Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
Add more filters

Publication year range
1.
J Cell Mol Med ; 27(17): 2583-2593, 2023 09.
Article in English | MEDLINE | ID: mdl-37525479

ABSTRACT

Previous studies have demonstrated that mitogen-activated protein kinase 11 (MAPK11) functions as an important point of integration in signalling transduction pathways and controlling endocellular processes, including viability of cells, differentiation, proliferation and apoptosis, through the sequence phosphorylation of the substrate protein Ser/Thr kinase protein cascade. Though MAPK 11 plays an important role in various tumours, especially in the invasive and metastatic processes, its expression and molecular mechanism in clear cell renal cell carcinoma (ccRCC) remain unclear. Runt-associated transcription factor 2 (RUNX2), a main transcription factor for osteoblast differentiation and chondrocyte maturation, has high expression in a number of tumours. In this study, the mRNA and protein levels of targeted genes in ccRCC tissues and adjacent tissues are analysed using the Cancer Genome Atlas (TCGA) database and western blotting. The ccRCC cell proliferation was measured with colony formation and EdU assay, and cell migration was examined through transwell assay. The interactive behaviour between proteins was detected with immunoprecipitation. Half-life period of RUNX2 protein was measured with cycloheximide chase assay. The results of the study indicated overexpression of MAPK11 and RUNX2 in ccRCC tissues and cell lines. MAPK11 and RUNX2 promoted the ccRCC cell proliferation and migration. Additionally, physical interaction took place between RUNX2 and P-MAPK11, which functioned to sustain the stability of RUNX2 protein. The high expression of RUNX2 could neutralize the functional degradation in MAPK11. And the outcomes of the study suggest that the P-MAPK11/RUNX2 axis may be used as a potential therapeutic target of ccRCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/pathology , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 11/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Kidney Neoplasms/pathology , Gene Expression Regulation, Neoplastic
2.
Biochem Biophys Res Commun ; 638: 43-50, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36436341

ABSTRACT

Stomatal movements allow the uptake of CO2 for photosynthesis and water loss through transpiration, therefore play a crucial role in determining water use efficiency. Both red and blue lights induce stomatal opening, and the stomatal apertures under light are finetuned by both positive and negative regulators in guard cells. However, the molecular mechanisms for precisely adjusting stomatal apertures under light have not been completely understood. Here, we provided evidence supporting that Arabidopsis thaliana mitogen-activated protein kinase 11 (MPK11) plays a negative role in red light-induced stomatal opening. First, MPK11 was found to be highly expressed in guard cells, and MPK11-GFP signals were detected in both nuclear and cytoplasm of guard cells. The transcript levels of MPK11 in guard cells were upregulated by white light, and the stomata of mpk11 opened wider than that of wild type under white light. Consistent with the larger stomatal aperture, mpk11 mutant exhibited higher stomatal conductance and CO2 assimilation rate under white light. The transcript levels of the genes responsible for osmolytes increases were higher in guard cells of mpk11 than that of wild type, which may contribute to the larger stomatal aperture of mpk11 under white light. Furthermore, MPK11 transcript levels in guard cells were upregulated by red light, and mpk11 mutant showed a larger stomatal aperture under red light. Taken together, these results demonstrate that red light-upregulated MPK11 plays a negative role in stomatal opening, which finetuning the stomatal opening apertures and preventing excessive water loss by transpiration under light.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Mitogen-Activated Protein Kinase 11/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Carbon Dioxide/metabolism , Plant Stomata/metabolism , Light , Water/metabolism
3.
Plant Cell Environ ; 46(5): 1453-1471, 2023 05.
Article in English | MEDLINE | ID: mdl-36691352

ABSTRACT

High temperatures (HT) cause pollen abortion and poor floret fertility in rice, which is closely associated with excessive accumulation of reactive oxygen species (ROS) in the developing anthers. However, the relationships between accumulation of abscisic acid (ABA) and ROS, and their effects on tapetum-specific programmed cell death (PCD) in HT-stressed anthers are poorly characterised. Here, we determined the spatiotemporal changes in ABA and ROS levels, and their relationships with tapetal PCD under HT exposure. Mutants lacking ABA-activated protein kinase 2 (SAPK2) functions and exogenous ABA treatments were used to explore the effects of ABA signalling on the induction of PCD and ROS accumulation during pollen development. HT-induced pollen abortion was tightly associated with ABA accumulation and oxidative stress. The higher ABA level in HT-stressed anthers resulted in the earlier initiation of PCD induction and subsequently abnormal tapetum degeneration by activating ROS accumulation in developing anthers. Interactions between SAPK2 and DEAD-box ATP-dependent RNA helicase elF4A-1 (RH4) were required for ABA-induced ROS generation in developing anthers. The OsSAPK2 knockout mutants showed the impaired PCD responses in the absence of HT. However, the deficiency of SAPK2 functions did not suppress the ABA-mediated ROS generation in HT-stressed anthers.


Subject(s)
Oryza , Reactive Oxygen Species/metabolism , Oryza/physiology , Abscisic Acid/pharmacology , Abscisic Acid/metabolism , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 11/metabolism , Pollen/physiology , Apoptosis/genetics , Heat-Shock Response , Gene Expression Regulation, Plant
4.
J Exp Bot ; 72(5): 1677-1690, 2021 02 27.
Article in English | MEDLINE | ID: mdl-33448300

ABSTRACT

Seed germination is a critical stage in the plant life cycle and it plays an important role in the efficiency of agricultural production. However, our knowledge of the mechanisms that regulate seed germination remains limited. In this study, we identified a novel gene, MAPK11, that encodes mitogen-activated protein kinase 11; its expression was significantly higher in seeds of tomato varieties with a low optimum germination temperature than in those with a high optimum germination temperature. In tests at 25 °C, overexpression of MAPK11 in an accession with optimum germination at 25 °C resulted in a decrease in germination, whereas RNAi of MAPK11 in an accession with optimum germination at 15 °C resulted in increased germination. Furthermore, we found that lines overexpressing MAPK11 exhibited hypersensitivity to ABA during germination. These observations were at least partially explained by the fact that MAPK11 up-regulated both NCED1 expression and ABA biosynthesis, and that it also affected ABA signaling and negatively regulated germination by influencing the phosphorylation of SnRK2.2 in vivo. In addition, we found that MAPK11 interacts with and phosphorylates SnRK1 in vivo, thereby potentially inhibiting its activation. SnRK1 interacted with ABI5 and suppressed the transcription of ABI5, thereby affecting ABA signaling and the regulation of germination. Our results demonstrate that ABA signaling in tomato is affected by a mechanism that depends on MAPK11 phosphorylating SnRKs, and this ultimately influences seed germination.


Subject(s)
Abscisic Acid/metabolism , Mitogen-Activated Protein Kinase 11/metabolism , Solanum lycopersicum , Basic-Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation, Plant , Germination , Solanum lycopersicum/genetics , Solanum lycopersicum/metabolism , Membrane Proteins , Phosphoproteins , Plants, Genetically Modified/metabolism , Protein Serine-Threonine Kinases/metabolism , Seeds/genetics , Seeds/metabolism
5.
Plant Cell Rep ; 40(3): 491-506, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33388892

ABSTRACT

KEY MESSAGE: StMAPK11 overexpression promotes potato growth, physiological activities and photosynthesis under drought conditions. Mitogen-activated protein kinases (MAPKs) are import regulators of MAPK pathway in plants under drought condition. However, the critical role in potato (Solanum tuberosum L.) drought resistance is not fully understood. In this study, we aimed to explore the role of StMAPK11 under drought stress. The result of RT-qPCR for assay of StMAPKs expression demonstrated that 15 StMAPKs were differentially expressed in leaves, flowers, petioles, stamens, pistils, stems, stolons, roots, tubers and tuber peels of potato. StMAPKs was dynamically modulated by abiotic stresses and plant hormone treatments, and StMAPK11 was apparently up-regulated under drought conditions. Therefore, the vectors pCPB-StMAPK11 and pCPBI121-miRmapk11 for over-expression and down-regulation of StMAPK11 were constructed, respectively, and introduced into potato cultivar Atlantic. The result showed that StMAPK11 promoted potato growth under drought conditions, as well as the physiological activities evidenced by changes in SOD, CAT and POD activity and H2O2, proline and MDA content. StMAPK11 up-regulation intensified drought resistance of potato plant by elevating antioxidant activities and photosynthesis. Moreover, we consolidated the protective role of StMAPK11 in tobacco and Arabidopsis against drought stress. The result could provide new insights into the function of StMAPK11 in drought response and its possible mechanisms.


Subject(s)
Droughts , Mitogen-Activated Protein Kinase 11/metabolism , Photosynthesis/physiology , Plant Proteins/metabolism , Solanum tuberosum/physiology , Arabidopsis/genetics , Arabidopsis/growth & development , Enzymes/metabolism , Gene Expression Regulation, Plant , Hydrogen Peroxide/metabolism , Mitogen-Activated Protein Kinase 11/genetics , Plant Proteins/genetics , Plants, Genetically Modified , Proline/metabolism , Stress, Physiological , Nicotiana/genetics , Nicotiana/growth & development
6.
PLoS Pathog ; 14(5): e1007020, 2018 05.
Article in English | MEDLINE | ID: mdl-29746571

ABSTRACT

The fruit fly Drosophila melanogaster is a powerful model system for the study of innate immunity in vector insects as well as mammals. For vector insects, it is particularly important to understand all aspects of their antiviral immune defenses, which could eventually be harnessed to control the transmission of human pathogenic viruses. The immune responses controlling RNA viruses in insects have been extensively studied, but the response to DNA virus infections is poorly characterized. Here, we report that infection of Drosophila with the DNA virus Invertebrate iridescent Virus 6 (IIV-6) triggers JAK-STAT signaling and the robust expression of the Turandots, a gene family encoding small secreted proteins. To drive JAK-STAT signaling, IIV-6 infection more immediately induced expression of the unpaireds, a family of IL-6-related cytokine genes, via a pathway that required one of the three Drosophila p38 homologs, p38b. In fact, both Stat92E and p38b were required for the survival of IIV-6 infected flies. In addition, in vitro induction of the unpaireds required an NADPH-oxidase, and in vivo studies demonstrated Nox was required for induction of TotA. These results argue that ROS production, triggered by IIV-6 infection, leads to p38b activation and unpaired expression, and subsequent JAK-STAT signaling, which ultimately protects the fly from IIV-6 infection.


Subject(s)
Drosophila melanogaster/immunology , Drosophila melanogaster/virology , Iridovirus/pathogenicity , Signal Transduction/immunology , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunity, Innate , Iridovirus/immunology , Janus Kinases/genetics , Janus Kinases/metabolism , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 11/metabolism , Reactive Oxygen Species/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
7.
RNA Biol ; 17(4): 500-516, 2020 04.
Article in English | MEDLINE | ID: mdl-31928144

ABSTRACT

Huntington's Disease (HD) is a monogenetic neurodegenerative disorder mainly caused by the cytotoxicity of the mutant HTT protein (mHTT) encoded by the mutant HTT gene. Lowering HTT mRNA has been extensively studied as a potential therapeutic strategy, but how its level is regulated endogenously has been unclear. Here we report that the RNA-binding protein (RBP) HuR interacts with and stabilizes HTT mRNA in an mHTT-dependent manner. In HD cells but not wild-type cells, siRNA knockdown or CRISPR-induced heterozygous knockout of HuR decreased HTT mRNA stability. HuR interacted with HTT mRNA at a conserved site in exon 11 rather than the 3'-UTR region of the mRNA. Interestingly, this interaction was dependent on the presence of mHTT, likely via the activation of MAPK11, which enhanced cytosolic localization of the HuR protein. Thus, mHTT, MAPK11 and HuR may form a positive feedback loop that stabilizes HTT mRNA and enhances mHTT accumulation, which may contribute to HD progression. Our data reveal a novel regulatory mechanism of HTT mRNA via non-canonical binding of HuR.


Subject(s)
ELAV-Like Protein 1/metabolism , Huntingtin Protein/chemistry , Huntingtin Protein/genetics , Huntington Disease/genetics , Mitogen-Activated Protein Kinase 11/metabolism , Mutation , 3' Untranslated Regions , Animals , Binding Sites , CRISPR-Cas Systems , Cell Line , ELAV-Like Protein 1/genetics , Exons , Feedback, Physiological , Gene Knockdown Techniques , HEK293 Cells , Humans , Huntington Disease/metabolism , Mice , RNA Stability
8.
Int J Mol Sci ; 21(20)2020 Oct 12.
Article in English | MEDLINE | ID: mdl-33053909

ABSTRACT

The p38 mitogen-activated protein kinase (MAPK) signaling pathway is implicated in cancer biology and has been widely studied over the past two decades as a potential therapeutic target. Most of the biological and pathological implications of p38MAPK signaling are often associated with p38α (MAPK14). Recently, several members of the p38 family, including p38γ and p38δ, have been shown to play a crucial role in several pathologies including cancer. However, the specific role of p38ß (MAPK11) in cancer is still elusive, and further investigation is needed. Here, we summarize what is currently known about the role of p38ß in different types of tumors and its putative implication in cancer therapy. All evidence suggests that p38ß might be a key player in cancer development, and could be an important therapeutic target in several pathologies, including cancer.


Subject(s)
Disease Susceptibility , Mitogen-Activated Protein Kinase 11/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mitogen-Activated Protein Kinase 11/genetics , Multigene Family , Neoplasms/pathology , Signal Transduction
9.
Int J Mol Sci ; 21(3)2020 Jan 23.
Article in English | MEDLINE | ID: mdl-31979341

ABSTRACT

Estrogen (E2) and polyunsaturated fatty acids (n-3PUFA) supplements independently support general wellbeing and enhance muscle regeneration in-vivo and myotube formation in-vitro. However, the combined effect of E2 and n-3PUFA on myoblast differentiation is not known. The purpose of the study was to identify whether E2 and n-3PUFA possess a synergistic effect on in-vitro myogenesis. Mouse C2C12 myoblasts, a reliable model to reiterate myogenic events in-vitro, were treated with 10nM E2 and 50µM eicosapentaenoic acid (EPA) independently or combined, for 0-24 h or 0-120 h during differentiation. Immunofluorescence, targeted qPCR and next generation sequencing (NGS) were used to characterize morphological changes and differential expression of key genes involved in the regulation of myogenesis and muscle function pathways. E2 increased estrogen receptor α (Erα) and the expression of the mitogen-activated protein kinase 11 (Mapk11) within 1 h of treatment and improved myoblast differentiation and myotube formation. A significant reduction (p < 0.001) in myotube formation and in the expression of myogenic regulatory factors Mrfs (MyoD, Myog and Myh1) and the myoblast fusion related gene, Tmem8c, was observed in the presence of EPA and the combined E2/EPA treatment. Additionally, EPA treatment at 48 h of differentiation inhibited the majority of genes associated with the myogenic and striated muscle contraction pathways. In conclusion, EPA and E2 had no synergistic effect on myotube formation in-vitro. Independently, EPA inhibited myoblast differentiation and overrides the stimulatory effect of E2 when used in combination with E2.


Subject(s)
Eicosapentaenoic Acid/pharmacology , Estrogens/pharmacology , Muscle Development/drug effects , Muscle Fibers, Skeletal/drug effects , Myoblasts/drug effects , Myoblasts/metabolism , Animals , Cell Line , DNA Glycosylases/metabolism , Drug Synergism , Estrogen Receptor alpha/metabolism , Fatty Acids, Unsaturated/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Ontology , High-Throughput Nucleotide Sequencing , Membrane Proteins/metabolism , Mice , Mitogen-Activated Protein Kinase 11/metabolism , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , MyoD Protein/metabolism , Myoblasts/cytology , Myogenin/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptome/drug effects , Transcriptome/genetics
10.
Apoptosis ; 24(5-6): 395-403, 2019 06.
Article in English | MEDLINE | ID: mdl-30879166

ABSTRACT

Lipoapoptosis of cardiomyocytes may underlie diabetic cardiomyopathy. Numerous forms of cardiomyopathies share a common end-pathway in which apoptotic loss of cardiomyocytes is mediated by p38α mitogen activated protein kinase (MAPK). Although we have previously shown that palmitic acid (PA), a saturated fatty acid (SFA) elevated in plasma of type 2 diabetes mellitus and morbid obesity, induces apoptosis in cardiomyocytes via p38α MAPK-dependent signaling, the downstream cascade events that cause cell death remain unknown. The objective of this study was to investigate mechanisms involved in palmitic acid-induced cardiomyocyte apoptosis. Human adult ventricular cardiomyocyte line (AC16 cells) exposed to high physiological levels of PA for 16 h showed enhanced transcription and phosphorylation of c-fos and c-jun subunits of AP-1 and transcription of caspase 8. When AC16 cells were transfected with small interfering RNA specific against p38α MAPK (si-p38α) for 24 or 48 h, the amplified phosphorylation of c-fos was dose-dependently attenuated, and procaspase 8 was dose-dependently reduced. With translational knockdown of c-fos, PA-induced apoptosis was diminished. Inhibition of caspase 8 for 24 h reduced apoptosis in PA-treated cardiomyocytes. These findings provide evidence for induction of apoptosis in cardiomyocytes exposed to high SFA by a novel pathway requiring activation of c-fos/AP-1 and caspase 8. These results demonstrate how elevated plasma SFA may lead to continual and cumulative loss of cardiomyocytes and potentially contribute to the development of diabetic cardiomyopathy.


Subject(s)
Apoptosis , Caspase 8/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Myocytes, Cardiac/pathology , Palmitic Acid/metabolism , Transcription Factor AP-1/metabolism , Apoptosis/drug effects , Caspase 8/genetics , Caspase Inhibitors/pharmacology , Cell Line, Transformed , Humans , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 11/metabolism , Mitogen-Activated Protein Kinase 14/genetics , Myocytes, Cardiac/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , RNA, Small Interfering , Transcription Factor AP-1/genetics , Transcription, Genetic
11.
Dev Biol ; 425(1): 58-69, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28322734

ABSTRACT

The Drosophila vestigial gene is required for proliferation and differentiation of the adult wing and for differentiation of larval and adult muscle identity. Vestigial is part of a multi-protein transcription factor complex, which includes Scalloped, a TEAD-class DNA binding protein. Binding Scalloped is necessary for translocation of Vestigial into the nucleus. We show that Vestigial is extensively post-translationally modified and at least one of these modifications is required for proper function during development. We have shown that there is p38-dependent phosphorylation of Serine 215 in the carboxyl-terminal region of Vestigial. Phosphorylation of Serine 215 occurs in the nucleus and requires the presence of Scalloped. Comparison of a phosphomimetic and non-phosphorylatable mutant forms of Vestigial shows differences in the ability to rescue the wing and muscle phenotypes associated with a null vestigial allele.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Nuclear Proteins/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Cell Line , Cell Nucleus/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/growth & development , Immunoblotting , Microscopy, Confocal , Mitogen-Activated Protein Kinase 11/metabolism , Muscles/embryology , Muscles/metabolism , Mutation , Nuclear Proteins/metabolism , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Serine/genetics , Serine/metabolism , Transcription Factors/metabolism , Wings, Animal/growth & development , Wings, Animal/metabolism
12.
J Cell Physiol ; 233(2): 968-978, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28383766

ABSTRACT

TWEAK regulates multiple physio-pathological processes in fibroblasts such as fibrosis. It also induces migration and invasion in tumors and it can activate p38 MAPK in various cell types. Moreover, p38α MAPK promotes migration and invasion in several cancer cells types and in mouse embryonic fibroblasts (MEFs). However, it remains unknown if TWEAK could promote migration in fibroblasts and whether p38α MAPK might play a role. Our results reveal that TWEAK activates ERKs, Akt, and p38α/ß MAPKs and reduces secreted Fibulin 3 in MEFs. TWEAK also increases migration and invasion in wt and p38α deficient MEFs, which indicates that p38α MAPK is not required to mediate these effects. In contrast, ERKs inhibition significantly decreases TWEAK-induced migration and Fibulin 3 knock-down mimics TWEAK effect. These results indicate that both ERKs activation and Fibulin 3 down-regulation would contribute to mediate TWEAK pro-migratory effect. In fact, the additional regulation of ERKs and/or p38ß as a consequence of Fibulin 3 decrease might be also involved in the pro-migratory effect of TWEAK in MEFs. In conclusion, our studies uncover novel mechanisms by which TWEAK would favor tissue repair by promoting fibroblasts migration.


Subject(s)
Cell Movement , Cytokine TWEAK/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/enzymology , Animals , Cells, Cultured , Cytokine TWEAK/genetics , Down-Regulation , Enzyme Activation , Extracellular Matrix Proteins/genetics , Mice , Mitogen-Activated Protein Kinase 11/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Time Factors
13.
BMC Plant Biol ; 18(1): 203, 2018 Sep 20.
Article in English | MEDLINE | ID: mdl-30236054

ABSTRACT

BACKGROUND: The sucrose non-fermenting-1-related protein kinase 2 family (SnRK2s) unifies different abiotic stress signals in plants. To date, the functions of two rice SnRK2s, osmotic stress/ABA-activated protein kinase 1 (SAPK1) and SAPK2, have been unknown. We investigated their roles in response to salt stress by generating loss-of-function lines using the CRISPR/Cas9 system and by overexpressing these proteins in transgenic rice plants. RESULTS: Expression profiling revealed that SAPK1 and SAPK2 expression were strongly induced by drought, NaCl, and PEG treatment, but not by ABA. SAPK2 expression was highest in the leaves, followed by the roots, whereas SAPK1 was highest expressed in roots followed by leaves. Both proteins were localized to the nucleus and the cytoplasm. Under salt stress, sapk1, sapk2 and, in particular, sapk1/2 mutants, exhibited reduced germination rates, more severe growth inhibition, more distinct chlorosis, reduced chlorophyll contents, and reduced survival rates in comparison with the wild-type plants. In contrast, SAPK1- and SAPK2-overexpression lines had increased germination rates and reduced sensitivities to salt; including mild reductions in growth inhibition, reduced chlorosis, increased chlorophyll contents and improved survival rates in comparison with the wild-type plants. These results suggest that SAPK1 and SAPK2 may function collaboratively as positive regulators of salt stress tolerance at the germination and seedling stages. We also found that SAPK1 and SAPK2 affected the osmotic potential following salt stress by promoting the generation of osmotically active metabolites such as proline. SAPK1 and SAPK2 also improved reactive oxygen species (ROS) detoxification following salt stress by promoting the generation of ROS scavengers such as ascorbic acid, and by increasing the expression levels of proteins such as superoxide dismutase (SOD) and catalase (CAT). SAPK1 and SAPK2 may function collaboratively in reducing Na+ toxicity by affecting the Na+ distribution between roots and shoots, Na+ exclusion from the cytoplasm, and Na+ sequestration into the vacuoles. These effects may be facilitated through the expression of Na+-and K+-homeostasis-related genes. CONCLUSION: SAPK1 and SAPK2 may function collaboratively as positive regulators of salt stress tolerance at the germination and seedling stages in rice. SAPK1 and SAPK2 may be useful to improve salt tolerance in crop plants.


Subject(s)
Mitogen-Activated Protein Kinase 11/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Oryza/physiology , Salt Tolerance/physiology , Chlorophyll/genetics , Chlorophyll/metabolism , Gene Expression Regulation, Plant , Germination , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 8/genetics , Mutation , Oryza/genetics , Osmosis , Plants, Genetically Modified/genetics , Potassium/metabolism , Reactive Oxygen Species/metabolism , Salt Tolerance/genetics , Seedlings/physiology , Sodium/metabolism
14.
Brain Behav Immun ; 72: 34-44, 2018 08.
Article in English | MEDLINE | ID: mdl-29128611

ABSTRACT

p38 mitogen-activated protein kinase (MAPK) consists of two major isoforms: p38α and p38ß; however, it remains unclear which isoform is more important for chronic pain development. Recently, we developed potent, long-lasting, and p38 MAPK subtype-specific antisense oligonucleotides (ASOs). We examined the therapeutic effects of isoform-specific ASOs in several chronic pain models following single intrathecal injection (300 µg/10 µl) in CD1 mice. In the chronic constriction injury (CCI) model, p38α MAPK ASO, given on post-operative day 5, reduced CCI-induced mechanical allodynia in male but not female mice. In contrast, mechanical allodynia after CCI in both sexes was not affected by p38ß MAPK ASO. Intrathecal injection of p38α or p38ß ASO resulted in a partial reduction (≈ 50%) of spinal p38α or p38ß mRNA level, respectively, in both sexes at two weeks. In contrast, intrathecal injection of the ASOs did not affect p38α and p38ß MAPK mRNA levels in dorsal root ganglia. Intrathecal p38α ASO also reduced postoperative pain (mechanical and cold allodynia) in male mice after tibia fracture. However, intrathecal p38α ASO had no effect on mechanical allodynia in male mice after paclitaxel treatment. Intrathecal p38α MAPK ASO pre-treatment also prevented TLR4-mediated mechanical allodynia and downregulated levels of p38α MAPK and phosphorylated p38 MAPK following intrathecal treatment of lipopolysaccharide. In summary, our findings suggest that p38α MAPK is the major p38 MAPK isoform in the spinal cord and regulates chronic pain in a sex and model-dependent manner. Intrathecal p38α MAPK ASO may offer a new treatment for some chronic pain conditions.


Subject(s)
Neuralgia/therapy , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Disease Models, Animal , Down-Regulation , Female , Ganglia, Spinal/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/therapy , Injections, Spinal , Male , Mice , Microglia/metabolism , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 11/metabolism , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/genetics , Pain Measurement , Pain, Postoperative/therapy , Peripheral Nervous System Diseases/metabolism , Phosphorylation , Protein Isoforms , Spinal Cord/metabolism , Toll-Like Receptor 4/metabolism
15.
Immunol Cell Biol ; 95(9): 814-823, 2017 10.
Article in English | MEDLINE | ID: mdl-28611474

ABSTRACT

The p38 mitogen-activated protein kinase (MAPK) pathway is involved in the regulation of immune and inflammatory processes. We used p38α-conditional, p38ß-deficient and p38α/ß double-null mouse models to address the role of these two p38 MAPK in CD4+ T cells, and found that p38α deficiency causes these cells to hyperproliferate. Our studies indicate that both p38α and p38ß are dispensable for T helper cell type 1 (Th1) differentiation but, by controlling interferon (IFN)γ and tumor necrosis factor (TNF)α production, are critical for normal Th1 effector function. We found that both p38α and p38ß modulate T-cell receptor-induced IFNγ and TNFα production, whereas only p38α regulates cytokine-induced IFNγ production. The lack of p38α and p38ß did not affect transcription and mRNA stability of Ifng. However, the absence of p38α in Th1 cells resulted in a decreased MNK1 phosphorylation after cytokine activation, and MNK1 inhibition blocked IFNγ production. Our results indicate that p38α regulates IFNγ secretion through the activation of the MNK1/eIF4E pathway of translation initiation and identify specific functions for p38α and p38ß in T-cell proliferation.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Mitogen-Activated Protein Kinase 11/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Protein Serine-Threonine Kinases/metabolism , Th1 Cells/physiology , Animals , Cell Proliferation/genetics , Cells, Cultured , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 14/genetics , Phosphorylation , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
16.
Am J Physiol Cell Physiol ; 309(7): C491-500, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26246429

ABSTRACT

The p38 MAP kinases play critical roles in skeletal muscle biology, but the specific processes regulated by these kinases remain poorly defined. Here we find that activity of p38α/ß is important not only in early phases of myoblast differentiation, but also in later stages of myocyte fusion and myofibrillogenesis. By treatment of C2 myoblasts with the promyogenic growth factor insulin-like growth factor (IGF)-I, the early block in differentiation imposed by the p38 chemical inhibitor SB202190 could be overcome. Yet, under these conditions, IGF-I could not prevent the later impairment of muscle cell fusion, as marked by the nearly complete absence of multinucleated myofibers. Removal of SB202190 from the medium of differentiating myoblasts reversed the fusion block, as multinucleated myofibers were detected several hours later and reached ∼90% of the culture within 30 h. Analysis by quantitative mass spectroscopy of proteins that changed in abundance following removal of the inhibitor revealed a cohort of upregulated muscle-enriched molecules that may be important for both myofibrillogenesis and fusion. We have thus developed a model system that allows separation of myoblast differentiation from muscle cell fusion and should be useful in identifying specific steps regulated by p38 MAP kinase-mediated signaling in myogenesis.


Subject(s)
Cell Differentiation/drug effects , Imidazoles/pharmacology , Insulin-Like Growth Factor I/pharmacology , Mitogen-Activated Protein Kinase 11/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Pyridines/pharmacology , Animals , Cell Fusion , Cells, Cultured , Mice , Mice, Inbred C3H , Mitogen-Activated Protein Kinase 11/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Muscle Development/physiology , Myoblasts, Skeletal/cytology , Signal Transduction/drug effects
17.
Mol Med ; 21: 626-36, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26225830

ABSTRACT

We recently confirmed that angiotensin II (Ang II) type 1 receptor (AT1R) was overexpressed in hepatocellular carcinoma tissue using a murine hepatoma model. Angiotensin(Ang)-(1-7) has been found beneficial in ameliorating lung cancer and prostate cancer. Which receptor of Ang-(1-7) is activated to mediate its effects is much speculated. This study was designed to investigate the effects of Ang-(1-7) on hepatocellular carcinoma, as well as the probable mechanisms. H22 hepatoma-bearing mice were randomly divided into five groups for treatment: mock group, low-dose Ang-(1-7), high-dose Ang-(1-7), high-dose Ang-(1-7) + A779 and high-dose Ang-(1-7) + PD123319. Ang-(1-7) treatment inhibited tumor growth time- and dose-dependently by arresting tumor proliferation and promoting tumor apoptosis as well as inhibiting tumor angiogenesis. The effects of Ang-(1-7) on tumor proliferation and apoptosis were reversed by coadministration with A779 or PD123319, whereas the effects on tumor angiogenesis were completely reversed by A779 but not by PD123319. Moreover, Ang-(1-7) downregulated AT1R mRNA, upregulated mRNA levels of Ang II type 2 receptor (AT2R) and Mas receptor (MasR) and p38-MAPK phosphorylation and suppressed H22 cell-endothelial cell communication. Thus, Ang-(1-7) administration suppresses hepatocellular carcinoma via complex interactions of AT1R, AT2R and MasR and may provide a novel and promising approach for the treatment of hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Neovascularization, Pathologic/genetics , Receptor, Angiotensin, Type 1/biosynthesis , Receptor, Angiotensin, Type 2/biosynthesis , Angiotensin I/administration & dosage , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Mice , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 11/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Peptide Fragments/administration & dosage , Phosphorylation , Proto-Oncogene Mas , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 2/genetics , Xenograft Model Antitumor Assays
18.
J Vasc Res ; 52(5): 347-59, 2015.
Article in English | MEDLINE | ID: mdl-27029009

ABSTRACT

R-Ras is a Ras family small GTPase that is highly expressed in mature functional blood vessels in normal tissues. It inhibits pathological angiogenesis and promotes vessel maturation and stabilization. Previous studies suggest that R-Ras affects cellular signaling in endothelial cells, pericytes and smooth-muscle cells to regulate vessel formation and remodeling in adult tissues. R-Ras suppresses VEGF-induced endothelial permeability and vessel sprouting while promoting normalization of pathologically developing vessels in mice. It attenuates VEGF receptor-2 (VEGFR2) activation by inhibiting internalization of the receptor upon VEGF ligand binding, leading to significant reduction of VEGFR2 autophosphorylation. Here, we show that R-Ras strongly suppresses the VEGF-dependent activation of stress-activated protein kinase-2/p38 mitogen-activated protein kinase (SAPK2/p38MAPK) and the phosphorylation of downstream heat-shock protein 27 (HSP27), a regulator of actin cytoskeleton organization, in endothelial cells. The suppression of p38MAPK activation and HSP27 phosphorylation by R-Ras concurred with altered actin cytoskeleton architecture, reduced membrane protrusion and inhibition of endothelial cell migration toward VEGF. Silencing of endogenous R-Ras by RNA interference increased membrane protrusion and cell migration stimulated by VEGF, and these effects were offset by p38MAPK inhibitor SB203580. These results suggest that R-Ras regulates angiogenic activities of endothelial cells in part via inhibition of the p38MAPK-HSP27 axis of VEGF signaling.


Subject(s)
HSP27 Heat-Shock Proteins/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Mitogen-Activated Protein Kinase 11/metabolism , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/pharmacology , ras Proteins/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Cell Movement/drug effects , Cells, Cultured , Enzyme Activation , Heat-Shock Proteins , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Mitogen-Activated Protein Kinase 11/antagonists & inhibitors , Molecular Chaperones , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA Interference , Signal Transduction/drug effects , Time Factors , Transfection , ras Proteins/genetics
19.
Fish Shellfish Immunol ; 47(1): 331-43, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26363230

ABSTRACT

The p38 kinases are one of the four subgroups of mitogen-activated protein kinase (MAPK) superfamily which are involved in the innate immunity. The p38 subfamily that includes four members namely p38α (MAPK14), p38ß (MAPK11), p38γ (MAPK12) and p38δ (MAPK13), regulates the activation of several transcription factors. In this study, a p38ß (OfMAPK11) homolog and a p38α (OfMAPK14) homolog of Oplegnathus fasciatus were identified at genomic level. Results clearly showed that both MAPK11 and MAPK14 are well-conserved at both genomic structural- and amino acid (aa)-levels. Genomic sequences of OfMAPK11 (∼ 15.6 kb) and OfMAPK14 (∼ 13.4 kb) had 12 exons. A comparison of exon-intron structural arrangement of these genes from different vertebrate lineages indicated that all the exon lengths are highly conserved, except their terminal exons. Full-length cDNAs of OfMAPK11 (3957 bp) and OfMAPK14 (2504 bp) encoded corresponding proteins of 361 aa and 360 aa, respectively. Both OfMAPK proteins harbored a Ser/Thr protein kinases catalytic domain (S_TKc domain) which includes an activation loop with a dual phosphorylation site (TGY motif) and several specific-binding sites for ATP and substrates. Molecular modeling of the activation loop and substrate binding sites of rock bream MAPKs revealed the conservation of crucial residues and their orientation in 3D space. Transcripts of OfMAPKs were ubiquitously detected in eleven tissues examined, however at different levels. The modulation of OfMAPKs' transcription upon pathogen-associated molecular patterns (PAMPs: flagellin, lipopolysaccharide and poly I:C) and pathogens (Edwardsiella tarda, Streptococcus iniae and rock bream iridovirus) was investigated. Among the seven examined tissues, the flagellin-challenge upregulated the mRNA level of both OfMAPKs in the head kidney. Meanwhile, modulation of OfMAPK mRNA expression in the liver upon other immune-challenges varied in a time-dependent manner. Collectively, these results suggest that OfMAPKs are true members of p38 subfamily, which might be induced by different immune stimuli.


Subject(s)
Fish Diseases/genetics , Fish Proteins/genetics , Gene Expression Regulation , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 14/genetics , Perciformes/genetics , Perciformes/metabolism , Amino Acid Sequence , Animals , Base Sequence , DNA Virus Infections/genetics , DNA Virus Infections/immunology , DNA Virus Infections/veterinary , DNA Virus Infections/virology , Edwardsiella tarda/physiology , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/veterinary , Fish Diseases/immunology , Fish Diseases/microbiology , Fish Diseases/virology , Fish Proteins/chemistry , Fish Proteins/metabolism , Flagellin/pharmacology , Iridoviridae/physiology , Lipopolysaccharides/pharmacology , Mitogen-Activated Protein Kinase 11/chemistry , Mitogen-Activated Protein Kinase 11/metabolism , Mitogen-Activated Protein Kinase 14/chemistry , Mitogen-Activated Protein Kinase 14/metabolism , Organ Specificity , Perciformes/classification , Phylogeny , Poly I-C/pharmacology , Sequence Alignment/veterinary , Streptococcal Infections/genetics , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcal Infections/veterinary , Streptococcus/physiology
20.
J Biol Chem ; 288(23): 17030-17041, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23625915

ABSTRACT

The Ca(2+) sensor STIM1 is crucial for activation of store-operated Ca(2+) entry (SOCE) through transient receptor potential canonical and Orai channels. STIM1 phosphorylation serves as an "off switch" for SOCE. However, the signaling pathway for STIM1 phosphorylation is unknown. Here, we show that SOCE activates AMP-activated protein kinase (AMPK); its effector p38ß mitogen-activated protein kinase (p38ß MAPK) phosphorylates STIM1, thus inhibiting SOCE in human lung microvascular endothelial cells. Activation of AMPK using 5-aminoimidazole-4-carboxamide-1-ß-d-ribofuranoside (AICAR) resulted in STIM1 phosphorylation on serine residues and prevented protease-activated receptor-1 (PAR-1)-induced Ca(2+) entry. Furthermore, AICAR pretreatment blocked PAR-1-induced increase in the permeability of mouse lung microvessels. Activation of SOCE with thrombin caused phosphorylation of isoform α1 but not α2 of the AMPK catalytic subunit. Moreover, knockdown of AMPKα1 augmented SOCE induced by thrombin. Interestingly, SB203580, a selective inhibitor of p38 MAPK, blocked STIM1 phosphorylation and led to sustained STIM1-puncta formation and Ca(2+) entry. Of the three p38 MAPK isoforms expressed in endothelial cells, p38ß knockdown prevented PAR-1-mediated STIM1 phosphorylation and potentiated SOCE. In addition, inhibition of the SOCE downstream target CaM kinase kinase ß (CaMKKß) or knockdown of AMPKα1 suppressed PAR-1-mediated phosphorylation of p38ß and hence STIM1. Thus, our findings demonstrate that SOCE activates CaMKKß-AMPKα1-p38ß MAPK signaling to phosphorylate STIM1, thereby suppressing endothelial SOCE and permeability responses.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Calcium/metabolism , Endothelial Cells/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 11/metabolism , Neoplasm Proteins/metabolism , Receptor, PAR-1/metabolism , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Calcium Channels , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cells, Cultured , Endothelial Cells/cytology , Gene Knockdown Techniques , Humans , Hypoglycemic Agents/pharmacology , Lung/blood supply , Lung/metabolism , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mitogen-Activated Protein Kinase 11/genetics , Neoplasm Proteins/genetics , Phosphorylation/drug effects , Phosphorylation/physiology , Receptor, PAR-1/genetics , Ribonucleotides/pharmacology , Stromal Interaction Molecule 1
SELECTION OF CITATIONS
SEARCH DETAIL