants, indicating PTEN as a critical downstream mediator of FXR signaling in BEC-driven liver regeneration. Consistent with the role of PTEN in inhibiting the PI3K-AKT-mTOR pathway, FXR activation reduced the expression of pS6, a marker of mTORC1 activation, in LPCs of regenerating livers. Importantly, suppressing PI3K and mTORC1 activities with their chemical inhibitors blocked BEC-driven liver regeneration, as did FXR activation. CONCLUSIONS: FXR activation impairs BEC-driven liver regeneration by enhancing PTEN activity; the PI3K-AKT-mTOR pathway controls the regeneration process. Given the clinical trials and use of FXR agonists for multiple liver diseases due to their beneficial effects on steatosis and fibrosis, the detrimental effects of FXR activation on LPCs suggest a rather personalized use of the agonists in the clinic.


Subject(s)
Cell Differentiation/drug effects , Liver Regeneration/drug effects , Receptors, Cytoplasmic and Nuclear/agonists , Stem Cells/drug effects , Animals , Animals, Genetically Modified , Biliary Tract/cytology , Cell Proliferation , Drug Evaluation, Preclinical , Epithelial Cells/drug effects , Epithelial Cells/physiology , Hepatocytes/drug effects , Hepatocytes/physiology , Liver/drug effects , Liver/physiology , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Stem Cells/physiology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Zebrafish , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Toxins (Basel) ; 12(10)2020 10 04.
Article in English | MEDLINE | ID: mdl-33020400

ABSTRACT

Cyanobacterial harmful algal blooms (CyanoHABs) produce microcystins (MCs) which are associated with animal and human hepatotoxicity. Over 270 variants of MC exist. MCs have been continually studied due of their toxic consequences. Monitoring water quality to assess the presence of MCs is of utmost importance although it is often difficult because CyanoHABs may generate multiple MC variants, and their low concentration in water. To effectively manage and control these toxins and prevent their health risks, sensitive, fast, and reliable methods capable of detecting MCs are required. This paper aims to review the three main analytical methods used to detect MCs ranging from biological (mouse bioassay), biochemical (protein phosphatase inhibition assay and enzyme linked immunosorbent assay), and chemical (high performance liquid chromatography, liquid chromatography-mass spectrometry, high performance capillary electrophoresis, and gas chromatography), as well as the newly emerging biosensor methods. In addition, the current state of these methods regarding their novel development and usage, as well as merits and limitations are presented. Finally, this paper also provides recommendations and future research directions towards method application and improvement.


Subject(s)
Cyanobacteria/metabolism , Environmental Monitoring , Enzyme Inhibitors/analysis , Microcystins/analysis , Water Pollutants, Chemical/analysis , Animals , Biological Assay , Biosensing Techniques , Chromatography , Cyanobacteria/growth & development , Enzyme Inhibitors/toxicity , Harmful Algal Bloom , Lethal Dose 50 , Mass Spectrometry , Mice , Microcystins/toxicity , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Reproducibility of Results , Water Microbiology , Water Pollutants, Chemical/toxicity
13.
Toxins (Basel) ; 12(9)2020 09 11.
Article in English | MEDLINE | ID: mdl-32932764

ABSTRACT

[D-Leu1]MC-LR and MC-LR, two microcystins differing in one amino acid, constitute a sanitary and environmental problem owing to their frequent and concomitant presence in water bodies of the Americas and their association with human intoxication during recreational exposure to cyanobacterial bloom. Present in reservoirs used for irrigation as well, they can generate problems in the development of crops such as Phaseolus vulgaris, of nutritional and economic interest to the region. Although numerous works address the toxic effects of MC-LR, information on the toxicity of [D-Leu1]MC-LR is limited. Our objective was to study the toxic effects of [D-Leu1]MC-LR and MC-LR (3.5 µg/ml) on P. vulgaris after a single contact at the imbibition stage. Our findings indicate that 10 days post treatment, [D-Leu1]MC-LR generates morphological and physiological alterations more pronounced than those caused by MC-LR. In addition to the alterations produced by [D-Leu1]MC-LR in the development of seedlings and the structure of the leaves, roots and stems, we also found alterations in leaf stomatal density and conductivity, a longer delay in the phototropic response and a decrease in the maximum curvature angles achieved with respect to that observed for MC-LR. Our findings indicate that these alterations are linked to the greater inhibition of phosphatase activity generated by [D-Leu1]MC-LR, rather than to oxidative damage. We observed that 30 days after treatment with MC-LR, plants presented better development and recovery than those treated with [D-Leu1]MC-LR. Further studies are required on [D-Leu1]MC-LR and MC-LR toxicity and their underlying mechanisms of action.


Subject(s)
Marine Toxins/toxicity , Microcystins/toxicity , Phaseolus/drug effects , Phototrophic Processes/drug effects , Plant Development/drug effects , Lipid Peroxidation/drug effects , Oxidative Stress/drug effects , Phaseolus/enzymology , Phaseolus/growth & development , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Plant Proteins/antagonists & inhibitors , Plant Proteins/metabolism , Time Factors
14.
Exp Hematol ; 91: 22-31, 2020 11.
Article in English | MEDLINE | ID: mdl-32976949

ABSTRACT

The discovery that the immunomodulatory imide drugs (IMiDs) possess antitumor properties revolutionized the treatment of specific types of hematological cancers. Since then, much progress has been made in understanding why the IMiDs are so efficient in targeting the malignant clones in difficult-to-treat diseases. Despite their efficacy, IMiD resistance arises eventually. Herein we summarize the mechanisms of sensitivity and resistance to lenalidomide in del(5q) myelodysplastic syndrome and multiple myeloma, two diseases in which these drugs are at the therapeutic frontline. Understanding the molecular and cellular mechanisms underlying IMiD efficacy and resistance may allow development of specific strategies to eliminate the malignant clone in otherwise incurable diseases.


Subject(s)
Antineoplastic Agents/pharmacology , Immunologic Factors/pharmacology , Lenalidomide/pharmacology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Anemia, Macrocytic/drug therapy , Anemia, Macrocytic/physiopathology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Cell Differentiation/drug effects , Chromosome Deletion , Chromosomes, Human, Pair 5 , Cytokines/metabolism , Disease Progression , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ikaros Transcription Factor/antagonists & inhibitors , Immunologic Factors/therapeutic use , Lenalidomide/therapeutic use , Megakaryocytes/drug effects , Multiple Myeloma/drug therapy , Multiple Myeloma/physiopathology , Neoplasm Proteins/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/physiopathology , Phosphoprotein Phosphatases/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/physiology
15.
J Biol Chem ; 295(45): 15342-15365, 2020 11 06.
Article in English | MEDLINE | ID: mdl-32868295

ABSTRACT

The contraction and relaxation of the heart is controlled by stimulation of the ß1-adrenoreceptor (AR) signaling cascade, which leads to activation of cAMP-dependent protein kinase (PKA) and subsequent cardiac protein phosphorylation. Phosphorylation is counteracted by the main cardiac protein phosphatases, PP2A and PP1. Both kinase and phosphatases are sensitive to intramolecular disulfide formation in their catalytic subunits that inhibits their activity. Additionally, intermolecular disulfide formation between PKA type I regulatory subunits (PKA-RI) has been described to enhance PKA's affinity for protein kinase A anchoring proteins, which alters its subcellular distribution. Nitroxyl donors have been shown to affect contractility and relaxation, but the mechanistic basis for this effect is unclear. The present study investigates the impact of several nitroxyl donors and the thiol-oxidizing agent diamide on cardiac myocyte protein phosphorylation and oxidation. Although all tested compounds equally induced intermolecular disulfide formation in PKA-RI, only 1-nitrosocyclohexalycetate (NCA) and diamide induced reproducible protein phosphorylation. Phosphorylation occurred independently of ß1-AR activation, but was abolished after pharmacological PKA inhibition and thus potentially attributable to increased PKA activity. NCA treatment of cardiac myocytes induced translocation of PKA and phosphatases to the myofilament compartment as shown by fractionation, immunofluorescence, and proximity ligation assays. Assessment of kinase and phosphatase activity within the myofilament fraction of cardiac myocytes after exposure to NCA revealed activation of PKA and inhibition of phosphatase activity thus explaining the increase in phosphorylation. The data suggest that the NCA-mediated effect on cardiac myocyte protein phosphorylation orchestrates alterations in the kinase/phosphatase balance.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Oxidants/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Receptors, Adrenergic, beta-1/metabolism , Signal Transduction , Acetates/pharmacology , Animals , Cattle , Cyclic AMP-Dependent Protein Kinases/metabolism , Diamide/pharmacology , Humans , Male , Mice , Myocytes, Cardiac/metabolism , Nitroso Compounds/pharmacology , Oxidation-Reduction , Phosphoprotein Phosphatases/metabolism , Phosphorylation/drug effects , Rabbits , Rats , Rats, Wistar , Signal Transduction/drug effects
16.
PLoS One ; 15(8): e0228002, 2020.
Article in English | MEDLINE | ID: mdl-32764831

ABSTRACT

Irinotecan specifically targets topoisomerase I (topoI), and is used to treat various solid tumors, but only 13-32% of patients respond to the therapy. Now, it is understood that the rapid rate of topoI degradation in response to irinotecan causes irinotecan resistance. We have published that the deregulated DNA-PKcs kinase cascade ensures rapid degradation of topoI and is at the core of the drug resistance mechanism of topoI inhibitors, including irinotecan. We also identified CTD small phosphatase 1 (CTDSP1) (a nuclear phosphatase) as a primary upstream regulator of DNA-PKcs in response to topoI inhibitors. Previous reports showed that rabeprazole, a proton pump inhibitor (PPI) inhibits CTDSP1 activity. The purpose of this study was to confirm the effects of rabeprazole on CTDSP1 activity and its impact on irinotecan-based therapy in colon cancer. Using differentially expressing CTDSP1 cells, we demonstrated that CTDSP1 contributes to the irinotecan sensitivity by preventing topoI degradation. Retrospective analysis of patients receiving irinotecan with or without rabeprazole has shown the effects of CTDSP1 on irinotecan response. These results indicate that CTDSP1 promotes sensitivity to irinotecan and rabeprazole prevents this effect, resulting in drug resistance. To ensure the best chance at effective treatment, rabeprazole may not be a suitable PPI for cancer patients treated with irinotecan.


Subject(s)
Colorectal Neoplasms/metabolism , DNA Topoisomerases, Type I/metabolism , Rabeprazole/metabolism , Cell Line, Tumor , Colonic Neoplasms/metabolism , Colorectal Neoplasms/physiopathology , DNA , DNA Topoisomerases, Type I/physiology , DNA-Activated Protein Kinase/metabolism , Drug Resistance/drug effects , Drug Resistance, Neoplasm/physiology , Humans , Irinotecan/metabolism , Irinotecan/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Proton Pump Inhibitors/pharmacology , Rabeprazole/pharmacology , Retrospective Studies , Topoisomerase I Inhibitors/pharmacology
17.
Pharmacol Ther ; 215: 107622, 2020 11.
Article in English | MEDLINE | ID: mdl-32650009

ABSTRACT

Protein phosphatases and kinases control multiple cellular events including proliferation, differentiation, and stress responses through regulating reversible protein phosphorylation, the most important post-translational modification. Members of metal-dependent protein phosphatase (PPM) family, also known as PP2C phosphatases, are Ser/Thr phosphatases that bind manganese/magnesium ions (Mn2+/Mg2+) in their active center and function as single subunit enzymes. In mammals, there are 20 isoforms of PPM phosphatases: PPM1A, PPM1B, PPM1D, PPM1E, PPM1F, PPM1G, PPM1H, PPM1J, PPM1K, PPM1L, PPM1M, PPM1N, ILKAP, PDP1, PDP2, PHLPP1, PHLPP2, PP2D1, PPTC7, and TAB1, whereas there are only 8 in yeast. Phylogenetic analysis of the DNA sequences of vertebrate PPM isoforms revealed that they can be divided into 12 different classes: PPM1A/PPM1B/PPM1N, PPM1D, PPM1E/PPM1F, PPM1G, PPM1H/PPM1J/PPM1M, PPM1K, PPM1L, ILKAP, PDP1/PDP2, PP2D1/PHLPP1/PHLPP2, TAB1, and PPTC7. PPM-family members have a conserved catalytic core region, which contains the metal-chelating residues. The different isoforms also have isoform specific regions within their catalytic core domain and terminal domains, and these regions may be involved in substrate recognition and/or functional regulation of the phosphatases. The twenty mammalian PPM phosphatases are involved in regulating diverse cellular functions, such as cell cycle control, cell differentiation, immune responses, and cell metabolism. Mutation, overexpression, or deletion of the PPM phosphatase gene results in abnormal cellular responses, which lead to various human diseases. This review focuses on the structures and biological functions of the PPM-phosphatase family and their associated diseases. The development of specific inhibitors against the PPM phosphatase family as a therapeutic strategy will also be discussed.


Subject(s)
Enzyme Inhibitors/pharmacology , Metals/metabolism , Phosphoprotein Phosphatases/metabolism , Animals , Drug Development , Gene Expression Regulation , Humans , Mutation , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/chemistry
18.
Int J Mol Sci ; 21(11)2020 May 27.
Article in English | MEDLINE | ID: mdl-32471161

ABSTRACT

Heparan sulfate proteoglycan syndecan-1, CD138, is known to be associated with cell proliferation, adhesion, and migration in malignancies. We previously reported that syndecan-1 (CD138) may contribute to urothelial carcinoma cell survival and progression. We investigated the role of heparanase, an enzyme activated by syndecan-1 in human urothelial carcinoma. Using human urothelial cancer cell lines, MGH-U3 and T24, heparanase expression was reduced with siRNA and RK-682, a heparanase inhibitor, to examine changes in cell proliferation activity, induction of apoptosis, invasion ability of cells, and its relationship to autophagy. A bladder cancer development mouse model was treated with RK-682 and the bladder tissues were examined using immunohistochemical analysis for Ki-67, E-cadherin, LC3, and CD31 expressions. Heparanase inhibition suppressed cellular growth by approximately 40% and induced apoptosis. The heparanase inhibitor decreased cell activity in a concentration-dependent manner and suppressed invasion ability by 40%. Inhibition of heparanase was found to suppress autophagy. In N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN)-induced bladder cancer mice, treatment with heparanase inhibitor suppressed the progression of cancer by 40%, compared to controls. Immunohistochemistry analysis showed that heparanase inhibitor suppressed cell growth, and autophagy. In conclusion, heparanase suppresses apoptosis and promotes invasion and autophagy in urothelial cancer.


Subject(s)
Cell Adhesion , Cell Movement , Glucuronidase/metabolism , Urinary Bladder Neoplasms/metabolism , Aged , Aged, 80 and over , Animals , Apoptosis , Autophagy , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Female , Glucuronidase/antagonists & inhibitors , Glucuronidase/genetics , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Middle Aged , Neoplasm Invasiveness , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/pharmacology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Urinary Bladder Neoplasms/pathology
19.
Cell Stress Chaperones ; 25(3): 383-394, 2020 05.
Article in English | MEDLINE | ID: mdl-32239474

ABSTRACT

Protein phosphatase 5 (PP5) is a serine/threonine protein phosphatase that regulates many cellular functions including steroid hormone signaling, stress response, proliferation, apoptosis, and DNA repair. PP5 is also a co-chaperone of the heat shock protein 90 molecular chaperone machinery that assists in regulation of cellular signaling pathways essential for cell survival and growth. PP5 plays a significant role in survival and propagation of multiple cancers, which makes it a promising target for cancer therapy. Though there are several naturally occurring PP5 inhibitors, none is specific for PP5. Here, we review the roles of PP5 in cancer progression and survival and discuss the unique features of the PP5 structure that differentiate it from other phosphoprotein phosphatase (PPP) family members and make it an attractive therapeutic target.


Subject(s)
Neoplasms/enzymology , Nuclear Proteins/chemistry , Nuclear Proteins/physiology , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/physiology , Breast Neoplasms/enzymology , Catalytic Domain , Female , HSP90 Heat-Shock Proteins/metabolism , Humans , Neoplasms/drug therapy , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism
20.
Comput Biol Chem ; 85: 107230, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32062376

ABSTRACT

Ser/thr phosphatase Stp1 is an important virulence factor for Staphylococcus aureus (S. aureus) and plays a key role in its infectivity, suggesting that it could serve as a potential target for treatment of S. aureus infection. Previous studies found that the activity of Stp1 was inhibited by MDSA and its derivatives. In this paper, we used molecular docking, molecular modeling, molecular dynamics simulations, binding free energy decomposition calculations, and hydrogen bond analyses to explore the structure-activity relationship. Energy decomposition indicated that MDSA, hydroxymethyl MDSA, carboxymethyl MDSA and methyl MDSA can bind to the catalytic pocket of Stp1. Furthermore, Met39, Ile163, Ile164, Val167, Gly195 and Asp233 were key residues in the Stp1-inhibitor complexes. Due to the lack of a double salicylate structure, salicylic acid cannot bind to the active site of Stpl, leading to loss of inhibitory activity. Based on these results, the structure-activity relationship at the atomic level was determined, which can promote the development of new and more effective anti-drug resistance inhibitors.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Salicylates/pharmacology , Staphylococcus aureus/enzymology , Bacterial Proteins/metabolism , Computational Biology , Enzyme Inhibitors/chemistry , Models, Molecular , Molecular Structure , Phosphoprotein Phosphatases/metabolism , Salicylates/chemistry , Structure-Activity Relationship , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...