Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
1.
Chembiochem ; 25(1): e202300551, 2024 01 02.
Article in English | MEDLINE | ID: mdl-37856284

ABSTRACT

Stress-activated signaling pathways orchestrate cellular behaviors and fates. Studying the precise role(s) of stress-activated protein kinases is challenging, because stress conditions induce adaptation and impose selection pressure. To meet this challenge, we have applied an optogenetic system with a single plasmid to express light-activated p38α or its upstream activator, MKK6, in conjunction with live-cell fluorescence microscopy. In starved cells, decaging of constitutively active p38α or MKK6 by brief exposure to UV light elicits rapid p38-mediated signaling, release of cytochrome c from mitochondria, and apoptosis with different kinetics. In parallel, light activation of p38α also suppresses autophagosome formation, similarly to stimulation with growth factors that activate PI3K/Akt/mTORC1 signaling. Active MKK6 negatively regulates serum-induced ERK activity, which is p38-independent as previously reported. Here, we reproduce that result with the one plasmid system and show that although decaging active p38α does not reduce basal ERK activity in our cells, it can block growth factor-stimulated ERK signaling in serum-starved cells. These results clarify the roles of MKK6 and p38α in dynamic signaling programs, which act in concert to actuate apoptotic death while suppressing cell survival mechanisms.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases , Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases , p38 Mitogen-Activated Protein Kinases , MAP Kinase Kinase 6/genetics
2.
Aging (Albany NY) ; 14(19): 7959-7971, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36205565

ABSTRACT

OBJECTIVE: Apurinic/apyrimidinic endonuclease 1 (APEX1), a key enzyme responsible for DNA base excision repair, has been linked to development and progression of cancers. In this work, we aimed to explore the role of APEX1 in hepatocellular carcinoma (HCC) and elucidate its molecular mechanism. METHODS: The expression of APEX1 in HCC tissues and matched adjacent normal tissues (n = 80 cases) was evaluated by immunohistochemistry. Web-based tools UALCAN and the Kaplan-Meier plotter were used to analyze the Cancer Genome Atlas database to compare expression of APEX1 mRNA to 5-year overall survival. APEX1 was stably silenced in two HCC cell lines, Hep 3B and Bel-7402, with shRNA technology. An in vivo tumorigenesis model was established by subcutaneously injecting sh-APEX1-transfected Bel-7402 cells into mice, and tumor growth was determined. We performed high-throughput transcriptome sequencing in sh-APEX1-treated HCC cells to identify the key KEGG signaling pathways induced by silencing of APEX1. RESULTS: APEX1 was significantly upregulated and predicted poor clinical overall survival in HCC patients. Silencing APEX1 inhibited the proliferation of HCC cells in vivo and in vitro, and it repressed invasion and migration and increased apoptosis and the percentage of cells in G1. Differentially expressed genes upon APEX1 silencing included genes involved in TNF signaling. A positive correlation between the expression of APEX1 and MAP2K6 was noted, and overexpressing MAP2K6 overcame cancer-related phenotypes associated with APEX1 silencing. CONCLUSION: APEX1 enhances the malignant properties of HCC via MAP2K6. APEX1 may represent a valuable prognostic biomarker and therapeutic target in HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Biomarkers , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , DNA , Endonucleases/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/pathology , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , RNA, Messenger , RNA, Small Interfering/genetics , Humans
3.
Viruses ; 14(10)2022 09 30.
Article in English | MEDLINE | ID: mdl-36298734

ABSTRACT

Interferon gamma (IFN-γ) may be potential adjuvant immunotherapy for COVID-19 patients. In this work, we assessed gene expression profiles associated with the IFN-γ pathway in response to SARS-CoV-2 infection. Employing a case-control study from SARS-CoV-2-positive and -negative patients, we identified IFN-γ-associated pathways to be enriched in positive patients. Bioinformatics analyses showed upregulation of MAP2K6, CBL, RUNX3, STAT1, and JAK2 in COVID-19-positive vs. -negative patients. A positive correlation was observed between STAT1/JAK2, which varied alongside the patient's viral load. Expression of MX1, MX2, ISG15, and OAS1 (four well-known IFN-stimulated genes (ISGs)) displayed upregulation in COVID-19-positive vs. -negative patients. Integrative analyses showcased higher levels of ISGs, which were associated with increased viral load and STAT1/JAK2 expression. Confirmation of ISGs up-regulation was performed in vitro using the A549 lung cell line treated with Poly (I:C), a synthetic analog of viral double-stranded RNA; and in different pulmonary human cell lines and ferret tracheal biopsies infected with SARS-CoV-2. A pre-clinical murine model of Coronavirus infection confirmed findings displaying increased ISGs in the liver and lungs from infected mice. Altogether, these results demonstrate the role of IFN-γ and ISGs in response to SARS-CoV-2 infection, highlighting alternative druggable targets that can boost the host response.


Subject(s)
COVID-19 , Humans , Animals , Mice , Interferon-gamma/genetics , SARS-CoV-2 , Case-Control Studies , RNA, Double-Stranded , Ferrets , MAP Kinase Kinase 6/genetics
4.
Elife ; 112022 08 16.
Article in English | MEDLINE | ID: mdl-35971771

ABSTRACT

Stress-activated p38 kinases control a plethora of functions, and their dysregulation has been linked to the development of steatosis, obesity, immune disorders, and cancer. Therefore, they have been identified as potential targets for novel therapeutic strategies. There are four p38 family members (p38α, p38ß, p38γ, and p38δ) that are activated by MKK3 and MKK6. Here, we demonstrate that lack of MKK6 reduces the lifespan in mice. Longitudinal study of cardiac function in MKK6 KO mice showed that young mice develop cardiac hypertrophy which progresses to cardiac dilatation and fibrosis with age. Mechanistically, lack of MKK6 blunts p38α activation while causing MKK3-p38γ/δ hyperphosphorylation and increased mammalian target of rapamycin (mTOR) signaling, resulting in cardiac hypertrophy. Cardiac hypertrophy in MKK6 KO mice is reverted by knocking out either p38γ or p38δ or by inhibiting the mTOR pathway with rapamycin. In conclusion, we have identified a key role for the MKK3/6-p38γ/δ pathway in the development of cardiac hypertrophy, which has important implications for the clinical use of p38α inhibitors in the long-term treatment since they might result in cardiotoxicity.


The human heart can increase its size to supply more blood to the body's organs. This process, called hypertrophy, can happen during exercise or be caused by medical conditions, such as high blood pressure or inherited genetic diseases. If hypertrophy is continually driven by illness, this can cause the heart to fail and no longer be able to properly pump blood around the body. For hypertrophy to happen, several molecular changes occur in the cells responsible for contracting the heart, including activation of the p38 pathway. Within this pathway is a p38 enzyme as well as a series of other proteins which are sequentially turned on in response to stress, such as inflammatory molecules or mechanical forces that alter the cell's shape. There are different types of p38 enzyme which have been linked to other diseases, making them a promising target for drug development. However, clinical trials blocking individual members of the p38 family have had disappointing results. An alternative approach is to target other proteins involved in the p38 pathway, such as MKK6, but it is not known what effect this might have. To investigate, Romero-Becerra et al. genetically modified mice to not have any MKK6 protein. As a result, these mice had a shorter lifespan, with hypertrophy developing at a young age that led to heart problems. Romero-Becerra et al. used different mice models to understand why this happened, showing that a lack of MKK6 reduces the activity of a specific member of the p38 family called p38α. However, this blockage boosted a different branch of the pathway which involved two other p38 proteins, p38γ and p38δ. This, in turn, triggered another key pathway called mTOR which also promotes hypertrophy of the heart. These results suggest that drugs blocking MKK6 and p38α could lead to side effects that cause further harm to the heart. A more promising approach for treating hypertrophic heart conditions could be to inhibit p38γ and/or p38δ. However, before this can be fully explored, further work is needed to generate compounds that specifically target these proteins.


Subject(s)
Heart Diseases , MAP Kinase Kinase 6 , Mitogen-Activated Protein Kinase 13 , Animals , Cardiomegaly , Heart Diseases/genetics , Heart Diseases/pathology , Longitudinal Studies , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase 6/genetics , Mice , Mitogen-Activated Protein Kinase 13/metabolism , TOR Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Cell Death Differ ; 29(5): 1042-1054, 2022 05.
Article in English | MEDLINE | ID: mdl-34815549

ABSTRACT

Somatic cell reprogramming is an ideal model for studying epigenetic regulation as it undergoes dramatic chromatin remodeling. However, a role for phosphorylation signaling in chromatin protein modifications for reprogramming remains unclear. Here, we identified mitogen-activated protein kinase kinase 6 (Mkk6) as a chromatin relaxer and found that it could significantly enhance reprogramming. The function of Mkk6 in heterochromatin loosening and reprogramming requires its kinase activity but does not depend on its best-known target, P38. We identified Gatad2b as a novel target of Mkk6 phosphorylation that acts downstream to elevate histone acetylation levels and loosen heterochromatin. As a result, Mkk6 over-expression facilitates binding of Sox2 and Klf4 to their targets and promotes pluripotency gene expression during reprogramming. Our studies not only reveal an Mkk phosphorylation mediated modulation of chromatin status in reprogramming, but also provide new rationales to further investigate and improve the cell fate determination processes.


Subject(s)
Chromatin , Heterochromatin , Cellular Reprogramming , Epigenesis, Genetic , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , Phosphorylation
6.
Int J Mol Sci ; 22(24)2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34948353

ABSTRACT

Obesity is a state of abnormal fat accumulation caused by an energy imbalance potentially caused by changes in multiple factors. MEK6 engages in cell growth, such as inflammation and apoptosis, as one of the MAPK signaling pathways. The MEK6 gene was found to be related to RMR, a gene associated with obesity. Because only a few studies have investigated the correlation between MEK6 and obesity or the relevant mechanisms, we conducted an experiment using a TgMEK6 model with MEK6 overexpression with non-Tg and chow diet as the control to determine changes in lipid metabolism in plasma, liver, and adipose tissue after a 15-week high-fat diet (HFD). MEK6 overexpression in the TgMEK6 model significantly increased body weight and plasma triglyceride and total cholesterol levels. p38 activity declined in the liver and adipose tissues and lowered lipolysis, oxidation, and thermogenesis levels, contributing to decreased energy consumption. In the liver, lipid formation and accumulation increased, and in adipose, adipogenesis and hypertrophy increased. The adiponectin/leptin ratio significantly declined in plasma and adipose tissue of the TgMEK6 group following MEK6 expression and the HFD, indicating the role of MEK6 expression in adipokine regulation. Plasma and bone-marrow-derived macrophages (BMDM) of the TgMEK6 group increased MEK6 expression-dependent secretion of pro-inflammatory cytokines but decreased levels of anti-inflammatory cytokines, further exacerbating the results exhibited by the diet-induced obesity group. In conclusion, this study demonstrated the synergistic effect of MEK6 with HFD in fat accumulation by significantly inhibiting the mechanisms of lipolysis in the adipose and M2 associated cytokines secretion in the BMDM.


Subject(s)
Cytokines/analysis , Fats/analysis , Inflammation/genetics , MAP Kinase Kinase 6/genetics , Obesity/genetics , Adipose Tissue/pathology , Animals , Diet, High-Fat/adverse effects , Inflammation/etiology , Inflammation/pathology , Male , Mice, Inbred C57BL , Obesity/etiology , Obesity/pathology , Up-Regulation
7.
Clin Transl Med ; 11(10): e613, 2021 10.
Article in English | MEDLINE | ID: mdl-34709743

ABSTRACT

BACKGROUND: The mitogen-activated protein kinase (MAPK) pathway is highly associated with the progression and metastasis of various solid tumours. MAPK14, a core molecule of the MAPK pathway, plays vital roles in the colorectal cancer (CRC). Recent studies have shown that circRNAs can affect tumour progression by encoding peptides. However, little is known regarding the potential protein translated from circMAPK14 and whether it plays a role in the carcinogenesis of colorectal cancer. METHODS: The RNA level and translatable potential of circMAPK14 in CRC was verified using qRT-PCR and public databases. RNase R digestion assay, qRT-PCR, sanger sequencing and FISH assays were utilised to verify the circular characteristics and subcellular localisation of circMAPK14. The suppressive role of circMAPK14 on the progression and metastasis of CRC was verified in vivo and in vitro. LC/MS analysis combined with western blotting demonstrated the presence and relative expression of circMAPK14-175aa. The underlying mechanism of circMAPK14-175aa action to inhibit CRC was identified by co-IP analysis. The binding of U2AF2 within the flanking introns of circMAPK14 was evaluated by RNA pull-down assay and RIP assay. Ultimately, luciferase reporter gene assays and ChIP assays confirmed that FOXC1 suppressed transcription of U2AF2 by binding to the U2AF2 promoter in the -400 bp to -100 bp region.  RESULTS: We identified that hsa_circ_0131663 (termed circMAPK14) showed significantly decreased expression level in cells and tissue samples of CRC, and was primarily localised in the cytoplasm. A series of function experiments demonstrated that circMAPK14 influenced CRC progression and metastasis by encoding a peptide of 175 amino acids (termed circMAPK14-175aa). We also found that circMAPK14-175aa reduced nuclear translocation of MAPK14 by competitively binding to MKK6, thus facilitating ubiquitin-mediated degradation of FOXC1. Moreover, we described a positive feedback loop in CRC in which elevated FOXC1 expression was caused by reduced circMAPK14-175aa expression. This, in turn, decreased circMAPK14 biogenesis by suppressing U2AF2 transcription. CONCLUSION: In summary, we reported for the first time that circMAPK14 functioned as a tumour-suppressor by encoding circMAPK14-175aa, which blocked the progression and metastasis of colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Proliferation/genetics , Cell-Free Nucleic Acids/genetics , Cell-Free Nucleic Acids/metabolism , Colorectal Neoplasms/metabolism , Disease Models, Animal , Disease Progression , Female , Genes, Tumor Suppressor , Humans , Mice , Mice, Inbred BALB C
8.
Cartilage ; 13(2_suppl): 785S-795S, 2021 12.
Article in English | MEDLINE | ID: mdl-34581623

ABSTRACT

OBJECTIVE: Osteoarthritis (OA) is a degenerative joint disease characterized by deterioration of articular cartilage functions. Previous studies have confirmed the role of circular RNAs (circRNAs) in OA, but the role of mechanical stress-related circRNA (circRNA-MSR) in OA is unknown. DESIGN: The human chondrocytes C28/I2 were cultured and treated with lipopolysaccharide (LPS) to establish the OA model. The mRNA and protein levels were measured by qRT-PCR or Western blot. Cell viability was analyzed by MTT assay. Flow cytometry was carried out to detect cell apoptosis. The levels of TNF-α, IL-1ß, and IL-6 were determined by enzyme-linked immunosorbent assay (ELISA). Pull-down assay was conducted to measure circRNA-MSR-related miRNA. Dual-luciferase reporter gene detection was performed to detect the target relationships between miR-643 and circRNA-MSR or Mitogen-activated protein kinase kinase 6 (MAP2K6). The RNA-fluorescence in situ hybridization (RNA-FISH) assay was conducted to verify the localization of circRNA-MSR and miR-643. RESULTS: The expressions of circRNA-MSR were upregulated in LPS stimulated C28/I2 cells. Knockdown of circRNA-MSR can inhibit LPS-induced apoptosis, inflammatory response, and extracellular matrix (ECM) degradation, and promote cell C28/I2 cells proliferation. Moreover, circRNA-MSR directly targeted miR-643. RNA-FISH exhibited that circRNA-MSR may act as a competing endogenous RNA (ceRNA) of miR-643. Over-expression of miR-643 could alleviate LPS-induced C28/I2 chondrocyte injury and promote cell proliferation. Besides, miR-643 directly bound to MAP2K6 mRNA. MiR-643 inhibition or MAP2K6 overexpression can reverse the role of circRNA-MSR knockdown on LPS-treated chondrocytes. CONCLUSION: circRNA-MSR can upregulate MAP2K6 by targeting miR-643, thereby inhibiting cell proliferation and promoting apoptosis of C28/I2 cells.


Subject(s)
Cartilage, Articular , MicroRNAs , Osteoarthritis , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Humans , In Situ Hybridization, Fluorescence , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , MicroRNAs/genetics , Osteoarthritis/genetics , Osteoarthritis/metabolism , RNA, Circular/genetics , Signal Transduction
9.
Cells ; 10(5)2021 05 04.
Article in English | MEDLINE | ID: mdl-34064531

ABSTRACT

To investigate whether the beiging process changes the interactive effects of salt and MEK6 gene on inflammatory adipogenesis, the salt treatment (NaCl 50 mM) and MEK6 transfection of Tg(+/+) cells were performed with white adipocytes (WAT) and beige-like-adipocytes (BLA). BLA induced by T3 were confirmed by UCP-1 expression and the MEK6 protein was 3.5 times higher in MEK6 transfected WAT than the control. The adipogenic genes, PPAR-γ and C/EBP-α, were 1.5 times more highly expressed in the salt-treated groups than the non-salt-treated groups, and adipogenesis was greatly increased in Tg(+/+) WAT compared to non-transfected Tg(-/-). The adipogenesis induced by salt treatment and MEK6 transfection was significantly reduced in BLA. The inflammatory adipocytokines, TNF-α, IL-1ß, and IL-6, were increased in the salt-treated Tg(+/+) WAT, but an anti-inflammation biomarker, the adiponectin/leptin ratio, was reduced in Tg(+/+), to tenth of that in Tg(-/-). However, the production of adipocytokines in WAT was strongly weakened in BLA, although a combination of salt and MEK6 transfection had the most significant effects on inflammation in both WAT and BLA. Oxygen consumption in mitochondria was maximized in salt-treated and MEK6 transfected WAT, but it was decreased by 50% in BLA. In conclusion, beiging controls the synergistic effects of salt and MEK6 on adipogenesis, inflammation, and energy expenditure.


Subject(s)
Adipocytes, Beige/metabolism , Adipocytes, White/metabolism , Adipogenesis , MAP Kinase Kinase 6/metabolism , Sodium Chloride/pharmacology , 3T3-L1 Cells , Adipocytes, Beige/cytology , Adipocytes, White/cytology , Adipocytes, White/drug effects , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Energy Metabolism , MAP Kinase Kinase 6/genetics , Mice , PPAR gamma/genetics , PPAR gamma/metabolism
10.
Protein Sci ; 30(4): 908-913, 2021 04.
Article in English | MEDLINE | ID: mdl-33554397

ABSTRACT

Mitogen-activated protein kinase (MAPK; p38, ERK, and JNK) cascades are evolutionarily conserved signaling pathways that regulate the cellular response to a variety of extracellular stimuli, such as growth factors and interleukins. The MAPK p38 is activated by its specific upstream MAPK kinases, MKK6 and MKK3. However, a comprehensive molecular understanding of how these cognate upstream kinases bind and activate p38 is still missing. Here, we combine NMR spectroscopy and isothermal titration calorimetry to define the binding interface between full-length MKK6 and p38. It was shown that p38 engages MKK6 not only via its hydrophobic docking groove, but also influences helix αF, a secondary structural element that plays a key role in organizing the kinase core. It was also shown that, unlike MAPK phosphatases, the p38 conserved docking (CD) site is much less affected by MKK6 binding. Finally, it was demonstrated that these interactions with p38 are conserved independent of the MKK6 activation state. Together, the results revealed differences between specificity markers of p38 regulation by upstream kinases, which do not effectively engage the CD site, and downstream phosphatases, which require the CD site for productive binding.


Subject(s)
MAP Kinase Kinase 6/chemistry , p38 Mitogen-Activated Protein Kinases/chemistry , Binding Sites , Enzyme Activation , Humans , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation, alpha-Helical , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Mol Syst Biol ; 17(1): e9684, 2021 01.
Article in English | MEDLINE | ID: mdl-33417276

ABSTRACT

To elucidate the contributions of specific lipid species to metabolic traits, we integrated global hepatic lipid data with other omics measures and genetic data from a cohort of about 100 diverse inbred strains of mice fed a high-fat/high-sucrose diet for 8 weeks. Association mapping, correlation, structure analyses, and network modeling revealed pathways and genes underlying these interactions. In particular, our studies lead to the identification of Ifi203 and Map2k6 as regulators of hepatic phosphatidylcholine homeostasis and triacylglycerol accumulation, respectively. Our analyses highlight mechanisms for how genetic variation in hepatic lipidome can be linked to physiological and molecular phenotypes, such as microbiota composition.


Subject(s)
Diet, High-Fat/adverse effects , Fatty Liver/genetics , Glucose/adverse effects , Insulin Resistance/genetics , MAP Kinase Kinase 6/genetics , Nuclear Proteins/genetics , Animals , Disease Models, Animal , Fatty Liver/chemically induced , Fatty Liver/metabolism , Gene Expression Profiling , Gene Expression Regulation , Genetic Variation , Lipidomics , Male , Mice , Phosphatidylcholines/metabolism , Triglycerides/metabolism
12.
Mol Med Rep ; 22(3): 1803-1810, 2020 09.
Article in English | MEDLINE | ID: mdl-32582987

ABSTRACT

Transdifferentiation of lung fibroblasts to myofibroblasts is a crucial pathophysiological process in pulmonary fibrosis. MicroRNA­375 (miR­375) was initially identified as a tumor­suppressive factor, and its expression was negatively associated with the severity of lung cancer; however, its role and potential mechanism in myofibroblast transdifferentiation and pulmonary fibrosis remain unclear. In the present study, human lung fibroblasts were stimulated with transforming growth factor­ß (TGF­ß) to induce myofibroblast transdifferentiation. A mimic and inhibitor of miR­375, and their negative controls, were used to overexpress or suppress miR­375 in lung fibroblasts, respectively. The mRNA expression levels of fibrotic markers, and protein expression of α­smooth muscle actin and periostin, were subsequently detected by reverse transcription­quantitative PCR and western blotting, to assess myofibroblast transdifferentiation. miR­375 was markedly upregulated in human lung fibroblasts after TGF­ß stimulation. The miR­375 mimic alleviated, whereas the miR­375 inhibitor aggravated TGF­ß­dependent transdifferentiation of lung fibroblasts. Mechanistically, miR­375 prevented myofibroblast transdifferentiation and collagen synthesis by blocking the P38 mitogen­activated protein kinases (P38) pathway, and P38 suppression abrogated the deleterious effect of the miR­375 inhibitor on myofibroblast transdifferentiation. Furthermore, the present study revealed that mitogen­activated protein kinase kinase 6 was involved in P38 inactivation by miR­375. In conclusion, miR­375 was implicated in modulating TGF­ß­dependent transdifferentiation of lung fibroblasts, and targeting miR­375 expression may help to develop therapeutic approaches for treating pulmonary fibrosis.


Subject(s)
Lung/cytology , MicroRNAs/genetics , Transforming Growth Factor beta/adverse effects , Up-Regulation , Cell Line , Cell Transdifferentiation , Collagen/metabolism , Humans , Lung/chemistry , Lung/drug effects , MAP Kinase Kinase 6/genetics , MAP Kinase Signaling System/drug effects , MicroRNAs/antagonists & inhibitors , Myofibroblasts/chemistry , Myofibroblasts/cytology , Myofibroblasts/drug effects
13.
J Biol Chem ; 295(25): 8494-8504, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32371393

ABSTRACT

The selective pressure imposed by extrinsic death signals and stressors adds to the challenge of isolating and interpreting the roles of proteins in stress-activated signaling networks. By expressing a kinase with activating mutations and a caged lysine blocking the active site, we can rapidly switch on catalytic activity with light and monitor the ensuing dynamics. Applying this approach to MAP kinase 6 (MKK6), which activates the p38 subfamily of MAPKs, we found that decaging active MKK6 in fibroblasts is sufficient to trigger apoptosis in a p38-dependent manner. Both in fibroblasts and in a murine melanoma cell line expressing mutant B-Raf, MKK6 activation rapidly and potently inhibited the pro-proliferative extracellular signal-regulated kinase (ERK) pathway; to our surprise, this negative cross-regulation was equally robust when all p38 isoforms were inhibited. These results position MKK6 as a new pleiotropic signal transducer that promotes both pro-apoptotic and anti-proliferative signaling, and they highlight the utility of caged, light-activated kinases for dissecting stress-activated signaling networks.


Subject(s)
MAP Kinase Kinase 6/metabolism , Signal Transduction/radiation effects , Ultraviolet Rays , Animals , Apoptosis/radiation effects , Cell Line , Cell Proliferation/radiation effects , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , MAP Kinase Kinase 6/genetics , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutagenesis , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Mol Cell Probes ; 49: 101478, 2020 02.
Article in English | MEDLINE | ID: mdl-31698040

ABSTRACT

BACKGROUND: Stroke is a leading cause of mortality worldwide. Rac-MAPK kinase 6 (Map2k6) plays important roles in cell proliferation and apoptosis. However, the role played by Map2k6 in stroke injury and the underlying mechanism of action remain unknown. METHODS: Mice received cerebral ischemia/reperfusion (I/R) injuries by transient middle cerebral artery occlusion. HT22 cells were subjected to oxygen glucose deprivation and reoxygenation (OGD/R) to simulate an I/R injury. Subsequently, the levels of circ_016719, miR-29c and Map2k6 expression were determined, and their interactions were examined by luciferase assays. Circ_016719 knockdown, miR-29c inhibition or Map2k6 overexpression was induced in HT22 cells; after which, the cells were examined for their viability, apoptosis, autophagy and proliferation, as well their levels of Map2k6, p38, p53, LC3B-I, LC3B-II, Beclin 1, and p62 expression. RESULTS: Significantly increased levels of circ_016719 and Map2k6, and decreased levels of miR-29c were observed in both in vivo and in vitro I/R injury models. In HT22 cells, circ_016719 knockdown significantly increased miR-29c expression and cell proliferation, but decreased Map2k6 expression and cell apoptosis. Additionally, significant increases in LC3B-I and p62 levels and decreased LC3B-II levels were observed, indicating that circ_016719 knockdown had significantly inhibited autophagy. Furthermore, additional inhibition of miR-29c markedly suppressed the effects of circ_016719 knockdown; however, that suppression was significantly attenuated by Map2k6 overexpression. Additionally, Map2k6 was identified as a direct target of miR-29c, which in turn, might be sponged by circ_016719. CONCLUSIONS: Our results suggest that circ_016719 directly targets miR-29c, and thereby regulates the expression and functions of Map2k6, which significantly contributes to the pro-apoptotic role of circ_016719.


Subject(s)
Apoptosis , MAP Kinase Kinase 6/metabolism , MicroRNAs/metabolism , Neurons/metabolism , Neurons/pathology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Animals , Autophagy , Base Sequence , Cell Line , Cell Survival , Disease Models, Animal , Gene Expression Regulation , Glucose , MAP Kinase Kinase 6/genetics , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Neurons/ultrastructure , Oxygen , RNA, Circular , Stroke/genetics
15.
Cytokine ; 126: 154862, 2020 02.
Article in English | MEDLINE | ID: mdl-31634687

ABSTRACT

Stimulation of H295R adrenocortical carcinoma cells with angiotensin II or cytokines induces the secretion of the chemokine interleukin-8 (IL-8). Here, we have analyzed the molecular mechanism of stimulus-induced IL-8 expression. IL-8 expression and IL-8 promoter activity increased in H295R cells expressing an activated Gαq-coupled designer receptor. H295R cells stimulated with either interleukin-1ß (IL-1ß) or phorbol ester also showed elevated IL-8 mRNA levels and higher IL-8 promoter activities. Deletion and point mutations of the IL-8 promoter revealed that the AP-1 binding site within the IL-8 promoter is essential to connect designer receptor stimulation with the transcriptional activation of the IL-8 gene. Expression of a constitutively active mutant of c-Jun, or expression of constitutively active mutants of the protein kinases MEKK1 and MKK6 confirmed that the IL-8 gene is a bona fide target of AP-1 in adrenocortical carcinoma cells. Upregulation of IL-8 expression in IL-1ß-treated H295R cells required NF-κB while the phorbol ester TPA used both the AP-1 and NF-κB sites of the IL-8 gene to stimulate IL-8 expression. These data were corroborated in experiments with chromatin-embedded AP-1 or NF-κB-responsive reporter genes. While stimulation of Gαq-coupled designer receptors increased the AP-1 activity in the cells, IL-1ß specifically stimulated NF-κB-regulated transcription. Stimulation of the cells with TPA increased both AP-1 and NF-κB activities. We conclude that stimulation of Gαq-coupled designer receptors or IL-1 receptors triggers distinct signaling pathways in H295R cells leading to the activation of either AP-1 or NF-κB. Nevertheless, both signaling cascades converge to the IL-8 gene, inducing IL-8 gene transcription.


Subject(s)
Adrenal Cortex Neoplasms/metabolism , Adrenocortical Carcinoma/metabolism , Interleukin-8/metabolism , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , Adrenal Cortex Neoplasms/genetics , Adrenocortical Carcinoma/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Genes, jun/genetics , Humans , Interleukin-1beta/pharmacology , Interleukin-8/genetics , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 1/metabolism , Point Mutation , Promoter Regions, Genetic , Sequence Deletion , Tetradecanoylphorbol Acetate/pharmacology , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Up-Regulation
16.
Fish Shellfish Immunol ; 92: 500-507, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31247318

ABSTRACT

Mitogen-activated protein kinase 6 (MKK6) is one of the major important central regulatory proteins response to environmental and physiological stimuli. In this study, a novel MKK6, EcMKK6, was isolated from Epinephelus coioides, an economically important cultured fish in China and Southeast Asian counties. The open reading frame (ORF) of EcMKK6 is 1077 bp encoding 358 amino acids. EcMKK6 contains a serine/threonine protein kinase (S_TKc) domain, a tyrosine kinase catalytic domain, a conserved dual phosphorylation site in the SVAKT motif and a conserved DVD domain. By in situ hybridization (ISH) with Digoxigenin-labeled probe, EcMKK6 mainly located at the cytoplasm of cells, and a little appears in the nucleus. EcMKK6 mRNA can be detected in all eleven tissues examined, but the expression level is different in these tissues. After challenge with Vibrio alginolyticus and Singapore grouper iridovirus (SGIV), the transcription level of EcMKK6 was apparently up-regulated in the tissues examined. The data demonstrated that the sequence and the characters of EcMKK6 were conserved, EcMKK6 showed tissue-specific expression profiles in healthy grouper, and the expression was significantly varied after pathogen infection, indicating that EcMKK6 may play important roles in E. coioides during pathogen-caused inflammation.


Subject(s)
Bass/genetics , Bass/immunology , Fish Diseases/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/immunology , Amino Acid Sequence , Animals , DNA Virus Infections/immunology , DNA Virus Infections/veterinary , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , MAP Kinase Kinase 6/chemistry , Phylogeny , Ranavirus/physiology , Sequence Alignment/veterinary , Vibrio Infections/immunology , Vibrio Infections/veterinary , Vibrio alginolyticus/physiology
17.
Cell Rep ; 27(12): 3473-3485.e5, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31216469

ABSTRACT

Reprogramming somatic cells to pluripotency by Oct4, Sox2, Klf4, and Myc represent a paradigm for cell fate determination. Here, we report a combination of Jdp2, Jhdm1b, Mkk6, Glis1, Nanog, Essrb, and Sall4 (7F) that reprogram mouse embryonic fibroblasts or MEFs to chimera competent induced pluripotent stem cells (iPSCs) efficiently. RNA sequencing (RNA-seq) and ATAC-seq reveal distinct mechanisms for 7F induction of pluripotency. Dropout experiments further reveal a highly cooperative process among 7F to dynamically close and open chromatin loci that encode a network of transcription factors to mediate reprogramming. These results establish an alternative paradigm for reprogramming that may be useful for analyzing cell fate control.


Subject(s)
Cellular Reprogramming/genetics , DNA-Binding Proteins/metabolism , F-Box Proteins/metabolism , Induced Pluripotent Stem Cells/cytology , Jumonji Domain-Containing Histone Demethylases/metabolism , MAP Kinase Kinase 6/metabolism , Nanog Homeobox Protein/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Chimera/genetics , DNA-Binding Proteins/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Euchromatin/genetics , Euchromatin/metabolism , F-Box Proteins/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Heterochromatin/genetics , Heterochromatin/metabolism , Induced Pluripotent Stem Cells/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Kruppel-Like Factor 4 , MAP Kinase Kinase 6/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nanog Homeobox Protein/genetics , RNA-Seq , Repressor Proteins/genetics , Transcription Factors/genetics
18.
Cell Death Dis ; 10(6): 376, 2019 05 15.
Article in English | MEDLINE | ID: mdl-31092814

ABSTRACT

Apoptosis and senescence are two mutually exclusive cell fate programs that can be activated by stress. The factors that instruct cells to enter into senescence or apoptosis are not fully understood, but both programs can be regulated by the stress kinase p38α. Using an inducible system that specifically activates this pathway, we show that sustained p38α activation suffices to trigger massive autophagosome formation and to enhance the basal autophagic flux. This requires the concurrent effect of increased mitochondrial reactive oxygen species production and the phosphorylation of the ULK1 kinase on Ser-555 by p38α. Moreover, we demonstrate that macroautophagy induction by p38α signaling determines that cancer cells preferentially enter senescence instead of undergoing apoptosis. In agreement with these results, we present evidence that the induction of autophagy by p38α protects cancer cells from chemotherapy-induced apoptosis by promoting senescence. Our results identify a new mechanism of p38α-regulated basal autophagy that controls the fate of cancer cells in response to stress.


Subject(s)
Autophagy , Cellular Senescence , Mitogen-Activated Protein Kinase 14/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Cell Line, Tumor , Cellular Senescence/drug effects , Doxorubicin/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase 6/antagonists & inhibitors , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 12/deficiency , Mitogen-Activated Protein Kinase 12/genetics , Mitogen-Activated Protein Kinase 12/metabolism , Mitogen-Activated Protein Kinase 14/deficiency , Mitogen-Activated Protein Kinase 14/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Signal Transduction
19.
Methods Enzymol ; 614: 187-205, 2019.
Article in English | MEDLINE | ID: mdl-30611424

ABSTRACT

Phosphorylation is a ubiquitous posttranslational modification that is essential for the regulation of many cellular processes. The human genome consists of more than 200,000 phosphorylation sites, whose phosphorylation is tightly controlled by ≥500 kinases and ~200 phosphatases. Given the large number of phosphorylation sites and the key role phosphorylation plays in regulating cellular processes, it is essential to characterize the impact of phosphorylation on substrate structure, dynamics, and function. However, a major challenge is the large-scale production of phosphorylated proteins in vitro for these structural, functional, and dynamic studies. Here, we describe an efficient protocol used routinely in our laboratory for the production of phosphorylated proteins. We also describe the methods used for identifying, characterizing, and separating the resulting phosphorylated proteins for subsequent studies.


Subject(s)
Ki-67 Antigen/metabolism , MAP Kinase Kinase 6/metabolism , Nuclear Magnetic Resonance, Biomolecular/methods , Protein Processing, Post-Translational , p38 Mitogen-Activated Protein Kinases/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Electrophoresis, Polyacrylamide Gel/methods , Humans , Ki-67 Antigen/genetics , MAP Kinase Kinase 6/genetics , Phosphorylation , Substrate Specificity , p38 Mitogen-Activated Protein Kinases/genetics
20.
Fish Shellfish Immunol ; 84: 795-801, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30393177

ABSTRACT

Mitogen-activated protein kinase kinase 6 (MKK6) is an essential component of the p38MAPK signaling pathway, which is involved in the modulation of inflammation, cell apoptosis and survival responses in mammals. However, the function of MKK6s in teleosts is still unclear. In this study, a fish MKK6 homolog (CiMKK6) was first identified from the grass carp (Ctenopharyngodon idella), a freshwater fish. CiMKK6 cDNA encodes a putative protein of 357 amino acids that contains conserved structural characteristics of the MKK6 family, including the S_TKc domain, SVAKT motif and DVD site. The deduced CiMKK6 protein exhibits high sequence homology with other reported fish MKK6s and shares the closest relationship with MKK6 from Danio rerio. Quantitative real-time PCR (qRT-PCR) analysis revealed that CiMKK6 mRNA was widely expressed in all tested tissues and stages of embryonic development. Additionally, the transcript levels of CiMKK6 in the intestine were significantly upregulated in response to bacterial muramyl dipeptide (MDP) and L-Ala-γ-D-Glu-meso-diaminopimelic acid (Tri-DAP) stimulation. Moreover, subcellular localization analysis indicated that CiMKK6 was distributed in both the cytoplasm and the nucleus of HEK293T cells. Finally, overexpression of CiMKK6 significantly enhanced the transcriptional activity of the AP-1 reporter gene in HEK293T cells. Overall, these findings may help better clarify the immune function of teleost MKK6s and provide new insight into the immune defense mechanisms of grass carp.


Subject(s)
Bacterial Proteins/immunology , Carps/genetics , Carps/immunology , Immunity, Innate/genetics , MAP Kinase Kinase 6/genetics , Animals , Bacterial Proteins/administration & dosage , Dipeptides/administration & dosage , Dipeptides/immunology , Fish Proteins/genetics , Fish Proteins/metabolism , HEK293 Cells , Humans , MAP Kinase Kinase 6/metabolism , Oligopeptides/administration & dosage , Oligopeptides/immunology , Random Allocation
SELECTION OF CITATIONS
SEARCH DETAIL
...