Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Publication year range
1.
J Pharmacol Sci ; 148(3): 326-330, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35177212

ABSTRACT

Extracellular signal-regulated protein kinase 5 (ERK5) has various physiological functions. However, the physiological role of ERK5 in the treatment of mice with an illicit drug such as methamphetamine (METH) remains unknown. We revealed that mice treated with METH showed hyperactivity, and increased p-ERK5 and Iba1 (a microglia marker) levels in the striatum. Additionally, these changes were inhibited by pretreatment with the ERK5 inhibitor BIX02189. The results suggest that METH-induced hyperactivity is associated with the activation of microglia via p-ERK5 in the striatum. Thus, the ERK5 pathway components in the central nervous system are potential therapeutic targets for preventing METH addiction.


Subject(s)
Aniline Compounds/pharmacology , Corpus Striatum/cytology , Hyperkinesis/chemically induced , Hyperkinesis/drug therapy , Indoles/pharmacology , Methantheline/adverse effects , Microglia/drug effects , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/physiology , Aniline Compounds/therapeutic use , Animals , Calcium-Binding Proteins/metabolism , Corpus Striatum/metabolism , Indoles/therapeutic use , Mice , Microfilament Proteins/metabolism , Microglia/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Psychomotor Agitation , Substance-Related Disorders/prevention & control
2.
Future Oncol ; 14(25): 2599-2613, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30073865

ABSTRACT

AIM: The airway epithelium of smokers exhibits upregulated SPRR3, an indicator of pathogenic keratinization. The mechanisms underlying this phenomenon require investigation. PATIENTS & METHODS: Human bronchial epithelial (HBE) SPRR3 expression was analyzed by smoking status. Primary HBE cells were exposed to cigarette smoke (CS). SPRR3 expression, SPRR3 promoter activity, AP-1 factor binding and AP-1 factors' effects were analyzed. RESULTS: Current smokers display SPRR3 upregulation relative to never smokers. CS upregulates SPRR3 transcription in an exposure-dependent manner. CS promotes c-Jun and Fra1 binding to the SPRR3-AP-1/TRE site. Wild-type c-Jun and Fra1 upregulate, whereas c-Jun and Fra1, dominant-negative mutants, suppress SPRR3 promoter activity. CONCLUSION: CS induces SPRR3 upregulation in HBE cells by promoting aberrant c-Jun/Fra1 dimerization.


Subject(s)
Bronchi/metabolism , Cornified Envelope Proline-Rich Proteins/genetics , Protein Multimerization , Proto-Oncogene Proteins c-fos/chemistry , Proto-Oncogene Proteins c-jun/chemistry , Tobacco Smoking/adverse effects , Aged , Aged, 80 and over , Cells, Cultured , Epithelial Cells/metabolism , Female , Gene Expression Regulation , Humans , Male , Middle Aged , Mitogen-Activated Protein Kinase 7/physiology , Mitogen-Activated Protein Kinase 8/physiology , Promoter Regions, Genetic , Transcription Factor AP-1/physiology , Up-Regulation
3.
Biol Reprod ; 97(3): 400-412, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-29025069

ABSTRACT

The differentiation of endometrial stromal cells into decidual cells, termed decidualization, is an integral step in the establishment of pregnancy. The mitogen-activated protein kinase homolog, WNK lysine deficient protein kinase 1 (WNK1), is activated downstream of epidermal growth factor receptor during decidualization. Primary human endometrial stromal cells (HESCs) were subjected to small interfering RNA knockdown of WNK1 followed by in vitro decidualization. This abrogated expression of the decidual marker genes, insulin like growth factor binding protein 1 (IGFBP1) and prolactin (PRL), and prevented adoption of decidual cell morphology. Analysis of the WNK1-dependent transcriptome by RNA-Seq demonstrated that WNK1 regulates the expression of 1858 genes during decidualization. Gene ontology and upstream regulator pathway analysis showed that WNK1 regulates cell migration, differentiation, and proliferation. WNK1 was required for many of the gene expression changes that drive decidualization, including the induction of the inflammatory cytokines, C-C motif chemokine ligand 8 (CCL8), interleukin 1 beta (IL1B), and interleukin 15 (IL15), and the repression of transforming growth factor-beta (TGF-beta) pathway genes, including early growth response 2 (EGR2), SMAD family member 3 (SMAD3), integrin subunit alpha 2 (ITGA2), integrin subunit alpha 4 (ITGA4), and integrin subunit beta 3 (ITGB3). In addition to abrogating decidualization, WNK1 knockdown decreased the migration and proliferation of HESCs. Furthermore, mitogen-activated protein kinase 7 (MAPK7), a known downstream target of WNK1, was activated during decidualization in a WNK1-dependent manner. Small interfering RNA knockdown of MAPK7 demonstrated that MAPK7 regulates a subset of WNK1-regulated genes and controls the migration and proliferation of HESCs. These results indicate that WNK1 and MAPK7 promote migration and proliferation during decidualization and regulate the expression of inflammatory cytokines and TGF-beta pathway genes in HESCs.


Subject(s)
Decidua/cytology , Endometrium/cytology , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/physiology , Stromal Cells/physiology , WNK Lysine-Deficient Protein Kinase 1/deficiency , WNK Lysine-Deficient Protein Kinase 1/genetics , Adult , Cell Movement , Cell Proliferation , Cytokines/biosynthesis , Cytokines/genetics , Female , Gene Expression Regulation , Gene Knockdown Techniques , Humans , RNA, Small Interfering/pharmacology , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics
4.
Arterioscler Thromb Vasc Biol ; 34(11): 2378-86, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25301843

ABSTRACT

Atherosclerosis is a focal disease that develops preferentially where nonlaminar, disturbed blood flow occurs, such as branches, bifurcations, and curvatures of large arteries. Endothelial cells sense and respond differently to disturbed flow compared with steady laminar flow. Disturbed flow that occurs in so-called atheroprone areas activates proinflammatory and apoptotic signaling, and this results in endothelial dysfunction and leads to subsequent development of atherosclerosis. In contrast, steady laminar flow as atheroprotective flow promotes expression of many anti-inflammatory genes, such as Kruppel-like factor 2 and endothelial nitric oxide synthase and inhibits endothelial inflammation and athrogenesis. Here we will discuss that disturbed flow and steady laminar flow induce pro- and antiatherogenic events via flow type-specific mechanotransduction pathways. We will focus on 5 mechanosensitive pathways: mitogen-activated protein kinases/extracellular signal-regulated kinase 5/Kruppel-like factor 2 signaling, extracellular signal-regulated kinase/peroxisome proliferator-activated receptor signaling, and mechanosignaling pathways involving SUMOylation, protein kinase C-ζ, and p90 ribosomal S6 kinase. We think that clarifying regulation mechanisms between these 2 flow types will provide new insights into therapeutic approaches for the prevention and treatment of atherosclerosis.


Subject(s)
Atherosclerosis/physiopathology , Endothelium, Vascular/physiopathology , Mechanotransduction, Cellular/physiology , Vascular Remodeling/physiology , Animals , Biomechanical Phenomena/physiology , Disease Models, Animal , Humans , Mitogen-Activated Protein Kinase 7/physiology , Peroxisome Proliferator-Activated Receptors/physiology , Signal Transduction/physiology
5.
J Biol Chem ; 288(4): 2623-31, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23223235

ABSTRACT

Prolactin-stimulated adult neurogenesis in the subventricular zone (SVZ) and olfactory bulb (OB) mediates several reproductive behaviors including mating/pregnancy, dominant male pheromone preference in females, and paternal recognition of offspring. However, downstream signaling mechanisms underlying prolactin-induced adult neurogenesis are completely unknown. We report here for the first time that prolactin activates extracellular signal-regulated kinase 5 (ERK5), a MAP kinase that is specifically expressed in the neurogenic regions of the adult mouse brain. Knockdown of ERK5 by retroviral infection of shRNA attenuates prolactin-stimulated neurogenesis in SVZ-derived adult neural stem/progenitor cells (aNPCs). Inducible erk5 deletion in adult neural stem cells of transgenic mice inhibits neurogenesis in the SVZ and OB following prolactin infusion or mating/pregnancy. These results identify ERK5 as a novel and critical signaling mechanism underlying prolactin-induced adult neurogenesis.


Subject(s)
Brain/metabolism , Mitogen-Activated Protein Kinase 7/physiology , Olfactory Bulb/metabolism , Prolactin/metabolism , Animals , Brain Mapping/methods , Female , Gene Deletion , Genotype , Mice , Mice, Knockout , Microscopy, Confocal/methods , Mitogen-Activated Protein Kinase 7/metabolism , Neurogenesis , Recombinant Proteins/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Tamoxifen/pharmacology
6.
J Neurochem ; 129(6): 980-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24479685

ABSTRACT

The gene encoding leucine-rich repeat kinase 2 (LRRK2) comprises a major risk factor for Parkinson's disease. Recently, it has emerged that LRRK2 plays important roles in the immune system. LRRK2 is induced by interferon-γ (IFN-γ) in monocytes, but the signaling pathway is not known. Here, we show that IFN-γ-mediated induction of LRRK2 was suppressed by pharmacological inhibition and RNA interference of the extracellular signal-regulated kinase 5 (ERK5). This was confirmed by LRRK2 immunostaining, which also revealed that the morphological responses to IFN-γ were suppressed by ERK5 inhibitor treatment. Both human acute monocytic leukemia THP-1 cells and human peripheral blood monocytes stimulated the ERK5-LRRK2 pathway after differentiation into macrophages. Thus, LRRK2 is induced via a novel, ERK5-dependent IFN-γ signal transduction pathway, pointing to new functions of ERK5 and LRRK2 in human macrophages. Leucine-rich repeat kinase 2 (LRRK2) is a major risk factor for the development of Parkinson's disease (PD). However, the role of LRRK2 in the affected neurons remains enigmatic. Recently, LRRK2 has been reported to be strongly expressed in the immune system. Here, we demonstrate that LRRK2 is induced by Interferon gamma via extracellular signal-regulated kinase 5 (ERK5) in macrophages, thus providing new insights in LRRK2 and ERK5 biology.


Subject(s)
Interferon-gamma/pharmacology , Macrophages/enzymology , Mitogen-Activated Protein Kinase 7/physiology , Protein Serine-Threonine Kinases/biosynthesis , Blotting, Western , Cells, Cultured , Cytokines/metabolism , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Humans , Immunohistochemistry , Interferon-gamma/antagonists & inhibitors , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Macrophages/drug effects , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects
7.
J Biol Chem ; 287(32): 26478-94, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22707717

ABSTRACT

Endothelial cell (EC) Toll-like receptor 2 (TLR2) activation up-regulates the expression of inflammatory mediators and of TLR2 itself and modulates important endothelial functions, including coagulation and permeability. We defined TLR2 signaling pathways in EC and tested the hypothesis that TLR2 signaling differs in EC and monocytes. We found that ERK5, heretofore unrecognized as mediating TLR2 activation in any cell type, is a central mediator of TLR2-dependent inflammatory signaling in human umbilical vein endothelial cells, primary human lung microvascular EC, and human monocytes. Additionally, we observed that, although MEK1 negatively regulates TLR2 signaling in EC, MEK1 promotes TLR2 signaling in monocytes. We also noted that activation of TLR2 led to the up-regulation of intracellularly expressed TLR2 and inflammatory mediators via NF-κB, JNK, and p38-MAPK. Finally, we found that p38-MAPK, JNK, ERK5, and NF-κB promote the attachment of human neutrophils to lung microvascular EC that were pretreated with TLR2 agonists. This study newly identifies ERK5 as a key regulator of TLR2 signaling in EC and monocytes and indicates that there are fundamental differences in TLR signaling pathways between EC and monocytes.


Subject(s)
Endothelium, Vascular/cytology , MAP Kinase Kinase 1/physiology , Mitogen-Activated Protein Kinase 7/physiology , Monocytes/cytology , Toll-Like Receptor 2/physiology , Cell Adhesion , Cell Line , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , NF-kappa B/metabolism , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Signal Transduction , Toll-Like Receptor 2/metabolism , Up-Regulation
8.
J Immunol ; 186(3): 1343-7, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21187442

ABSTRACT

The transcription factor Krüppel-like factor 2 (KLF2) controls the emigration of conventional T cells from the thymus through its regulation of the cell surface receptor S1P1. Prior to KLF2 expression, developing T cells require a positive selection signal through the TCR. However, following positive selection there are time, spatial, and maturational events that occur before KLF2 is finally upregulated and emigration occurs. We are interested in determining the signals that upregulate KLF2 and allow thymocytes to emigrate into circulation and whether they are linked to functional maturation. In endothelial cells KLF2 expression has been shown to be dependent on the mitogen-activated protein kinase ERK5. Furthermore, it has been reported that IL-7 signaling leads to the phosphorylation of ERK5. Thus, we hypothesized that IL-7R signaling through ERK5 could drive the expression of KLF2. In this study, we provide evidence that this hypothesis is incorrect. We also found that CD8 lineage specification occurred normally in the absence of IL-7R signaling, in contrast to a recently proposed model. We showed that both CD4 and CD8 T cells complete maturation and express KLF2 independently of ERK5 and IL-7.


Subject(s)
Cell Differentiation/immunology , Cell Movement/immunology , Interleukin-7/physiology , Mitogen-Activated Protein Kinase 7/physiology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , Cell Differentiation/genetics , Cell Movement/genetics , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/physiology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase 7/deficiency , Mitogen-Activated Protein Kinase 7/genetics , Receptors, Interleukin-7/antagonists & inhibitors , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/genetics , T-Lymphocyte Subsets/metabolism , Thymus Gland/metabolism
9.
Biochim Biophys Acta ; 1813(10): 1854-62, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21767578

ABSTRACT

Hepatitis C virus (HCV) is an RNA positive strand virus, member of the Flaviviridae family. The viral particle is composed of a capsid containing the genome, surrounded by E1 and E2 proteins, however different forms of viral particles have been observed including non-enveloped particles. Previous reports have proposed that hepatitis C non-enveloped capsid-like particles (HCVne) enter cells of hepatic origin via clathrin-mediated endocytosis, during which different signaling events occur. In this report we show that HCVne particles are capable of inducing the recently discovered ERK5 pathway, in a dose dependent way. The ERK5 pathway can be activated by growth factors and other extracellular signals. This specific activation occurs through a well characterized upstream kinase, MEK5, and is capable of inducing gene regulation of mef2. In contrast, when HCV core structural and NS5A non-structural proteins were expressed endogenously no activation of this pathway was detected. These cell signaling events could be of critical importance and might give clues for the elucidation of cellular manifestations associated with HCV infection.


Subject(s)
Capsid Proteins/pharmacology , Hepacivirus , MADS Domain Proteins/metabolism , MAP Kinase Kinase 5/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Myogenic Regulatory Factors/metabolism , Virion/physiology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Hep G2 Cells , Hepacivirus/physiology , Humans , MADS Domain Proteins/physiology , MAP Kinase Kinase 5/physiology , MEF2 Transcription Factors , Mitogen-Activated Protein Kinase 7/physiology , Models, Biological , Myogenic Regulatory Factors/physiology , Signal Transduction/drug effects , Spodoptera , Viral Core Proteins/pharmacology , Viral Envelope Proteins/pharmacology , Viral Nonstructural Proteins/pharmacology
10.
J Pharmacol Sci ; 120(4): 259-63, 2012.
Article in English | MEDLINE | ID: mdl-23165802

ABSTRACT

Big mitogen-activated protein kinase 1 (BMK1), also known as extracellular signal-regulated kinase 5 (ERK5), is a newly identified member of the mitogen-activated protein (MAP) kinase family. BMK1 has been reported to be sensitive to various neuro-humoral factors and oxidative stress in various cells. In this review, we focused on the role of BMK1 in atherosclerosis in a cultured rat aortic smooth muscle cell model. Treatment with platelet-derived growth factor caused vascular smooth muscle cell (VSMC) migration in a BMK1 activation-dependent manner. H(2)O(2) caused BMK1 activation and VSMC death, including apoptosis of VSMCs. An inhibitory function for BMK1 against cell death from oxidative stress was discovered using siRNA techniques to downregulate the expression of BMK1. These findings suggest a role for BMK1 in the pathogenesis and/or progression of atherosclerosis.


Subject(s)
Atherosclerosis/etiology , Mitogen-Activated Protein Kinase 7/physiology , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Animals , Apoptosis/genetics , Atherosclerosis/genetics , Cell Movement/genetics , Cells, Cultured , Disease Progression , Hydrogen Peroxide/adverse effects , Mice , Mitogen-Activated Protein Kinase 7/metabolism , Oxidative Stress/physiology , RNA, Small Interfering , Rats , Signal Transduction/physiology
11.
J Immunol ; 185(6): 3498-503, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20729331

ABSTRACT

Most cancer cells use anaerobic-like glycolysis to generate energy instead of oxidative phosphorylation. They also avoid recognition by CTLs, which occurs primarily through decreasing the level of MHC class I (MHC-I) at the cell surface. We find that the two phenomena are linked; culture conditions that force respiration in leukemia cells upregulate MHC-I transcription and protein levels at the cell surface, whereas these decrease in cells forced to perform fermentation as well as in leukemia cells lacking a functional mitochondrial respiratory chain. Forced respiration leads to increased expression of the MAPK ERK5, which activates MHC-I gene promoters, and ERK5 accumulation in mitochondria. Respiration-induced MHC-I upregulation is reversed upon short hairpin RNA-mediated ERK5 downregulation and by inactive mutants of ERK5. Moreover, short hairpin RNA for ERK5 leukemia cells do not tolerate forced respiration. Thus, the expression of ERK5 and MHC-I is linked to cell metabolism and notably diminished by the metabolic adaptations found in tumor cells.


Subject(s)
Gene Expression Regulation/immunology , Histocompatibility Antigens Class I/biosynthesis , Leukemia, B-Cell/immunology , MAP Kinase Signaling System/immunology , Mitogen-Activated Protein Kinase 7/physiology , Oxidative Phosphorylation , Adenosine Triphosphate/biosynthesis , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival/immunology , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Down-Regulation/immunology , Glutamine/metabolism , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Jurkat Cells , Leukemia L1210 , Leukemia, B-Cell/enzymology , Leukemia, B-Cell/pathology , MAP Kinase Signaling System/genetics , Mice , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/genetics , Up-Regulation/immunology
12.
Gut ; 60(5): 648-57, 2011 May.
Article in English | MEDLINE | ID: mdl-21199832

ABSTRACT

BACKGROUND AND AIMS: Radiotherapy for neoplastic disease is associated with significant adverse enteric effects associated with excessive cell death. Ionising radiation induces cell death by a mechanism that is dependent on JNK (c-jun N-terminal kinase) pathway signalling. Additionally, it is known that cells exposed to extracellular bacterial products such as flagellin, pleiotropically activate a number of innate immune pathways, including that of JNK. The JNK pathway controls its own activity by inducing the transcription of mitogen-activated protein kinase phosphatase-7 (MKP-7) which directly targets phosphorylated JNK, thus functioning as a negative feedback loop. Previously, it has been shown that flagellin limits ionising radiation-induced mortality in mice, but the cellular mechanism of protection remained unknown. METHODS: Wild-type C57BL/6 or tlr5(-/-) C57BL/6 were injected with flagellin 2 h before exposure to irradiation, and their intestines were examined for apoptosis. Candidate proteins mediating cytoprotection from irradiation were identified by expression profiling. One of these candidates, MKP-7, was cloned and packaged into adenovirus particles, used to infect cultured cells, and examined for the extent to which its activity reduced cellular apoptosis by flow cytometry or immunoblot analysis. RESULTS: Flagellin pretreatment protected mice from radiation-induced intestinal mucosal injury and apoptosis via a Toll-like receptor 5 (TLR5)-dependent mechanism. Expression profiling of flagellin-treated mice showed upregulation of MKP-7, an inducible repressor of the JNK pathway. MKP-7 expression reached a maximum at 2 h after flagellin treatment, coinciding with suppression of phosphorylated JNK and JNK pathway inhibition. Furthermore, constitutive MKP-7 expression protected cultured cells from radiation-induced apoptosis. CONCLUSIONS: Flagellin is a promising adjuvant for suppressing ionising radiation-induced injury. MKP-7 activity exhibits cytoprotective effects, and is thus a candidate cellular molecule for limiting the damaging effect of radiotherapy on the gastreointestinal system.


Subject(s)
Flagellin/therapeutic use , Intestinal Mucosa/radiation effects , Mitogen-Activated Protein Kinase 7/physiology , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/therapeutic use , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Apoptosis Inducing Factor/antagonists & inhibitors , Cells, Cultured , Cytoprotection/genetics , Drug Evaluation, Preclinical/methods , Flagellin/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Intestine, Small/drug effects , Intestine, Small/metabolism , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 7/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Phosphorylation/radiation effects , RNA, Messenger/genetics , Radiation Injuries, Experimental/enzymology , Radiation Injuries, Experimental/pathology , Radiation-Protective Agents/pharmacology , Toll-Like Receptor 5/physiology , Up-Regulation/drug effects
13.
J Biol Chem ; 285(34): 26199-210, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20551324

ABSTRACT

The MEK5/Erk5 MAPK cascade has recently been implicated in the regulation of endothelial integrity and represents a candidate pathway mediating the beneficial effects of laminar flow, a major factor preventing vascular dysfunction and disease. Here we expressed a constitutively active mutant of MEK5 (MEK5D) to study the transcriptional and functional responses to Erk5 activation in human primary endothelial cells. We provide evidence that constitutive Erk5 activation elicits an overall protective phenotype characterized by increased apoptosis resistance and a decreased angiogenic, migratory, and inflammatory potential. This is supported by bioinformatic microarray analysis, which uncovered a statistical overrepresentation of corresponding functional clusters as well as a significant induction of anti-thrombotic, hemostatic, and vasodilatory genes. We identify KLF4 as a novel Erk5 target and demonstrate a critical role of this transcription factor downstream of Erk5. We show that KLF4 expression largely reproduces the protective phenotype in endothelial cells, whereas KLF4 siRNA suppresses expression of various Erk5 targets. Additionally, we show that vasoprotective statins potently induce KLF4 and KLF4-dependent gene expression via activation of Erk5. Our data underscore a major protective function of the MEK5/Erk5/KLF4 module in ECs and implicate agonistic Erk5 activation as potential strategy for treatment of vascular diseases.


Subject(s)
Endothelial Cells/physiology , Kruppel-Like Transcription Factors/genetics , MAP Kinase Kinase 5/metabolism , Mitogen-Activated Protein Kinase 7/physiology , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Gene Expression Regulation , Hemostasis/genetics , Humans , Inflammation/genetics , Kruppel-Like Factor 4 , Mitogen-Activated Protein Kinase 7/metabolism , Phenotype , Protective Agents , Signal Transduction/physiology , Transcriptional Activation , Vasodilation/genetics
14.
Br J Cancer ; 104(4): 664-72, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21266977

ABSTRACT

BACKGROUND: Aberrant mitogen/extracellular signal-regulated kinase 5 (MEK5)-extracellular signal-regulated protein kinase 5 (ERK5)-mediated signalling has been implicated in a number of tumour types including prostate cancer (PCa). The molecular basis of ERK5-driven carcinogenesis and its clinical relevance remain to be fully characterised. METHODS: Modulation of ERK5 expression or function in human PCa PC3 and PC3-ERK5 (stably transfected with ERK5) cells was performed using siRNA-mediated knockdown or the MEK inhibitor PD18435 respectively. In vitro significance of ERK5 signalling was assessed by assays for proliferation, motility, invasion and invadopodia. Expression of matrix metalloproteinases/tissue inhibitors of metalloproteases was determined by Q-RT-PCR. Extracellular signal-regulated protein kinase 5 expression in primary and metastatic PCa was examined using immunohistochemistry. RESULTS: Reduction of ERK5 expression or signalling significantly inhibited the motility and invasive capability of PC3 cells. Extracellular signal-regulated protein kinase 5-mediated signalling significantly promoted formation of in vivo metastasis in an orthotopic PCa model (P<0.05). Invadopodia formation was also enhanced by forced ERK5 expression in PC3 cells. Furthermore, in metastatic PCa, nuclear ERK5 immunoreactivity was significantly upregulated when compared with benign prostatic hyperplasia and primary PCa (P=0.013 and P<0.0001, respectively). CONCLUSION: Our in vitro, in vivo and clinical data support an important role for the MEK5-ERK5 signalling pathway in invasive PCa, which represents a potential target for therapy in primary and metastatic PCa.


Subject(s)
Mitogen-Activated Protein Kinase 7/physiology , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/pathology , Animals , Benzamides/pharmacology , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cells, Cultured , Drug Evaluation, Preclinical , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , MAP Kinase Kinase 5/genetics , MAP Kinase Kinase 5/metabolism , MAP Kinase Kinase 5/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Neoplasm Invasiveness , Phenotype , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/pharmacology , Transfection , Transplantation, Heterologous
15.
J Neurosci Res ; 89(10): 1542-50, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21647938

ABSTRACT

Neurotrophins play essential roles in the development, differentiation, and survival of neuronal and nonneuronal cells. Alterations in neurotrophin expression have been implicated in a variety of neurodegenerative disorders. Dysregulation of brain-derived neurotrophic factor (BDNF) has been implicated in deficits of long-term potentiation and cognition and may contribute to the development of Alzheimer's disease (AD). In this study, we used complementary pharmacological and molecular approaches to evaluate the role of ERK1/2 and ERK5, two members of the MAPK pathway associated with neuroprotection, in regulating BDNF expression in C6 glial cells and primary astrocytes. Our data revealed that U0126, an inhibitor of both ERK5 and ERK1/2, increased the levels of BDNF mRNA, whereas the MEK1/2-specific inhibitor PD184352 did not, suggesting that ERK5 exerts negative control over BDNF expression. This was supported by experiments in which RNAi-mediated depletion of ERK5 led to an increase in BDNF. In contrast, transfection with constitutively active MEK5 resulted in an inhibition of BDNF expression, confirming the inhibitory role of ERK5 in the regulation of BDNF. Interestingly, transfection with the dominant active mutant of MEK1 (MEKR4F), the upstream activator of ERK1/2, resulted in a modest increase in BDNF levels. Collectively, our data suggest that ERK5 and ERK1/2 exert opposite effects on BDNF expression and support the hypothesis that an imbalance of these two signaling pathways may contribute to the pathology of diseases in which neurotrophin dysregulation is noted.


Subject(s)
Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Mitogen-Activated Protein Kinase 7/physiology , Protein Kinase Inhibitors/pharmacology , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/enzymology , Astrocytes/pathology , Benzamides/pharmacology , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Butadienes/pharmacology , Cell Line, Tumor , Glioma/enzymology , Glioma/pathology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/genetics , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Nitriles/pharmacology , Primary Cell Culture , Rats , Rats, Sprague-Dawley
16.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 33(6): 615-9, 2011 Dec.
Article in Zh | MEDLINE | ID: mdl-22509542

ABSTRACT

OBJECTIVE: To study the role of extracellular signal-regulated protein kinase 5 (ERK5) during the biosynthesis of follicle-stimulating hormone (FSH)-mediated progesterone in primary granulosa cells. METHODS: The expressions of phosphorylated and general forms of ERKS in primary granulosa cells after the treatment of FSH were detected by Western blot analysis. The subcellular localization of ERK5 was observed by confocal microscopy. The effect of ERK5 on FSH-mediated progesterone biosynthesis in primary granulosa cells was analyzed using recombinant adenovirus vectors. RESULTS: ERK5 activation was induced by FSH in a time-dependent manner in primary cultured granulosa cells, although the general ERK5 protein level decreased also in a time-dependent manner. The treatment of FSH showed no remarkable effect on the subcellular distribution of endogenous ERK5, which was mainly in the cytoplasm of granulosa cells. The co-infection of Ad-caMEK5 and Ad-wtERK5 increased the progesterone production and StAR expression in primary cultured granulosa cells, whereas inhibition of ERK5 activation suppressed the FSH-stimulated progesterone production. CONCLUSION: ERK5 may stimulate FSH-mediated progesterone production in primary cultured granulosa cells.


Subject(s)
Granulosa Cells/metabolism , Mitogen-Activated Protein Kinase 7/physiology , Progesterone/biosynthesis , Animals , Cells, Cultured , Female , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/drug effects , Mitogen-Activated Protein Kinase 7/metabolism , Rats , Rats, Sprague-Dawley
17.
J Invest Dermatol ; 141(2): 250-252, 2021 02.
Article in English | MEDLINE | ID: mdl-33504438

ABSTRACT

Resistance to targeted therapy and immunotherapy remains a major obstacle in improving care for patients with advanced melanoma. MicroRNAs play important roles in regulating gene networks involved in disease progression and resistance to therapy in cancers such as melanoma. MicroRNA miR-211 contributes to melanocyte and melanoma biology and has been implicated in targeted therapy resistance. Lee et al. (2020) report a novel mechanism by which miR-211 promotes resistance to BRAFV600E inhibitor therapy via the upregulation of the extracellular signal-regulated kinase 5 signaling pathway.


Subject(s)
Melanoma/drug therapy , MicroRNAs/physiology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Drug Resistance, Neoplasm , Humans , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 7/physiology , TRPM Cation Channels/physiology
18.
Sci Rep ; 11(1): 17764, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34493753

ABSTRACT

Endothelial-mesenchymal transition (EndMT) is a form of endothelial dysfunction wherein endothelial cells acquire a mesenchymal phenotype and lose endothelial functions, which contributes to the pathogenesis of intimal hyperplasia and atherosclerosis. The mitogen activated protein kinase 7 (MAPK7) inhibits EndMT and decreases the expression of the histone methyltransferase Enhancer-of-Zeste homologue 2 (EZH2), thereby maintaining endothelial quiescence. EZH2 is the catalytic subunit of the Polycomb Repressive Complex 2 that methylates lysine 27 on histone 3 (H3K27me3). It is elusive how the crosstalk between MAPK7 and EZH2 is regulated in the endothelium and if the balance between MAPK7 and EZH2 is disturbed in vascular disease. In human coronary artery disease, we assessed the expression levels of MAPK7 and EZH2 and found that with increasing intima/media thickness ratio, MAPK7 expression decreased, whereas EZH2 expression increased. In vitro, MAPK7 activation decreased EZH2 expression, whereas endothelial cells deficient of EZH2 had increased MAPK7 activity. MAPK7 activation results in increased expression of microRNA (miR)-101, a repressor of EZH2. This loss of EZH2 in turn results in the increased expression of the miR-200 family, culminating in decreased expression of the dual-specificity phosphatases 1 and 6 who may repress MAPK7 activity. Transfection of endothelial cells with miR-200 family members decreased the endothelial sensitivity to TGFß1-induced EndMT. In endothelial cells there is reciprocity between MAPK7 signaling and EZH2 expression and disturbances in this reciprocal signaling associate with the induction of EndMT and severity of human coronary artery disease.


Subject(s)
Cell Transdifferentiation/physiology , Coronary Artery Disease/pathology , Endothelium, Vascular/pathology , Enhancer of Zeste Homolog 2 Protein/physiology , Mesoderm/pathology , Mitogen-Activated Protein Kinase 7/physiology , Signal Transduction/physiology , Tunica Intima/pathology , 3' Untranslated Regions/genetics , Coronary Artery Disease/enzymology , Coronary Stenosis/enzymology , Coronary Stenosis/pathology , Dual Specificity Phosphatase 1/biosynthesis , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 6/biosynthesis , Dual Specificity Phosphatase 6/genetics , Endothelium, Vascular/enzymology , Enzyme Activation , Gene Expression Regulation , Genes, Reporter , Histone Code , Human Umbilical Vein Endothelial Cells , Humans , Hyperplasia , Mesoderm/enzymology , MicroRNAs/biosynthesis , MicroRNAs/genetics , Tunica Media/pathology
19.
Carcinogenesis ; 31(10): 1734-41, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20729391

ABSTRACT

We have recently shown that American ginseng (AG) prevents and treats mouse colitis. Because both mice and humans with chronic colitis have a high colon cancer risk, we tested the hypothesis that AG can be used to prevent colitis-driven colon cancer. Using the azoxymethane (AOM)/dextran sulfate sodium (DSS) mouse model of ulcerative colitis, we show that AG can suppress colon cancer associated with colitis. To explore the molecular mechanisms of the anticancer effects of AG, we also carried out antibody array experiments on colon cells isolated at a precancerous stage. We found there were 82 protein end points that were either significantly higher (41 proteins) or significantly lower (41 proteins) in the AOM + DSS group compared with the AOM-alone (control) group. In contrast, there were only 19 protein end points that were either significantly higher (10 proteins) or significantly lower (9 proteins) in the AOM + DSS + AG group compared with the AOM-alone (control) group. Overall, these results suggest that AG keeps the colon environment in metabolic equilibrium when mice are treated with AOM + DSS and gives insight into the mechanisms by which AG protects from colon cancer associated with colitis.


Subject(s)
Colitis/drug therapy , Colonic Neoplasms/prevention & control , Panax , Phytotherapy , Plant Extracts/therapeutic use , Adaptor Proteins, Signal Transducing/physiology , Animals , Azoxymethane/toxicity , Colon/drug effects , Colon/pathology , Dextran Sulfate/toxicity , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 7/physiology , PAX2 Transcription Factor/physiology
20.
Mol Cell Biol ; 27(12): 4566-77, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17452462

ABSTRACT

MEKK2 and MEK5 encode Phox/Bem1p (PB1) domains that heterodimerize with one another. MEKK2, MEK5, and extracellular signal-related kinase 5 (ERK5) form a ternary complex through interactions involving the MEKK2 and MEK5 PB1 domains and a 34-amino-acid C-terminal extension of the MEK5 PB1 domain. This C-terminal extension encodes an ERK5 docking site required for MEK5 activation of ERK5. The PB1 domains bind in a front-to-back arrangement, with a cluster of basic amino acids in the front of the MEKK2 PB1 domain binding to the back-end acidic clusters of the MEK5 PB1 domain. The C-terminal moiety, including the acidic cluster of the MEKK2 PB1 domain, is not required for MEK5 binding and binds MKK7. Quiescent MEKK2 preferentially binds MEK5, and MEKK2 activation results in ERK5 activation. Activated MEKK2 binds and activates MKK7, leading to JNK activation. The findings define how the MEKK2 and MEK5 PB1 domains are uniquely used for differential binding of two mitogen-activated protein kinase kinases, MEK5 and MKK7, for the coordinated control of ERK5 and c-Jun N-terminal kinase activation.


Subject(s)
MAP Kinase Kinase 5/chemistry , MAP Kinase Kinase 7/physiology , MAP Kinase Kinase Kinase 2/chemistry , Mitogen-Activated Protein Kinase 7/physiology , Signal Transduction , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mice, Knockout , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL