Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
J Cancer Res Clin Oncol ; 148(1): 57-70, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34981193

ABSTRACT

PURPOSE: Therapy resistance is the principal obstacle to achieving cures in cancer patients and its successful tackling requires a deep understanding of the resistance mediators. Increasing evidence indicates that tumor phosphatases are novel and druggable targets in translational oncology and their modulation may hinder tumor growth and motility and potentiate therapeutic sensitivity in various neoplasms via regulation of various signal transduction pathways. Dual-specificity phosphatases (DUSPs) are key players of cell growth, survival and death and have essential roles in tumor initiation, malignant progression and therapy resistance through regulation of the MAPK signaling pathway. In this review, different aspects of DUSPs are discussed. METHODS: A comprehensive literature review was performed using various websites including PubMed. RESULTS: We provide mechanistic insights into the roles of well-known DUSPs in resistance to a wide range of cancer therapeutic approaches including chemotherapy, radiation and molecular targeted therapy in human malignancies. Moreover, we discuss the development of DUSP modulators, with a focus on DUSP1 and 6 inhibitors. Ultimately, the preclinical investigations of small molecule inhibitors of DUSP1 and 6 are outlined. CONCLUSION: Emerging evidence indicates that the DUSP family is aberrantly expressed in human malignancies and plays critical roles in determining sensitivity to a wide range of cancer therapeutic strategies through regulation of the MAPK signaling pathways. Consequently, targeting DUSPs and their downstream molecules can pave the way for more effective cancer therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 6/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Neoplasms/drug therapy , Benzofurans/pharmacology , Carcinogenesis/pathology , Drug Resistance, Neoplasm/genetics , Dual Specificity Phosphatase 1/biosynthesis , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 6/biosynthesis , Dual Specificity Phosphatase 6/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Targeted Therapy/methods , Neoplasms/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
2.
J Pain ; 21(11-12): 1149-1159, 2020.
Article in English | MEDLINE | ID: mdl-30660765

ABSTRACT

It is widely accepted that neuroinflammation in the spinal cord contributes to the development of central sensitization in neuropathic pain. Mitogen-activated protein kinase (MAPK) activation plays a vital role in the development of neuroinflammation in the spinal cord. In this study, we investigated the effect of bexarotene (bex), a retinoid X receptor agonist, on MAPKs activation in chronic constriction injury (CCI)-induced neuropathic pain. The data showed that daily treatment with bex 50 mg/kg significantly alleviated CCI-induced nociceptive hypersensitivity in rats. Bex 50 mg/kg/day inhibited CCI-induced MAPKs (p38MAPK, ERK1/2, and JNK) activation and upregulation of proinflammatory factors (IL-1ß, tumor necrosis factor-α and IL-6). Bex also reversed CCI-induced microglia activation in the ipsilateral spinal cord. Furthermore, bex treatment significantly upregulated MKP-1 in the spinal cord. These effects were completely abrogated by MKP-1 inhibitor BCI. These results indicated that bex relieved CCI-induced neuroinflammation and neuropathic pain by targeting MKP-1. Therefore, bex might be a potential agent for the treatment of neuropathic pain. PERSPECTIVE: Bex could relieve neuropathic pain behaviors in animals by reversing MKP-1 downregulation and MAPKs activation in the spinal cord. Therapeutic applications of bex may be extended beyond cutaneous T-cell lymphoma.


Subject(s)
Bexarotene/administration & dosage , Drug Delivery Systems/methods , Dual Specificity Phosphatase 1/metabolism , Inflammation Mediators/metabolism , Neuralgia/metabolism , Spinal Cord/metabolism , Animals , Constriction , Dose-Response Relationship, Drug , Dual Specificity Phosphatase 1/antagonists & inhibitors , Inflammation Mediators/antagonists & inhibitors , Male , Neuralgia/drug therapy , Rats , Spinal Cord/drug effects
3.
Am J Physiol Cell Physiol ; 317(6): C1183-C1193, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31532716

ABSTRACT

Dual-specificity phosphatase 1 (DUSP1) is differentially expressed in cumulus cells of different physiological states, but its specific function and mechanism of action remain unclear. In this study, we explored the effects of DUSP1 expression inhibition on cell cycle progression, proliferation, apoptosis, and lactate and cholesterol levels in cumulus cells and examined reactive oxygen species levels, mitochondrial function, autophagy, and the expression of key cytokine genes. The results showed that inhibition of DUSP1 in cumulus cells caused abnormal cell cycle progression, increased cell proliferation, decreased apoptosis rates, increased cholesterol synthesis and lactic acid content, and increased cell expansion. The main reason for these effects was that inhibition of DUSP1 reduced ROS accumulation, increased glutathione level and mitochondrial membrane potential, and reduced autophagy levels in cells. These results indicate that DUSP1 limits the biological function of bovine cumulus cells under normal physiological conditions and will greatly contribute to further explorations of the physiological functions of cumulus cells and the interactions of the cumulus-oocyte complex.


Subject(s)
Apoptosis/genetics , Cell Cycle/genetics , Cumulus Cells/metabolism , Dual Specificity Phosphatase 1/genetics , Mitochondria/physiology , Reactive Oxygen Species/metabolism , Animals , Autophagy/genetics , Cattle , Cell Proliferation/genetics , Cholesterol/metabolism , Cumulus Cells/cytology , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 1/metabolism , Female , Gene Expression Regulation , Glutathione/metabolism , Lactic Acid/metabolism , Membrane Potential, Mitochondrial/genetics , Oxidative Stress , Primary Cell Culture , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
4.
Bioorg Med Chem Lett ; 29(14): 1746-1748, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31103445

ABSTRACT

Structure based virtual screening attempts to discover DUSP1 inhibitors have yielded a scaffold featuring benzoxazole and acylthiourea pharmacophore. A series of its analogues were synthesized to explore structure activity relationship (SAR) of DUSP1 inhibition.


Subject(s)
Dual Specificity Phosphatase 1/antagonists & inhibitors , Humans , Structure-Activity Relationship
5.
Clin Cancer Res ; 25(13): 4117-4127, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30936125

ABSTRACT

PURPOSE: In neurofibromatosis type 1 (NF1) and in highly aggressive malignant peripheral nerve sheath tumors (MPNSTs), constitutively active RAS-GTP and increased MAPK signaling are important in tumorigenesis. Dual specificity phosphatases (DUSPs) are negative regulators of MAPK signaling that dephosphorylate p38, JNK, and ERK in different settings. Although often acting as tumor suppressors, DUSPs may also act as oncogenes, helping tumor cells adapt to high levels of MAPK signaling. We hypothesized that inhibiting DUSPs might be selectively toxic to cells from NF1-driven tumors. EXPERIMENTAL DESIGN: We examined DUSP gene and protein expression in neurofibroma and MPNSTs. We used small hairpin RNA (shRNA) to knock down DUSP1 and DUSP6 to evaluate cell growth, downstream MAPK signaling, and mechanisms of action. We evaluated the DUSP inhibitor, (E)-2-benzylidene-3-(cyclohexylamino)-2,3-dihydro-1H-inden-1-one (BCI), in MPNST cell lines and in cell-line and patient-derived MPNST xenografts. RESULTS: DUSP1 and DUSP6 are expressed in NF1-deleted tumors. Knockdown of DUSP1 and DUSP6, alone or in combination, reduced MPNST cell growth and led to ERK and JNK hyperactivation increasing downstream TP53 and p-ATM. The DUSP inhibitor, BCI, diminished the survival of NF1-deleted Schwann cells and MPNST cell lines through activation of JNK. In vivo, treatment of an established cell-line xenograft or a novel patient-derived xenograft (PDX) of MPNSTs with BCI increased ERK and JNK activation, caused tumor necrosis and fibrosis, and reduced tumor volume in one model. CONCLUSIONS: Targeting DUSP1 and DUSP6 genetically or with BCI effectively inhibits MPNST cell growth and promotes cell death, in vitro and in xenograft models. The data support further investigation of DUSP inhibition in MPNSTs.


Subject(s)
Antineoplastic Agents/pharmacology , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 6/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Nerve Sheath Neoplasms/metabolism , Nerve Sheath Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Animals , Cell Line, Tumor , DNA Copy Number Variations , Disease Models, Animal , Gene Knockdown Techniques , Humans , Mice , Neurofibromatosis 1/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
6.
Clin Cancer Res ; 24(17): 4201-4214, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29739788

ABSTRACT

Purpose: Loss of cell-cycle control is a hallmark of cancer, which can be targeted with agents, including cyclin-dependent kinase-4/6 (CDK4/6) kinase inhibitors that impinge upon the G1-S cell-cycle checkpoint via maintaining activity of the retinoblastoma tumor suppressor (RB). This class of drugs is under clinical investigation for various solid tumor types and has recently been FDA-approved for treatment of breast cancer. However, development of therapeutic resistance is not uncommon.Experimental Design: In this study, palbociclib (a CDK4/6 inhibitor) resistance was established in models of early stage, RB-positive cancer.Results: This study demonstrates that acquired palbociclib resistance renders cancer cells broadly resistant to CDK4/6 inhibitors. Acquired resistance was associated with aggressive in vitro and in vivo phenotypes, including proliferation, migration, and invasion. Integration of RNA sequencing analysis and phosphoproteomics profiling revealed rewiring of the kinome, with a strong enrichment for enhanced MAPK signaling across all resistance models, which resulted in aggressive in vitro and in vivo phenotypes and prometastatic signaling. However, CDK4/6 inhibitor-resistant models were sensitized to MEK inhibitors, revealing reliance on active MAPK signaling to promote tumor cell growth and invasion.Conclusions: In sum, these studies identify MAPK reliance in acquired CDK4/6 inhibitor resistance that promotes aggressive disease, while nominating MEK inhibition as putative novel therapeutic strategy to treat or prevent CDK4/6 inhibitor resistance in cancer. Clin Cancer Res; 24(17); 4201-14. ©2018 AACR.


Subject(s)
Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/genetics , Dual Specificity Phosphatase 1/genetics , MAP Kinase Kinase Kinases/genetics , Neoplasms/drug therapy , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Dual Specificity Phosphatase 1/antagonists & inhibitors , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase Kinases/antagonists & inhibitors , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation/drug effects , Piperazines/pharmacology , Pyridines/pharmacology , Retinoblastoma Protein/genetics , Sequence Analysis, RNA , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
J Innate Immun ; 10(3): 202-214, 2018.
Article in English | MEDLINE | ID: mdl-29455206

ABSTRACT

BACKGROUND: ß-Adrenergic agents suppress inflammation and may play an important role in posttraumatic infections. Mechanisms may include inhibition of MAP kinase signaling. We sought to determine whether MKP-1 contributed to catecholamine suppression of innate immunity and also wanted to know whether early catecholamine treatment after traumatic injury increases the risk of later nosocomial infection. METHODS: We performed experiments using THP-1 cells and peripheral blood mononuclear cells from healthy individuals. We exposed cells to epinephrine and/or LPS and measured inflammatory gene transcription and MAP kinase activation. We inhibited MKP-1 activity to determine its role in catecholamine-induced immune suppression. Finally, we studied injured subjects to determine whether early catecholamine treatment was associated with nosocomial infection. RESULTS: Epinephrine increases MKP-1 transcripts and protein and decreases LPS-induced p38 and JNK phosphorylation and TNF-α gene transcription. RNAi inhibition of MKP-1 at least partially restores LPS-induced TNF-α gene expression (p = 0.024). In the clinical cohort, subjects treated with ß-adrenergic agents had an increased risk of ventilator-associated pneumonia (aOR = 1.9; 95% CI = 1.3-2.6) and bacteremia (aOR = 1.5; 95% CI = 1.1-2.3). CONCLUSIONS: MKP-1 may have a role in catecholamine-induced suppression of innate immunity, and exogenous catecholamines might contribute to nosocomial infection risk.


Subject(s)
Adrenergic beta-Agonists/therapeutic use , Dual Specificity Phosphatase 1/metabolism , Immunity, Innate/drug effects , Wounds and Injuries/drug therapy , Adolescent , Adrenergic beta-Agonists/pharmacology , Adult , Bacteremia/epidemiology , Bacteremia/etiology , Child , Child, Preschool , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 1/genetics , Epinephrine/pharmacology , Female , Humans , Infant , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , Male , Middle Aged , Phosphorylation/drug effects , Pneumonia, Ventilator-Associated/epidemiology , Pneumonia, Ventilator-Associated/etiology , THP-1 Cells , Tumor Necrosis Factor-alpha/genetics , Vasoconstrictor Agents/adverse effects , Vasoconstrictor Agents/pharmacology , Young Adult
8.
Oncogene ; 36(43): 5939-5947, 2017 10 26.
Article in English | MEDLINE | ID: mdl-28650468

ABSTRACT

Understanding the mechanisms of platinum compound resistance, including cisplatin resistance, has important implications for improving cancer treatments. Previous studies identified a potential role for mitogen-activated protein kinase phosphatase-1 (MKP-1) in cisplatin resistance. This work focuses on the regulation of poly(ADP-ribose) polymerase-1 (PARP-1) expression by MKP-1. We found that MKP-1 overexpression stimulates PARP-1 and poly(ADP-ribose) (PAR) protein expression and cisplatin resistance while its downregulation suppresses PARP-1 and PAR protein expression and cisplatin resistance. Silencing MKP-1 promoted PARP-1 ubiquitination, which decreased PARP-1 protein levels. We also found that silencing c-Jun N-terminal kinase 1/2 (JNK1/2) decreased PARP-1 ubiquitination while increasing total PARP-1 protein levels. Furthermore, we showed that acquired cisplatin-resistant ovarian cancer cells expressed high levels of MKP-1 and PARP-1 proteins, and that silencing MKP-1 or PARP-1 increased cisplatin sensitivity in resistant cells. Notably, the pharmacologic inhibition of PARP activity restored cisplatin sensitivity in MKP-1 overexpressing cells. Thus, this work indicates that suppression of JNK1/2 activity by MKP-1 maintains PARP-1 levels and suggests that MKP-1-mediated cisplatin resistance can be bypassed by PARP-1 inhibition.


Subject(s)
Drug Resistance, Neoplasm/genetics , Dual Specificity Phosphatase 1/genetics , Ovarian Neoplasms/drug therapy , Poly (ADP-Ribose) Polymerase-1/genetics , Cell Line, Tumor , Cisplatin/administration & dosage , Dual Specificity Phosphatase 1/antagonists & inhibitors , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Proteolysis/drug effects
9.
Am J Physiol Cell Physiol ; 310(11): C921-30, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27053523

ABSTRACT

Vascular smooth muscle contraction is primarily regulated by phosphorylation of myosin light chain. There are also modulatory pathways that control the final level of force development. We tested the hypothesis that protein kinase C (PKC) and mitogen-activated protein (MAP) kinase modulate vascular smooth muscle activity via effects on MAP kinase phosphatase-1 (MKP-1). Swine carotid arteries were mounted for isometric force recording and subjected to histamine stimulation in the presence and absence of inhibitors of PKC [bisindolylmaleimide-1 (Bis)], MAP kinase kinase (MEK) (U0126), and MKP-1 (sanguinarine) and flash frozen for measurement of MAP kinase, PKC-potentiated myosin phosphatase inhibitor 17 (CPI-17), and caldesmon phosphorylation levels. CPI-17 was phosphorylated in response to histamine and was inhibited in the presence of Bis. Caldesmon phosphorylation levels increased in response to histamine stimulation and were decreased in response to MEK inhibition but were not affected by the addition of Bis. Inhibition of PKC significantly increased p42 MAP kinase, but not p44 MAP kinase. Inhibition of MEK with U0126 inhibited both p42 and p44 MAP kinase activity. Inhibition of MKP-1 with sanguinarine blocked the Bis-dependent increase of MAP kinase activity. Sanguinarine alone increased MAP kinase activity due to its effects on MKP-1. Sanguinarine increased MKP-1 phosphorylation, which was inhibited by inhibition of MAP kinase. This suggests that MAP kinase has a negative feedback role in inhibiting MKP-1 activity. Therefore, PKC catalyzes MKP-1 phosphorylation, which is reversed by MAP kinase. Thus the fine tuning of vascular contraction is due to the concerted effort of PKC, MAP kinase, and MKP-1.


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Muscle, Smooth, Vascular/enzymology , Protein Kinase C/metabolism , Vasoconstriction , Animals , Calmodulin-Binding Proteins/metabolism , Carotid Arteries/enzymology , Dual Specificity Phosphatase 1/antagonists & inhibitors , Feedback, Physiological , In Vitro Techniques , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Swine , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
11.
Biochem Biophys Res Commun ; 467(2): 254-60, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26435497

ABSTRACT

Dual-specificity phosphatases (DUSPs) show distinct substrate preferences for specific MAPKs. DUSPs sharing a substrate preference for ERK1/2 may be classified as inducible or constitutive. In contrast to the inducible DUSPs which also dephosphorylate p38 MAPK and JNK in the major inflammatory pathways, constitutive DUSP6 and DUSP7 are specific to ERK1/2 and have not been studied in microglia and other immune cells to date. In the present study, we differentiated mRNA expression profiles of inducible and constitutive DUSPs that dephosphorylate ERK1/2 in microglia. Lipopolysaccharide (LPS) at 1 ng/ml induced prompt phosphorylation of ERK1/2 with peak induction at 30 min. LPS induced expression of DUSP1, DUSP2, and DUSP5 within 60 min, whereas DUSP4 expression was induced more slowly. DUSP6 and DUSP7 exhibited constitutive basal expression, which decreased immediately after LPS stimulation but subsequently returned to basal levels. The expression of DUSP6 and DUSP7 was regulated inverse to the phosphorylation of ERK1/2 in LPS-stimulated microglia. Therefore, we next investigated the correlation between DUSP6 and DUSP7 expression and ERK1/2 phosphorylation in resting and LPS-stimulated microglia. Inhibition of the ERK1/2 pathway by PD98059 and FR180204 resulted in a decrease in DUSP6 and DUSP7 expression, both in resting and LPS-stimulated microglia. These inhibitors partially blocked the LPS-induced expression of DUSP1, DUSP2, and DUSP4, but had no effect on DUSP5. Finally, we examined the role of DUSP6 activity in the downregulation of ERK1/2 phosphorylation. BCI, an inhibitor of DUSP6, increased the phosphorylation of ERK1/2. However, pretreatment with BCI inhibited the LPS-induced phosphorylation of ERK1/2. These results demonstrate that constitutive DUPS6 and DUSP7 expression was downregulated inverse to the expression of inducible DUSPs and the phosphorylation of ERK1/2 in LPS-stimulated microglia. The expression of DUPS6 and DUSP7 was mediated by ERK1/2 activity both in resting and LPS-stimulated microglia. In turn, DUSP6 suppressed the basal phosphorylation of ERK1/2, but exerted no suppressive effect on LPS-induced phosphorylation. Although DUSP6 is acknowledged as a negative regulator of the ERK1/2 pathway, such roles of DUSP6 need to be examined further in activated microglia.


Subject(s)
Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 6/genetics , Microglia/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Animals , Animals, Newborn , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 1/metabolism , Dual Specificity Phosphatase 6/antagonists & inhibitors , Dual Specificity Phosphatase 6/metabolism , Flavonoids/pharmacology , Gene Expression Regulation , Isoenzymes/genetics , Isoenzymes/metabolism , Lipopolysaccharides/pharmacology , Microglia/cytology , Microglia/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyridazines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Mutat Res ; 778: 71-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26111828

ABSTRACT

Histone mark, H3S10 phosphorylation plays a dual role in a cell by maintaining relaxed chromatin for active transcription in interphase and condensed chromatin state in mitosis. The level of H3S10P has also been shown to alter on DNA damage; however, its cell cycle specific behavior and regulation during DNA damage response is largely unexplored. In the present study, we demonstrate G1 cell cycle phase specific reversible loss of H3S10P in response to IR-induced DNA damage is mediated by opposing activities of phosphatase, MKP1 and kinase, MSK1 of the MAP kinase pathway. We also show that the MKP1 recruits to the chromatin in response to DNA damage and correlates with the decrease of H3S10P, whereas MKP1 is released from chromatin during recovery phase of DDR. Furthermore, blocking of H3S10 dephosphorylation by MKP1 inhibition impairs DNA repair process and results in poor survival of WRL68 cells. Collectively, our data proposes a pathway regulating G1 cell cycle phase specific reversible reduction of H3S10P on IR induced DNA damage and also raises the possibility of combinatorial modulation of H3S10P with specific inhibitors to target the cancer cells in G1-phase of cell cycle.


Subject(s)
DNA Damage , DNA Repair/physiology , Dual Specificity Phosphatase 1/physiology , G1 Phase/physiology , Histones/metabolism , Point Mutation , Protein Processing, Post-Translational/physiology , Benzophenanthridines/pharmacology , Cell Line , Chromatin/genetics , Chromatin/ultrastructure , Colony-Forming Units Assay , Comet Assay , DNA/genetics , DNA/radiation effects , DNA Repair/genetics , Dual Specificity Phosphatase 1/antagonists & inhibitors , Gamma Rays , Histones/genetics , Histones/physiology , Humans , Isoquinolines/pharmacology , MAP Kinase Signaling System , Nucleosomes/radiation effects , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/physiology , Sulfonamides/pharmacology
13.
Genet Mol Res ; 14(2): 4495-504, 2015 May 04.
Article in English | MEDLINE | ID: mdl-25966222

ABSTRACT

In this study, we investigated the antidepressant effects of hippocampal neuron administration of ß-asarone, a selective mitogen-activated protein kinase phosphatase-1 inhibitor, in a rat model of depression. Our previous studies showed that the extracellular signal-regulated kinase signaling pathway and brain-derived neurotrophic factor expression, which is regulated by extracellular signal-regulated kinase, are key links in the biological mechanism of depression. Mitogen-activated protein kinase phosphatase-1 (MKP-1) is a negative regulatory protein of extracellular signal-regulated kinase signaling pathways. In this study, we explored the regulation of MKP-1 by ß-asarone in producing an antidepressant effect.


Subject(s)
Acorus , Anisoles/pharmacology , Antidepressive Agents/pharmacology , Depression/drug therapy , Dual Specificity Phosphatase 1/antagonists & inhibitors , Allylbenzene Derivatives , Animals , Depression/genetics , Depression/metabolism , Dual Specificity Phosphatase 1/genetics , Gene Expression , Male , Rats , Signal Transduction
14.
Diabetes ; 64(12): 4285-97, 2015 12.
Article in English | MEDLINE | ID: mdl-25858560

ABSTRACT

Activation of AMPK suppresses inflammation, but the underlying mechanisms remain poorly understood. This study was designed to characterize the molecular mechanisms by which AMPK suppresses vascular inflammation. In cultured human aortic smooth muscle cells, pharmacologic or genetic activation of AMPK inhibited the signal transducer and activator of transcription-1 (STAT1), while inhibition of AMPK had opposite effects. Deletion of AMPKα1 or AMPKα2 resulted in activation of STAT1 and in increases in proinflammatory mediators, both of which were attenuated by administration of STAT1 small interfering RNA or fludarabine, a selective STAT1 inhibitor. Moreover, AMPK activation attenuated the proinflammatory actions induced by STAT1 activators such as interferon-γ and angiotensin II (AngII). Mechanistically, we found that AMPK activation increased, whereas AMPK inhibition decreased, the levels of mitogen-activated protein kinase phosphatase-1 (MKP-1), an inducible nuclear phosphatase, by regulating proteasome-dependent degradation of MKP-1. Gene silencing of MKP-1 increased STAT1 phosphorylation and prevented 5-aminoimidazole-4-carboxyamide ribonucleoside-reduced STAT1 phosphorylation. Finally, we found that infusion of AngII caused a more severe inflammatory response in AMPKα2 knockout mouse aortas, all of which were suppressed by chronic administration of fludarabine. We conclude that AMPK activation suppresses STAT1 signaling and inhibits vascular inflammation through the upregulation of MKP-1.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Dual Specificity Phosphatase 1/metabolism , Muscle, Smooth, Vascular/metabolism , STAT1 Transcription Factor/metabolism , Vasculitis/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/genetics , Angiotensin II/adverse effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aorta, Thoracic , Cells, Cultured , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 1/chemistry , Dual Specificity Phosphatase 1/genetics , Enzyme Activation/drug effects , Humans , Interferon-gamma/adverse effects , MAP Kinase Signaling System/drug effects , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/pathology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , RNA Interference , Random Allocation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/genetics , Vasculitis/chemically induced , Vasculitis/immunology , Vasculitis/pathology
15.
Cell Biol Int ; 39(1): 113-20, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25044272

ABSTRACT

Chonic hypoxia, smooth muscle cell (SMC) proliferation and vascular remodeling are hallmark features of pathogenic pulmonary artery hypertension. MicroRNAs (miRNAs), endogenously expressed small noncoding RNAs, regulate gene expression at the post-transcriptional level. MiR-210 is considered a "master miRNA" in the control of diverse functions in hypoxic cells and tissues and has a cytoprotective function in pulmonary artery SMCs during hypoxic stress. MiR-210 is also upregulated in lung tissue of chonically hypoxic mice suffering from pulmonary hypertension. Jin et al. () showed that mice deficient in mitogen-activated protein kinase phosphatase 1 (MKP-1) had severe hypoxia-induced pulmonary hypertension, so MKP-1 may be important in the progression of hypoxic pulmonary artery hypertension. We investigated the possible interactions between miR-210 and MKP-1 and the effect on cell proliferation in hypoxic human pulmonary artery SMCs (hPASMCs). miR-210 was significantly increased in cultured hPASMCs exposed to 1% O2 hypoxia for 48 h, as was MKP-1 mRNA and protein expression. Furthermore, inhibiting miR-210 expression increased MKP-1 mRNA and protein expression in hPASMCs and decreased cell proliferation under hypoxia. Conversely, overexpressing miR-210 prevented hypoxia-induced MKP-1 expression with no effect on cell proliferation. siRNA knockdown of MKP-1 abolished the miR-210-inhibition prevention of cell proliferation under hypoxia. MKP-1 is a target of miR-210 and could mediate the negative regulation of miR-210 inhibition on hypoxic hPASMCs.


Subject(s)
Cell Hypoxia , Dual Specificity Phosphatase 1/metabolism , MicroRNAs/metabolism , Cell Line , Cell Proliferation , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 1/genetics , Humans , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Up-Regulation
16.
Cancer Lett ; 357(1): 265-278, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25444924

ABSTRACT

Maintaining cellular redox homeostasis is imperative for the survival and normal functioning of cells. This study describes the role and regulation of MAPKinases in oxidative stress mediated apoptosis. Plumbagin, a vitamin K3 analog and a pro-oxidant, was employed and it induced apoptosis in both mouse and human T-cell lymphoma cell lines via increased oxidative stress, caspase activity and loss of mitochondrial membrane potential. The pro-oxidant and cytotoxic effects of plumbagin were sensitive to antioxidants indicating a decisive role of cellular redox balance. Plumbagin induced persistent activation of JNK and pharmacological inhibition as well as shRNA-mediated JNK knock-down rescued cells from plumbagin-induced apoptosis. Further, plumbagin induced cytochrome c release, FasL expression and Bax levels via activation of JNK pathway. Exposure of lymphoma cells to plumbagin led to inhibition of total and specific phosphatase activity, increased total protein S-glutathionylation and induced glutathionylation of dual specific phosphatase- 1 and 4 (MKP-1 and MKP-2). The in vivo anti-tumor efficacy of plumbagin was demonstrated using a mouse model. In conclusion, oxidative stress mediated tumor cytotoxicity operates through sustained JNK activation via a novel redox-mediated regulation of MKP-1 and MKP-2.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual-Specificity Phosphatases/antagonists & inhibitors , Glutathione/metabolism , Lymphoma, T-Cell/drug therapy , Mitogen-Activated Protein Kinase Phosphatases/antagonists & inhibitors , Naphthoquinones/pharmacology , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Disease Models, Animal , Dual Specificity Phosphatase 1/metabolism , Dual-Specificity Phosphatases/metabolism , Enzyme Activation , Humans , Jurkat Cells , Lymphoma, T-Cell/enzymology , Lymphoma, T-Cell/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Models, Molecular , Phosphorylation , Random Allocation
17.
Cell Signal ; 26(10): 2167-74, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25007998

ABSTRACT

CaMKs are a widely distributed family of kinases with multiple and often cell specific effects on intracellular signal transduction pathway. In endothelial cells, it has been recognized a role for CamKII in several pathways such as eNOS activation and nitric oxide production. It is not clear though, whether CaMKII interfere with other endothelial cell functions such as ERK activation and cell proliferation. We explored this issue in primary cultured rat endothelial cells and we evaluated the effect on endothelial cell proliferation and DNA synthesis. CaMKII inhibition through Cantide, conducted into the cell through Antoennapedia (ANT-CN), showed positive effects on proliferation and H(3)-thimdine incorporation similar to insulin stimulation. Accordingly, both CaMKII pharmacological inhibition and silencing through shRNA produced activation of the p44/42 MAPK. These observations leaded to the hypothesis that CamKII could regulate p44/p42 by interfering with specific ERK phosphatases. Indeed, we found that CaMKII interacts and protect the dual specific phosphatase MKP-1 from proteasome mediated degradation while this complex is disrupted by CaMKII inhibitors. This study reveals that CaMKII, besides phosphorylation through the known ras-raf-mek pathway, can regulate also dephosphorylation of p44/p42 by modulation of MKP-1 level. This novel finding opens to a novel scenario in regulation of endothelial cell functions.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dual Specificity Phosphatase 1/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Aorta/cytology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Proliferation/drug effects , Cells, Cultured , DNA/biosynthesis , Dual Specificity Phosphatase 1/antagonists & inhibitors , Endothelial Cells/cytology , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , Insulin/pharmacology , Leupeptins/pharmacology , MAP Kinase Signaling System/drug effects , Marine Toxins , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oxazoles/pharmacology , Phosphorothioate Oligonucleotides/pharmacology , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/chemistry , Protein Binding , Proto-Oncogene Proteins c-raf/metabolism , Rats
18.
Neurosci Lett ; 569: 49-54, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24686178

ABSTRACT

Mitogen-activated protein kinase phosphatase 1 (MKP-1) is shown to negatively regulate MAPK signaling in various peripheral tissues as well as the central nervous system such as cortex, striatum and hippocampus. In this study, we examined whether MKP-1 regulates MAPK signaling in the mouse hypothalamus. Intraperitoneal injection of TNFα significantly increased MKP-1 mRNA expression in paraventricular and arcuate nuclei in the hypothalamus. TNFα treatment induced increases in MKP-1 expression at both mRNA and protein levels, accompanied by the inactivation of MAPK signaling in mouse hypothalamic explants. Inhibition of MKP-1 by its inhibitor or siRNA increased MAPK activity in the explants. Our data indicate that MKP-1 negatively regulates MAPK signaling in the mouse hypothalamus.


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Hypothalamus/metabolism , MAP Kinase Signaling System , Animals , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 1/genetics , Male , Mice, Inbred C57BL , Organ Culture Techniques , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Tumor Necrosis Factor-alpha/pharmacology
19.
Chem Biol Drug Des ; 84(2): 158-68, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24605883

ABSTRACT

Plasmodium falciparum, the causative agent of malaria, contributes to significant morbidity and mortality worldwide. Forward genetic analysis of the blood-stage asexual cycle identified the putative phosphatase from PF3D7_1305500 as an important element of intraerythrocytic development expressed throughout the life cycle. Our preliminary evaluation identified it as an atypical mitogen-activated protein kinase phosphatase. Additional bioinformatic analysis delineated a conserved signature motif and three residues with potential importance to functional activity of the atypical dual-specificity phosphatase domain. A homology model of the dual-specificity phosphatase domain was developed for use in high-throughput in silico screening of the available library of antimalarial compounds from ChEMBL-NTD. Seven compounds from this set with predicted affinity to the active site were tested against in vitro cultures, and three had reduced activity against a ∆PF3D7_1305500 parasite, suggesting PF3D7_1305500 is a potential target of the selected compounds. Identification of these compounds provides a novel starting point for a structure-based drug discovery strategy that moves us closer toward the discovery of new classes of clinical antimalarial drugs. These data suggest that mitogen-activated protein kinase phosphatases represent a potentially new class of P. falciparum drug target.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Dual Specificity Phosphatase 1/antagonists & inhibitors , Plasmodium falciparum/enzymology , Amino Acid Sequence , Drug Discovery , Dual Specificity Phosphatase 1/chemistry , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Humans , Malaria, Falciparum/drug therapy , Models, Molecular , Molecular Sequence Data , Phylogeny , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Sequence Alignment
20.
Cell Death Dis ; 4: e521, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23449457

ABSTRACT

Sporadic occurrence of transformed tumor cells is under the surveillance of the host immune system and such cells are effectively eliminated by immune-mediated cell death. During tumor progression, the antitumor effects of the tumor microenvironment are suppressed by diverse immunosuppressive mechanisms. In this research, we suggest novel immune evasion strategy of tumor cells through a transforming growth factor (TGF)-ß1- and hypoxia-dependent mechanism. Experimental results showed that TGF-ß1 and hypoxia induced mitogen-activated protein kinase phosphatase (MKP)-1 expression within 1 h, resulting in attenuation of c-Jun N-terminal kinase (JNK) phosphorylation and subsequent death receptor-mediated cell death. In addition, analysis of microarray data and immunostaining of MKP-1 in hepatocellular carcinoma (HCC) patient samples revealed that expression of MKP-1 is notably higher in tumors than in normal tissues, implying that MKP-1-dependent suppression of immune-mediated cell death takes place only in the tumor. To prove that MKP-1 can act as a mediator of immune escape by tumors, we determined whether chemo-resistance against several anticancer drugs could be overcome by knockdown of MKP-1. Cytotoxic assays showed that chemotherapy with siRNA targeting MKP-1 was significantly more effective than chemotherapy in the presence of MKP-1. Thus, we conclude that TGF-ß1 and hypoxia ensure tumor cell survival and growth through expression of MKP-1.


Subject(s)
Apoptosis/drug effects , Cell Hypoxia , Dual Specificity Phosphatase 1/metabolism , Receptors, Death Domain/metabolism , Transforming Growth Factor beta1/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Databases, Factual , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 1/genetics , Hep G2 Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Transgenic , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Smad2 Protein/antagonists & inhibitors , Smad2 Protein/genetics , Smad2 Protein/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...