Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Med Chem ; 65(9): 6690-6709, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35442672

ABSTRACT

Tumor necrosis factor α (TNF-α) has been demonstrated to be a therapeutic target for autoimmune diseases. However, this biological therapy exhibits some inevitable disadvantages, such as risk of infection. Thus, small-molecule alternatives by targeting TNF-α production signaling pathway are still in demand. Herein, we describe the design, synthesis, and structure-activity relationships of 3-aryindanone compounds regarding their modulation of TNF-α production. Among them, (R)-STU104 exhibited the most potent inhibitory activity on TNF-α production, which suppressed the TAK1/MKK3/p38/MnK1/MK2/elF4E signal pathways through binding with MKK3 and disrupting the TAK1 phosphorylating MKK3. As a result, (R)-STU104 demonstrated remarkable dose-effect relationships on both acute and chronic mouse UC models. In addition to its good pharmacokinetic (PK) and safety profile, (R)-STU104 showed better anti-UC efficacy in vivo at 10 mg/kg/d than mesalazine at the dose of 50 mg/kg/d. These results suggested that TAK1-MKK3 interaction inhibitors could be potentially utilized for the treatment of UC.


Subject(s)
Colitis, Ulcerative , MAP Kinase Kinase 3 , MAP Kinase Kinase Kinases , Protein Kinase Inhibitors , Tumor Necrosis Factor-alpha , Animals , Colitis, Ulcerative/drug therapy , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Mice , Protein Kinase Inhibitors/pharmacology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Bioorg Med Chem ; 45: 116324, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34333394

ABSTRACT

The transcription master regulator MYC plays an essential role in regulating major cellular programs and is a well-established therapeutic target in cancer. However, MYC targeting for drug discovery is challenging. New therapeutic approaches to control MYC-dependent malignancy are urgently needed. The mitogen-activated protein kinase kinase 3 (MKK3) binds and activates MYC in different cell types, and disruption of MKK3-MYC protein-protein interaction may provide a new strategy to target MYC-driven programs. However, there is no perturbagen available to interrogate and control this signaling arm. In this study, we assessed the drugability of the MKK3-MYC complex and discovered the first chemical tool to regulate MKK3-mediated MYC activation. We have designed a short 44-residue inhibitory peptide and developed a cell lysate-based time-resolved fluorescence resonance energy transfer (TR-FRET) assay to discover the first small molecule MKK3-MYC PPI inhibitor. We have optimized and miniaturized the assay into an ultra-high-throughput screening (uHTS) 1536-well plate format. The pilot screen of ~6,000 compounds of a bioactive chemical library followed by multiple secondary and orthogonal assays revealed a quinoline derivative SGI-1027 as a potent inhibitor of MKK3-MYC PPI. We have shown that SGI-1027 disrupts the MKK3-MYC complex in cells and in vitro and inhibits MYC transcriptional activity in colon and breast cancer cells. In contrast, SGI-1027 does not inhibit MKK3 kinase activity and does not interfere with well-known MKK3-p38 and MYC-MAX complexes. Together, our studies demonstrate the drugability of MKK3-MYC PPI, provide the first chemical tool to interrogate its biological functions, and establish a new uHTS assay to enable future discovery of potent and selective inhibitors to regulate this oncogenic complex.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , MAP Kinase Kinase 3/antagonists & inhibitors , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , MAP Kinase Kinase 3/chemistry , Molecular Docking Simulation , Molecular Structure , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-myc/chemistry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
3.
Int J Mol Sci ; 22(14)2021 Jul 10.
Article in English | MEDLINE | ID: mdl-34299039

ABSTRACT

Zinc chloride is known to be effective in combatting hepatitis A virus (HAV) infection, and zinc ions seem to be especially involved in Toll-like receptor (TLR) signaling pathways. In the present study, we examined this involvement in human hepatoma cell lines using a human TLR signaling target RT-PCR array. We also observed that zinc chloride inhibited mitogen-activated protein kinase kinase 3 (MAP2K3) expression, which could downregulate HAV replication in human hepatocytes. It is possible that zinc chloride may inhibit HAV replication in association with its inhibition of MAP2K3. In that regard, this study set out to determine whether MAP2K3 could be considered a modulating factor in the development of the HAV pathogen-associated molecular pattern (PAMP) and its triggering of interferon-ß production. Because MAP2K3 seems to play a role in antiviral immunity against HAV infection, it is a promising target for drug development. The inhibition of MAP2K3 may also prevent HAV patients from developing a severe hepatitis A infection.


Subject(s)
Carcinoma, Hepatocellular/virology , Chlorides/pharmacology , Hepatitis A/complications , Hepatocytes/virology , Liver Neoplasms/virology , MAP Kinase Kinase 3/antagonists & inhibitors , Virus Replication , Zinc Compounds/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/enzymology , Hepatitis A/virology , Hepatitis A virus/isolation & purification , Hepatocytes/drug effects , Hepatocytes/enzymology , Host-Pathogen Interactions , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/enzymology , Tumor Cells, Cultured
4.
J Alzheimers Dis ; 71(2): 671-684, 2019.
Article in English | MEDLINE | ID: mdl-31424393

ABSTRACT

BACKGROUND: Oligomeric amyloid-ß peptide (Aß) is associated with dysfunctional neuronal networks and neuronal loss in the development of Alzheimer's disease (AD). Ischemic postconditioning protects against post-ischemic excitotoxicity, oxidative stress, and inflammatory process that have also been implicated in the pathogenesis of AD. Evaluating the roles of ischemic postconditioning in oligomeric Aß-induced neurotoxicity and underlying signal events may provide potential strategy for medical therapy in AD. OBJECTIVES: The aim of the present study was to explore whether and how a brief ischemic postconditioning protects against Aß neurotoxicity in rat hippocampus. METHODS: Oligomeric Aß25-35 (20 nmol/rat) or Aß1-42 (5 nmol/rat) was infused by intracerebroventricular injection in adult male Sprague-Dawley rats. Ischemic postconditioning, a brief episode of global brain ischemia (3 min), was conducted at 1, 3, or 7 days after Aß treatment, respectively. RESULTS: A brief ischemic postconditioning reduced neuronal loss and inhibited the activation of MLK3, MKK3/6, and P38MAPKs in rat hippocampal CA1 and CA3 subfields after Aß oligomer infusion. An N-methyl-D-aspartate (NMDA) receptor antagonist amantadine, but not non-NMDA receptor antagonist CNQX, reversed the MLK3-MKK3/6-P38MAPK signal events and beneficial effect of ischemic postconditioning on neuronal survival. Such reversion was also realized by NVP-AAM077, a GluN2A-subunit-selective NMDA receptor antagonist. Moreover, posttreatment with low doses of NMDA (5 nmol-40 nmol/rat) suppressed the Aß-induced P38MAPK signaling and imitated the neuroprotection of ischemic postconditioning against Aß neurotoxicity. CONCLUSIONS: Ischemic postconditioning provides neuroprotection against Aß neurotoxicity by moderate upregulation of NMDA receptor signaling, especially GluN2A-containing NMDA receptor pathway, and thereafter downregulation of MLK3-MKK3/6-P38MAPK signal events.


Subject(s)
Hippocampus/metabolism , Ischemic Postconditioning/methods , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase 6/metabolism , MAP Kinase Kinase Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Amyloid beta-Peptides/toxicity , Animals , Down-Regulation/drug effects , Down-Regulation/physiology , Hippocampus/drug effects , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 6/antagonists & inhibitors , MAP Kinase Kinase Kinases/antagonists & inhibitors , Male , Peptide Fragments/toxicity , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinase Kinase 11
5.
Br J Cancer ; 120(9): 941-951, 2019 04.
Article in English | MEDLINE | ID: mdl-30944457

ABSTRACT

BACKGROUND: Mitogen-activated protein kinases (MEK 1/2) are central components of the RAS signalling pathway and are attractive targets for cancer therapy. These agents continue to be investigated in KRAS mutant colon cancer but are met with significant resistance. Clinical investigations have demonstrated that these strategies are not well tolerated by patients. METHODS: We investigated a biomarker of response for MEK inhibition in KRAS mutant colon cancers by LC-MS/MS analysis. We tested the MEK inhibitor in PIK3CA wild(wt) and mutant(mt) colon cancer cells. In addition, we tested the combinational effects of MEK and TNKS inhibitor in vitro and in vivo. RESULTS: We identified ß-catenin, a key mediator of the WNT pathway, in response to MEK inhibitor. MEK inhibition led to a decrease in ß-catenin in PIK3CA wt colon cancer cells but not in mt. Tumour regression was promoted by combination of MEK inhibition and NVP-TNS656, which targets the WNT pathway. Furthermore, inhibition of MEK promoted tumour regression in colon cancer patient-derived xenograft models expressing PIK3CA wt. CONCLUSIONS: We propose that inhibition of the WNT pathway, particularly ß-catenin, may bypass resistance to MEK inhibition in human PIK3CA mt colon cancer. Therefore, we suggest that ß-catenin is a potential predictive marker of MEK inhibitor resistance.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , beta Catenin/metabolism , Acetamides/pharmacology , Animals , Biomarkers, Pharmacological/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/metabolism , Colonic Neoplasms/metabolism , Drug Resistance, Viral , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 3/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , Pyrimidinones/pharmacology , Xenograft Model Antitumor Assays , beta Catenin/antagonists & inhibitors
6.
Article in English | MEDLINE | ID: mdl-30348669

ABSTRACT

Cystic echinococcosis is a zoonosis caused by the larval stage of Echinococcus granulosussensu lato There is an urgent need to develop new drugs for the treatment of this disease. In this study, we identified two new members of mitogen-activated protein kinase (MAPK) cascades, MKK3/6 and MEK1/2 homologs (termed EgMKK1 and EgMKK2, respectively), from E. granulosussensu stricto Both EgMKK1 and EgMKK2 were expressed at the larval stages. As shown by yeast two-hybrid and coimmunoprecipitation analyses, EgMKK1 interacted with the previously identified Egp38 protein but not with EgERK. EgMKK2, on the other hand, interacted with EgERK. In addition, EgMKK1 and EgMKK2 displayed kinase activity toward the substrate myelin basic protein. When sorafenib tosylate, PD184352, or U0126-ethanol (EtOH) was added to the medium for in vitro culture of E. granulosus protoscoleces (PSCs) or cysts, an inhibitory and cytolytic effect was observed via suppressed phosphorylation of EgMKKs and EgERK. Nonviability of PSCs treated with sorafenib tosylate or U0126-EtOH, and not with PD184352, was confirmed through bioassays, i.e., inoculation of treated and untreated protoscoleces into mice. In vivo treatment of E. granulosussensu stricto-infected mice with sorafenib tosylate or U0126-EtOH for 4 weeks demonstrated a reduction in parasite weight, but the results did not show a significant difference. In conclusion, the MAPK cascades were identified as new targets for drug development, and E. granulosus was efficiently inhibited by their inhibitors in vitro The translation of these findings into in vivo efficacy requires further adjustment of treatment regimens using sorafenib tosylate or, possibly, other kinase inhibitors.


Subject(s)
Benzamides/pharmacology , Butadienes/pharmacology , Echinococcosis/drug therapy , Echinococcus granulosus/drug effects , MAP Kinase Signaling System/drug effects , Nitriles/pharmacology , Sorafenib/pharmacology , Animals , Echinococcosis/parasitology , Echinococcosis/pathology , Echinococcus granulosus/metabolism , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 6/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Phosphorylation/drug effects
7.
Cancer Lett ; 442: 126-136, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30391783

ABSTRACT

Gossypetin as a hexahydroxylated flavonoid found in many flowers and Hibiscus. It exerts various pharmacological activities, including antioxidant, antibacterial and anticancer activities. However, the anticancer capacity of gossypetin has not been fully elucidated. In this study, gossypetin was found to inhibit anchorage-dependent and -independent growth of esophageal cancer cells. To identify the molecular target(s) of gossypetin, various signaling protein kinases were screened and results indicate that gossypetin strongly attenuates the MKK3/6-p38 signaling pathway by directly inhibiting MKK3 and MKK6 protein kinase activity in vitro. Mechanistic investigations showed that arginine-61 in MKK6 is critical for binding with gossypetin. Additionally, the inhibition of cell growth by gossypetin is dependent on the expression of MKK3 and MKK6. Gossypetin caused G2 phase cell cycle arrest and induced intrinsic apoptosis by activating caspases 3 and 7 and increasing the expression of BAX and cytochrome c. Notably, gossypetin suppressed patient-derived esophageal xenograft tumor growth in an in vivo mouse model. Our findings suggest that gossypetin is an MKK3 and MKK6 inhibitor that could be useful for preventing or treating esophageal cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Flavonoids/pharmacology , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 6/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Binding Sites , Cell Line, Tumor , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/enzymology , Esophageal Squamous Cell Carcinoma/pathology , Female , Flavonoids/chemistry , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase 6/chemistry , MAP Kinase Kinase 6/metabolism , Mice, SCID , Molecular Docking Simulation , Protein Conformation , Protein Kinase Inhibitors/chemistry , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
8.
J Mol Neurosci ; 65(1): 43-53, 2018 May.
Article in English | MEDLINE | ID: mdl-29696468

ABSTRACT

The mechanism contributing to blood-brain barrier (BBB) disruption, involved in poststroke edema and hemorrhagic transformation, is important but elusive. We investigated microRNA-21 (miR-21)-mediated mechanism in the disruption of BBB following cerebral ischemia-reperfusion (I/R) injury. Rats with cerebral I/R injury were prepared after middle cerebral artery occlusion and subsequent reperfusion. The underlying regulatory mechanisms of miR-382 were investigated with treatment of miR-382 mimics, miR-382 inhibitors, or SB203580 (an inhibitor of the MAPK signaling pathway) prior to I/R modeling. Compared with sham-operated rats, rats following I/R showed increased Longa's scores, ischemic hemisphere volume, cerebral infarct volume, EB content in brain tissues, enhanced levels of p38, iNOS, and MMP-9. The ectopic expression of miR-21 by mimics and MAPK signaling inhibition by SB203580 reduced Longa's scores, ischemic hemisphere volume, cerebral infarct volume, EB content in brain tissues, decreased levels of p38, MAP2K3, iNOS, and MMP-9. The luciferase activity determination showed miR-21 bound to MAP2K3 in its 3'UTR. miR-21 downregulation mediated by inhibitors appeared to yield an opposed trend. We also found that MAPK signaling inhibition by SB203580 could rescue rats with treatment of miR-382 inhibitors. The study highlights the neuroprotective role of MiR-21 during cerebral I/R injury and its preventive effect against BBB disruption by blocking the MAPK signaling pathway via targeted inhibition of MAP2K3, potentially opening a novel therapeutic avenue for the treatment of cerebral ischemia.


Subject(s)
Blood-Brain Barrier/metabolism , Infarction, Middle Cerebral Artery/metabolism , MAP Kinase Signaling System , MicroRNAs/genetics , Animals , HEK293 Cells , Humans , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 3/metabolism , Male , Matrix Metalloproteinase 9/metabolism , MicroRNAs/metabolism , Nitric Oxide Synthase Type II/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Int J Mol Sci ; 17(11)2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27879664

ABSTRACT

Chloranthalactone B (CTB), a lindenane-type sesquiterpenoid, was obtained from the Chinese medicinal herb Sarcandra glabra, which is frequently used as a remedy for inflammatory diseases. However, the anti-inflammatory mechanisms of CTB have not been fully elucidated. In this study, we investigated the molecular mechanisms underlying these effects in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. CTB strongly inhibited the production of nitric oxide and pro-inflammatory mediators such as prostaglandin E2, tumor necrosis factor α (TNF-α), interleukin-1ß (IL-1ß), and IL-6 in RAW264.7 cells stimulated with LPS. A reverse-transcription polymerase chain reaction assay and Western blot further confirmed that CTB inhibited the expression of inducible nitric oxide synthase, cyclooxygenase-2, TNF-α, and IL-1ß at the transcriptional level, and decreased the luciferase activities of activator protein (AP)-1 reporter promoters. These data suggest that inhibition occurred at the transcriptional level. In addition, CTB blocked the activation of p38 mitogen-activated protein kinase (MAPK) but not c-Jun N-terminal kinase or extracellular signal-regulated kinase 1/2. Furthermore, CTB suppressed the phosphorylation of MKK3/6 by targeting the binding sites via formation of hydrogen bonds. Our findings clearly show that CTB inhibits the production of inflammatory mediators by inhibiting the AP-1 and p38 MAPK pathways. Therefore, CTB could potentially be used as an anti-inflammatory agent.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Lactones/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Macrophages/drug effects , Sesquiterpenes/pharmacology , Transcription Factor AP-1/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Line , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/antagonists & inhibitors , Dinoprostone/biosynthesis , Gene Expression Regulation , Inflammation/prevention & control , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 3/chemistry , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase 6/antagonists & inhibitors , MAP Kinase Kinase 6/chemistry , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Models, Molecular , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Signal Transduction , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription, Genetic , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Bioorg Med Chem Lett ; 26(3): 1086-1089, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26704264

ABSTRACT

The MAPK signaling cascade, comprised of several linear and intersecting pathways, propagates signaling into the nucleus resulting in cytokine and chemokine release. The Map Kinase Kinase isoforms 3 and 6 (MKK3 and MKK6) are responsible for the phosphorylation and activation of p38, and are hypothesized to play a key role in regulating this pathway without the redundancy seen in downstream effectors. Using FBDD, we have discovered efficient and selective inhibitors of MKK3 and MKK6 that can serve as tool molecules to help further understand the role of these kinases in MAPK signaling, and the potential impact of inhibiting kinases upstream of p38.


Subject(s)
Drug Design , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 6/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Binding Sites , Humans , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase 6/metabolism , MAP Kinase Signaling System/drug effects , Molecular Docking Simulation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , U937 Cells , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Free Radic Biol Med ; 83: 139-48, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25697779

ABSTRACT

Mitochondria are increasingly recognized as drivers of inflammatory responses. MAP kinase kinase 3 (MKK3), a dual-specificity protein kinase, is activated in inflammation and in turn activates p38 MAP kinase signaling. Here we show that MKK3 influences mitochondrial function and acts as a critical mediator of inflammation. MKK3-deficient (MKK3(-/-)) mice and bone marrow-derived macrophages (BMDMs) secreted smaller amounts of cytokines than wild type (WT) after lipopolysaccharide (LPS) exposure. There was improved mitochondrial function, as measured by basal oxygen consumption rate, mitochondrial membrane potential, and ATP production, in MKK3(-/-) BMDMs. After LPS exposure, MKK3(-/-) BMDMs did not show a significant increase in cellular reactive oxygen species production or in mitochondrial superoxide compared to WT. Activation of two important inflammatory mediators, i.e., the nuclear translocation of NF-κB and caspase-1 activity (a key component of the inflammasome), was lower in MKK3(-/-) BMDMs. p38 and JNK activation was lower in MKK3(-/-) BMDMs compared to WT after exposure to LPS. Knockdown of MKK3 by siRNA in wild-type BMDMs improved mitochondrial membrane potential, reduced LPS-induced caspase-1 activation, and attenuated cytokine secretion. Our studies establish MKK3 as a regulator of mitochondrial function and inflammatory responses to LPS and suggest that MKK3 may be a therapeutic target in inflammatory disorders such as sepsis.


Subject(s)
Bone Marrow/immunology , Inflammation/immunology , MAP Kinase Kinase 3/physiology , Macrophages/immunology , Mitochondria/immunology , Animals , Blotting, Western , Bone Marrow/drug effects , Bone Marrow/metabolism , Cells, Cultured , Cytokines/metabolism , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/pharmacology , MAP Kinase Kinase 3/antagonists & inhibitors , Macrophages/drug effects , Macrophages/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Phosphorylation/drug effects , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
13.
Cell Death Dis ; 6: e1621, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25633290

ABSTRACT

Mitogen-activated protein kinase kinase 3 (MAP2K3, MKK3) is a member of the dual specificity protein kinase group that belongs to the MAP kinase kinase family. This kinase is activated by mitogenic or stress-inducing stimuli and participates in the MAP kinase-mediated signaling cascade, leading to cell proliferation and survival. Several studies highlighted a critical role for MKK3 in tumor progression and invasion, and we previously identified MKK3 as transcriptional target of mutant (mut) p53 to sustain cell proliferation and survival, thus rendering MKK3 a promising target for anticancer therapies. Here, we found that targeting MKK3 with RNA interference, in both wild-type (wt) and mutp53-carrying cells, induced endoplasmic reticulum stress and autophagy that, respectively, contributed to stabilize wtp53 and degrade mutp53. MKK3 depletion reduced cancer cell proliferation and viability, whereas no significant effects were observed in normal cellular context. Noteworthy, MKK3 depletion in combination with chemotherapeutic agents increased tumor cell response to the drugs, in both wtp53 and mutp53 cancer cells, as demonstrated by enhanced poly (ADP-ribose) polymerase cleavage and reduced clonogenic ability in vitro. In addition, MKK3 depletion reduced tumor growth and improved biological response to chemotherapeutic in vivo. The overall results indicate MKK3 as a novel promising molecular target for the development of more efficient anticancer treatments in both wtp53- and mutp53-carrying tumors.


Subject(s)
MAP Kinase Kinase 3/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/enzymology , Animals , Autophagy , Cell Line , Cell Proliferation , Cell Survival , Endoplasmic Reticulum Stress , Female , Humans , MAP Kinase Kinase 3/metabolism , Mice, Nude , Mutant Proteins/metabolism , Neoplasms/pathology , Protein Stability , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
14.
J Immunol ; 190(3): 1264-75, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23275604

ABSTRACT

Sepsis is a leading cause of intensive care unit admissions, with high mortality and morbidity. Although outcomes have improved with better supportive care, specific therapies are limited. Endothelial activation and oxidant injury are key events in the pathogenesis of sepsis-induced lung injury. The signaling pathways leading to these events remain poorly defined. We sought to determine the role of MAPK kinase 3 (MKK3), a kinase of the p38 group, in the pathogenesis of sepsis. We used a murine i.p. LPS model of systemic inflammation to mimic sepsis. Lung injury parameters were assessed in lung tissue and bronchoalveolar lavage specimens. Primary lung endothelial cells were cultured and assessed for mediators of inflammation and injury, such as ICAM-1, AP-1, NF-κB, and mitochondrial reactive oxygen species. Our studies demonstrate that MKK3 deficiency confers virtually complete protection against organ injury after i.p. LPS. Specifically, MKK3(-/-) mice were protected against acute lung injury, as assessed by reduced inflammation, mitochondrial reactive oxygen species generation, endothelial injury, and ICAM-1 expression after LPS administration. Our results show that endothelial MKK3 is required for inflammatory cell recruitment to the lungs, mitochondrial oxidant-mediated AP-1, NF-κB activation, and ICAM-1 expression during LPS challenge. Collectively, these studies identify a novel role for MKK3 in lethal LPS responses and provide new therapeutic targets against sepsis and acute lung injury.


Subject(s)
Acute Lung Injury/enzymology , Endothelial Cells/enzymology , Endotoxemia/enzymology , MAP Kinase Kinase 3/physiology , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Animals , Apoptosis , Bronchoalveolar Lavage Fluid , Chemotaxis, Leukocyte/physiology , Disease Models, Animal , Endothelial Cells/pathology , Endotoxemia/pathology , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Lipopolysaccharides/toxicity , Lung/pathology , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 3/deficiency , MAP Kinase Kinase 3/genetics , Mice , Mice, Knockout , Mitochondria/metabolism , NF-kappa B/metabolism , Neutrophil Activation , Peritonitis/chemically induced , Peritonitis/enzymology , RNA, Small Interfering/pharmacology , Radiation Chimera , Reactive Oxygen Species/metabolism , Sepsis/enzymology , Transcription Factor AP-1/metabolism
15.
J Biol Chem ; 285(19): 14160-9, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20223820

ABSTRACT

Mitogen-activated protein kinase kinase 3 (MAP2K3) is a member of the dual specificity kinase group. Growing evidence links MAP2K3 to invasion and tumor progression. Here, we identify MAP2K3 as a transcriptional target of endogenous gain-of-function p53 mutants R273H, R175H, and R280K. We show that MAP2K3 modulation occurred at the mRNA and protein levels and that endogenous mutant p53 proteins are capable of binding to and activate the MAP2K3 promoter. In addition, we found that the studied p53 mutants regulate MAP2K3 gene expression through the involvement of the transcriptional cofactors NF-Y and NF-kappaB. Finally, functional studies showed that endogenous MAP2K3 knockdown inhibits proliferation and survival of human tumor cells, whereas the ectopic expression of MAP2K3 can rescue the proliferative defect induced by mutant p53 knockdown. Taken together, our findings define a novel player through which mutant p53 exerts its gain-of-function activity in cancer cells.


Subject(s)
Gene Expression Regulation, Neoplastic , MAP Kinase Kinase 3/genetics , Mutation/genetics , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Flow Cytometry , Humans , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 3/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasms/pathology , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transcriptional Activation , Up-Regulation
16.
Oncogene ; 29(10): 1519-30, 2010 Mar 11.
Article in English | MEDLINE | ID: mdl-20023695

ABSTRACT

The Ras oncogene is known to activate three major MAPK pathways, ERK, JNK, p38 and exert distinct cellular phenotypes, that is, apoptosis and invasion through the Ras-MKK3-p38-signaling cascade. We attempted to identify the molecular targets of this pathway that selectively govern the invasive phenotype. Stable transfection of NIH3T3 fibroblasts with MKK3(act) cDNA construct revealed similar p38-dependent in vitro characteristics observed in Ha-Ras(EJ)-transformed NIH3T3 cells, including enhanced invasiveness and anchorage-independent growth correlating with p38 phosphorylation status. To identify the consensus downstream targets of the Ras-MKK3-p38 cascade involved in invasion, in vitro invasion assays were used to isolate highly invasive cells from both, MKK3 and Ha-Ras(EJ) transgenic cell lines. Subsequently a genome-wide transcriptome analysis was employed to investigate differentially regulated genes in invasive Ha-Ras(EJ)- and MKK3(act)-transfected NIH3T3 fibroblasts. Using this phenotype-assisted approach combined with system level protein-interaction network analysis, we identified FOXM1, PLK1 and CDK1 to be differentially regulated in invasive Ha-Ras(EJ)-NIH3T3 and MKK3(act)-NIH3T3 cells. Finally, a FOXM1 RNA-knockdown approach revealed its requirement for both invasion and anchorage-independent growth of Ha-Ras(EJ)- and MKK3(act)-NIH3T3 cells. Together, we identified FOXM1 as a key downstream target of Ras and MKK3-induced cellular in vitro invasion and anchorage-independent growth signaling.


Subject(s)
Forkhead Transcription Factors/genetics , Gene Expression Profiling , MAP Kinase Kinase 3/genetics , p38 Mitogen-Activated Protein Kinases/genetics , ras Proteins/genetics , Animals , Blotting, Western , Cell Adhesion , Cell Movement , Flavonoids/pharmacology , Forkhead Box Protein M1 , Forkhead Transcription Factors/metabolism , Imidazoles/pharmacology , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 3/metabolism , Mice , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Pyridines/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism , ras Proteins/metabolism
17.
Redox Rep ; 14(6): 243-50, 2009.
Article in English | MEDLINE | ID: mdl-20003709

ABSTRACT

Previous studies have reported that selenite, a known antioxidant, protects brain against ischemia/reperfusion injury, which is mediated by oxidative stress. The aim of this study was to investigate whether selenite can protect kidney against ischemic injury by reducing activation of the apoptosis signal regulating kinase 1 (ASK1)/mitogen-activated protein kinase kinase 3 (MKK3)/p38 mitogen-activated protein kinase signaling pathway. The activation and expression of ASK1, MKK3, p38, caspase 3 and cleaved PARP were analyzed by Western blot. Apoptosis of renal tubular epithelial cells was assessed by the terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling method. Malondialdehyde (MDA) levels were measured by the thiobarbituric acid reaction. Blood serum creatinine and blood urea nitrogen level were measured with an Olympus automatic multi-analyzer. We found that selenite attenuated significantly ASK1, MKK3, and p38 phosphorylation at 3 h after renal ischemia. Furthermore, selenite decreased significantly renal epithelial tubular cell apoptosis. In addition, selenite reduced the MDA level. These findings suggest that the protective action of selenite on ischemia renal injury is associated closely with reducing activation of the ASK1-MKK3-p38 signal pathway.


Subject(s)
Kidney/drug effects , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , Reperfusion Injury/drug therapy , Signal Transduction/drug effects , Sodium Selenite/therapeutic use , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Apoptosis/physiology , Caspase 3/metabolism , Enzyme Activation , Humans , Kidney/metabolism , Kidney/pathology , Lipid Peroxidation/drug effects , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Male , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/physiology , Sodium Selenite/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
18.
J Periodontal Res ; 43(2): 201-11, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18302623

ABSTRACT

BACKGROUND AND OBJECTIVE: Lipopolysaccharide from gram-negative bacteria is one of the microbial-associated molecular patterns that initiate the immune/inflammatory response, leading to the tissue destruction observed in periodontitis. The aim of this study was to evaluate the role of the p38 mitogen-activated protein kinase (MAPK) signaling pathway in lipopolysaccharide-induced receptor activator of nuclear factor-kappaB ligand (RANKL) expression by murine periodontal ligament cells. MATERIAL AND METHODS: Expression of RANKL and osteoprotegerin mRNA was studied by reverse transcription-polymerase chain reaction upon stimulation with lipopolysaccharide from Escherichia coli and Aggregatibacter actinomycetemcomitans. The biochemical inhibitor SB203580 was used to evaluate the contribution of the p38 MAPK signaling pathway to lipopolysaccharide-induced RANKL and osteoprotegerin expression. Stable cell lines expressing dominant-negative forms of MAPK kinase (MKK)-3 and MKK6 were generated to confirm the role of the p38 MAPK pathway. An osteoclastogenesis assay using a coculture model of the murine monocytic cell line RAW 264.7 was used to determine if osteoclast differentiation induced by lipopolysaccharide-stimulated periodontal ligament was correlated with RANKL expression. RESULTS: Inhibiting p38 MAPK prior to lipopolysaccharide stimulation resulted in a significant decrease of RANKL mRNA expression. Osteoprotegerin mRNA expression was not affected by lipopolysaccharide or p38 MAPK. Lipopolysaccharide-stimulated periodontal ligament cells increased osteoclast differentiation, an effect that was completely blocked by osteoprotegerin and significantly decreased by inhibition of MKK3 and MKK6, upstream activators of p38 MAPK. Conditioned medium from murine periodontal ligament cultures did not increase osteoclast differentiation, indicating that periodontal ligament cells produced membrane-bound RANKL. CONCLUSION: Lipopolysaccharide resulted in a significant increase of RANKL in periodontal ligament cells. The p38 MAPK pathway is required for lipopolysaccharide-induced membrane-bound RANKL expression in these cells.


Subject(s)
MAP Kinase Signaling System , Osteoclasts , Periodontal Ligament/metabolism , RANK Ligand/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism , Aggregatibacter actinomycetemcomitans , Animals , Cell Differentiation , Cell Line, Transformed , Escherichia coli , Gene Expression , Lipopolysaccharides , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 6/antagonists & inhibitors , Mice , Osteoprotegerin/biosynthesis , Periodontal Ligament/cytology
19.
Mol Cancer Ther ; 6(4): 1212-22, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17406030

ABSTRACT

Although astrocytic brain tumors do not metastasize systemically, during tumorigenesis glioma cells adopt an invasive phenotype that is poorly targeted by conventional therapies; hence, glioma patients die of recurrence from the locally invasive tumor population. Our work is aimed at identifying and validating novel therapeutic targets and biomarkers in invasive human gliomas. Transcriptomes of invasive glioma cells relative to stationary cognates were produced from a three-dimensional spheroid in vitro invasion assay by laser capture microdissection and whole human genome expression microarrays. Qualitative differential expression of candidate invasion genes was confirmed by quantitative reverse transcription-PCR, clinically by immunohistochemistry on tissue microarray, by immunoblotting on surgical specimens, and on two independent gene expression data sets of glial tumors. Cell-based assays and ex vivo brain slice invasion studies were used for functional validation. We identify mitogen-activated protein kinase (MAPK) kinase 3 (MKK3) as a key activator of p38 MAPK in glioma; MKK3 activation is strongly correlated with p38 activation in vitro and in vivo. We further report that these members of the MAPK family are strong promoters of tumor invasion, progression, and poor patient survival. Inhibition of either candidate leads to significantly reduced glioma invasiveness in vitro. Consistent with the concept of synthetic lethality, we show that inhibition of invasion by interference with these genes greatly sensitizes arrested glioma cells to cytotoxic therapies. Our findings therefore argue that interference with MKK3 signaling through a novel treatment combination of p38 inhibitor plus temozolomide heightens the vulnerability of glioma to chemotherapy.


Subject(s)
Glioma/enzymology , Glioma/pathology , MAP Kinase Kinase 3/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Astrocytoma/enzymology , Astrocytoma/pathology , Biomarkers/metabolism , Cell Line, Tumor , Collagen Type I/metabolism , Disease Progression , Enzyme Activation/drug effects , Gene Expression Profiling , Glioma/diagnosis , Glioma/genetics , Humans , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 3/genetics , Male , Neoplasm Invasiveness , Phosphorylation/drug effects , Prognosis , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Survival Analysis , Up-Regulation/drug effects , Up-Regulation/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
20.
J Interferon Cytokine Res ; 26(10): 719-29, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17032166

ABSTRACT

Coupled bone turnover is directed by the expression of receptor-activated NF-kappaB ligand (RANKL) and its decoy receptor, osteoprotegerin (OPG). Proinflammatory cytokines, such as interleukin-1beta (IL-1beta) and tumor necrosis factor-alpha (TNF-alpha) induce RANKL expression in bone marrow stromal cells. Here, we report that IL-1beta and TNF-alpha-induced RANKL requires p38 mitogen-activating protein kinase (MAPK) pathway activation for maximal expression. Real-time PCR was used to assess the p38 contribution toward IL-1beta and TNF-alpha-induced RANKL mRNA expression. Steady-state RANKL RNA levels were increased approximately 17-fold by IL-1beta treatment and subsequently reduced approximately 70%-90% when p38 MAPK was inhibited with SB203580. RANKL mRNA stability data indicated that p38 MAPK did not alter the rate of mRNA decay in IL-1beta-induced cells. Using a RANKL-luciferase cell line receptor containing a 120-kB segment of the 5' flanking region of the RANKL gene, reporter expression was stimulated 4-5-fold by IL-1beta or TNF-alpha treatment. IL-1beta-induced RANKL reporter expression was completely blocked with specific p38 inhibitors as well as dominant negative mutant constructs of MAPK kinase-3 and -6. In addition, blocking p38 signaling in bone marrow stromal cells partially inhibited IL-1beta and TNF-alpha-induced osteoclastogenesis in vitro. Results from these studies indicate that p38 MAPK is a major signaling pathway involved in IL-1beta and TNF-alpha-induced RANKL expression in bone marrow stromal cells.


Subject(s)
Bone Marrow Cells/metabolism , Carrier Proteins/biosynthesis , Cytokines/pharmacology , Membrane Glycoproteins/biosynthesis , Mitogen-Activated Protein Kinase Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/enzymology , Carrier Proteins/genetics , Cell Differentiation , Cell Line , Genes, Reporter , Imidazoles/pharmacology , Interleukin-1/antagonists & inhibitors , Interleukin-1/pharmacology , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase 6/antagonists & inhibitors , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , MAP Kinase Signaling System/drug effects , Membrane Glycoproteins/genetics , Mice , Mutation , Osteoclasts/cytology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , RANK Ligand , RNA, Messenger/metabolism , Receptor Activator of Nuclear Factor-kappa B , Stromal Cells/drug effects , Stromal Cells/enzymology , Stromal Cells/metabolism , Transcriptional Activation , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...