Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
1.
Physiol Rep ; 12(11): e16108, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38872461

ABSTRACT

ERK3/MAPK6 activates MAP kinase-activated protein kinase (MK)-5 in selected cell types. Male MK5 haplodeficient mice show reduced hypertrophy and attenuated increase in Col1a1 mRNA in response to increased cardiac afterload. In addition, MK5 deficiency impairs cardiac fibroblast function. This study determined the effect of reduced ERK3 on cardiac hypertrophy following transverse aortic constriction (TAC) and fibroblast biology in male mice. Three weeks post-surgery, ERK3, but not ERK4 or p38α, co-immunoprecipitated with MK5 from both sham and TAC heart lysates. The increase in left ventricular mass and myocyte diameter was lower in TAC-ERK3+/- than TAC-ERK3+/+ hearts, whereas ERK3 haploinsufficiency did not alter systolic or diastolic function. Furthermore, the TAC-induced increase in Col1a1 mRNA abundance was diminished in ERK3+/- hearts. ERK3 immunoreactivity was detected in atrial and ventricular fibroblasts but not myocytes. In both quiescent fibroblasts and "activated" myofibroblasts isolated from adult mouse heart, siRNA-mediated knockdown of ERK3 reduced the TGF-ß-induced increase in Col1a1 mRNA. In addition, intracellular type 1 collagen immunoreactivity was reduced following ERK3 depletion in quiescent fibroblasts but not myofibroblasts. Finally, knocking down ERK3 impaired motility in both atrial and ventricular myofibroblasts. These results suggest that ERK3 plays an important role in multiple aspects of cardiac fibroblast biology.


Subject(s)
Fibroblasts , Animals , Male , Mice , Fibroblasts/metabolism , Collagen Type I/metabolism , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain/metabolism , Myocardium/metabolism , Myocardium/cytology , Mitogen-Activated Protein Kinase 6/metabolism , Mitogen-Activated Protein Kinase 6/genetics , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Cells, Cultured , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/genetics , Myocytes, Cardiac/metabolism
2.
Eur Spine J ; 33(5): 2056-2067, 2024 May.
Article in English | MEDLINE | ID: mdl-38551688

ABSTRACT

OBJECTIVE: This research was to unravel the impact of the lncRNA differentiation antagonizing non-protein coding RNA (DANCR)/microRNA (miR)-146a-5p/mitogen-activated protein kinase 6 (MAPK6) axis on spinal cord injury (SCI). METHODS: SCI mouse models were established and injected with si-DANCR or miR-146a-5p agomir. The recovery of motor function was assessed by Basso Mouse Scale. SCI was pathologically evaluated, and serum inflammatory factors were measured in SCI mice. Mouse spinal cord neurons were injured by H2O2 and transfected, followed by assessment of proliferation and apoptosis. DANCR, miR-146a-5p, and MAPK6 in tissues and cells were detected, as well as their relationship. RESULTS: DANCR increased and miR-146a-5p decreased in SCI. Silencing DANCR or enhancing miR-146a-5p stimulated the proliferation of mouse spinal cord neurons and reduced apoptosis. DANCR was bound to miR-146a-5p to target MAPK6. DANCR affected the proliferation and apoptosis of spinal cord neurons by mediating the miR-146a-5p/MAPK6 axis. Downregulating DANCR or upregulating miR-146a-5p improved inflammation, the destruction of spinal cord tissue structure, and apoptosis in SCI mice. CONCLUSION: DANCR affects spinal cord neuron apoptosis and inflammation of SCI by mediating the miR-146a-5p/MAPK6 axis.


Subject(s)
Apoptosis , MicroRNAs , Neurons , RNA, Long Noncoding , Spinal Cord Injuries , Animals , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Apoptosis/genetics , Apoptosis/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Mice , Neurons/metabolism , Mitogen-Activated Protein Kinase 6/genetics , Mitogen-Activated Protein Kinase 6/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Inflammation/genetics , Inflammation/metabolism , Mice, Inbred C57BL , Male
3.
Int Immunopharmacol ; 124(Pt B): 110984, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37757635

ABSTRACT

Diabetic cardiomyopathy (DCM) is a challenging diabetic complication that manifests as chronic inflammation. Yet, the mechanism underlying diabetes-associated myocardial injury is not fully understood. We investigated the pharmacological effects and mechanisms of linderalactone, a natural compound that can prevent diabetes-induced cardiomyopathy in mice. Diabetes was induced by a single dose of streptozotocin (120 mg/kg, i.p.). Diabetic mice were administrated with linderalactone (2.5 or 5 mg/kg) by gavage for five weeks. Harvested heart tissues were then subjected to RNA-sequencing analysis to explore the potential mechanism of linderalactone. Linderalactone prevented heart dysfunction by inhibiting myocardial hypertrophy, fibrosis, and inflammation, without altering blood glucose. RNA-sequencing indicated that linderalactone exerted its cardioprotective effects mainly by affecting the mitogen-activated protein kinase (MAPK)/ activating transcription factor 6 (ATF6) pathway. Linderalactone also suppressed endoplasmic reticulum (ER) stress mediated by the diabetes-activated MAPKs/ATF6 pathway, thereby reducing myocardial hypertrophy and inflammation in heart tissues and in cultured cardiomyocytes. Inhibition of MAPKs or a deficiency of ATF6 in cardiomyocytes mimicked the linderalactone-associated decreases in high glucose-induced hypertrophy and inflammation. Linderalactone showed beneficial effects in alleviating diabetic cardiomyopathy, in part by modulating the MAPK/ATF6 signaling pathway to mitigate myocardial hypertrophy and inflammation. Linderalactone may have clinical utility in the treatment for diabetes-associated cardiomyopathy.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Mice , Animals , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/metabolism , Diabetes Mellitus, Experimental/metabolism , Mitogen-Activated Protein Kinase 6/metabolism , Myocytes, Cardiac/metabolism , Inflammation/metabolism , Hypertrophy , RNA/metabolism
4.
Elife ; 122023 04 14.
Article in English | MEDLINE | ID: mdl-37057894

ABSTRACT

The actin cytoskeleton is tightly controlled by RhoGTPases, actin binding-proteins and nucleation-promoting factors to perform fundamental cellular functions. We have previously shown that ERK3, an atypical MAPK, controls IL-8 production and chemotaxis (Bogueka et al., 2020). Here, we show in human cells that ERK3 directly acts as a guanine nucleotide exchange factor for CDC42 and phosphorylates the ARP3 subunit of the ARP2/3 complex at S418 to promote filopodia formation and actin polymerization, respectively. Consistently, depletion of ERK3 prevented both basal and EGF-dependent RAC1 and CDC42 activation, maintenance of F-actin content, filopodia formation, and epithelial cell migration. Further, ERK3 protein bound directly to the purified ARP2/3 complex and augmented polymerization of actin in vitro. ERK3 kinase activity was required for the formation of actin-rich protrusions in mammalian cells. These findings unveil a fundamentally unique pathway employed by cells to control actin-dependent cellular functions.


Subject(s)
Actins , Mitogen-Activated Protein Kinase 6 , Animals , Humans , Actins/metabolism , Mitogen-Activated Protein Kinase 6/metabolism , Polymerization , Cell Movement , Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Mammals/metabolism , rac1 GTP-Binding Protein/metabolism
5.
J Transl Med ; 21(1): 186, 2023 03 09.
Article in English | MEDLINE | ID: mdl-36895010

ABSTRACT

BACKGROUND: Breast cancer (BC) is a common malignant tumor in women worldwide. Circular RNA (circRNA) has been proven to play a critical role in BC progression. However, the exact biological functions and underlying mechanisms of circRNAs in BC remain largely unknown. METHODS: Here, we first screened for differentially expressed circRNAs in 4 pairs of BC tissues and adjacent non-tumor tissues using a circRNA microarray. Functionally, gain- and loss-of-function experiments in vitro and in vivo showed that circDNAJC11 promoted BC cell proliferation, migration, invasion, and tumor growth. Mechanistically, RNA pull-down, mass spectrum, RNA immunoprecipitation, fluorescence in situ hybridization assays, and rescue experiments were executed. RESULTS: We found that circDNAJC11 was significantly upregulated in triple-negative breast cancer tissues and cells. Clinical data revealed that the high expression of circDNAJC11 was closely correlated with a poor prognosis of BC patients and could be an independent risk factor for BC prognosis. Functionally, gain- and loss-of-function experiments in vitro and in vivo showed that circDNAJC11 promoted BC cell proliferation, migration, invasion, and tumor growth. Mechanistically, RNA pull-down, mass spectrum, RNA immunoprecipitation, fluorescence in situ hybridization assays, and rescue experiments were executed. We demonstrated that circDNAJC11 combined with TAF15 to promote BC progression via stabilizing MAPK6 mRNA and activating the MAPK signaling pathway. CONCLUSIONS: The circDNAJC11/TAF15/MAPK6 axis played a crucial role in the progression and development of BC, suggesting that circDNAJC11 might be a novel biomarker and therapeutical target for BC.


Subject(s)
Breast Neoplasms , MicroRNAs , TATA-Binding Protein Associated Factors , Triple Negative Breast Neoplasms , Female , Humans , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , In Situ Hybridization, Fluorescence , MicroRNAs/genetics , RNA, Circular/genetics , Signal Transduction/genetics , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , Triple Negative Breast Neoplasms/genetics , Mitogen-Activated Protein Kinase 6/metabolism
6.
Biochem Biophys Res Commun ; 612: 119-125, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35523049

ABSTRACT

Kinases represent one of the largest druggable families of proteins. Importantly, many kinases are aberrantly activated/de-activated in multiple organs during obesity, which contributes to the development of diabetes and associated diseases. Previous results indicate that the complex between Extracellular-regulated kinase 3 (ERK3) and Mitogen-Activated Protein Kinase (MAPK)-activated protein kinase 5 (MK5) suppresses energy dissipation and promotes fatty acids (FAs) output in adipose tissue and, therefore promotes obesity and diabetes. However, the therapeutic potential of targeting this complex at the systemic level has not been fully explored. Here we applied a translational approach to target the ERK3/MK5 complex in mice. Importantly, deletion of ERK3 in the whole body or administration of MK5-specific inhibitor protects against obesity and promotes insulin sensitivity. Finally, we show that the expression of ERK3 and MK5 correlates with the degree of obesity and that ERK3/MK5 complex regulates energy dissipation in human adipocytes. Altogether, we demonstrate that ERK3/MK5 complex can be targeted in vivo to preserve metabolic health and combat obesity and diabetes.


Subject(s)
Diabetes Mellitus , Protein Serine-Threonine Kinases , Animals , Intracellular Signaling Peptides and Proteins , Mice , Mitogen-Activated Protein Kinase 6/metabolism , Obesity
7.
Cancer Gene Ther ; 29(8-9): 1285-1295, 2022 08.
Article in English | MEDLINE | ID: mdl-35228660

ABSTRACT

Lung squamous cell carcinoma (LUSC) represents one of the commonest types of lung cancer featured with high morbidity and poor prognosis. Many types of research have documented that long noncoding RNAs (lncRNAs) exert crucial functions in the development of cancers, and LUSC is included. In our study, we aimed at unveiling the mechanism underlying long intergenic nonprotein coding RNA 0649 (LINC00649) in LUSC cells. As a result, LINC00649 was discovered to be with high expression in LUSC cells. Moreover, it was confirmed through functional assays that the knockdown of LINC00649 hindered the occurrence and progression of LUSC. Results of mechanism assays validated that E2F transcription factor 7 (E2F7) was a transcription activator of LINC00649 and induced its up-regulation in LUSC cells. Furthermore, LINC00649 recruited TAF15, which is TATA-box binding protein associated factor 15 to stabilize mitogen-activated protein kinase 6 (MAPK6) expression and activate the transcription of MAPK6, thereby enhancing MAPK6 expression to activate the MAPK signaling pathway. Eventually, results of rescue assays suggested that overexpression of MAPK6 offset the influence of LINC00649 silencing on LUSC progression. In summary, our research determined the E2F7/LINC00649/TAF15/MAPK6/MAPK signaling pathway in regulating LUSC development, which made LINC00649 a potential biomarker for LUSC treatment.


Subject(s)
Carcinoma, Squamous Cell , Lung Neoplasms , Mitogen-Activated Protein Kinase 6/metabolism , RNA, Long Noncoding , TATA-Binding Protein Associated Factors , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic , Humans , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Signal Transduction , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism
8.
Oncogene ; 41(12): 1752-1766, 2022 03.
Article in English | MEDLINE | ID: mdl-35124697

ABSTRACT

EGLN3 is critically important for growth of various cancers including lung cancer. However, virtually nothing is known about the role and mechanism for EGLN3 hydroxylase activity in cancers. EGLN3 catalyzes the hydroxylation of extracellular signal-regulated kinase 3 (Erk3), a potent driver of cancers. The role and mechanism for EGLN3-induced stabilization of Erk3 remain to be defined. Here, we show that Erk3 interacts with heat shock cognate protein of 70 kDa (HSC70) and lysosome-associated membrane protein type 2 A (LAMP2A), two core components of chaperone-mediated autophagy (CMA). As a consequence, Erk3 is degraded by the CMA-lysosome pathway. EGLN3-catalyzed hydroxylation antagonizes CMA-dependent destruction of Erk3. Mechanistically, hydroxylation blunts the interaction of Erk3 with LAMP2A, thereby blocking lysosomal decay of Erk3. EGLN3 inactivation inhibits macrophage migration, efferocytosis, and M2 polarization. Studies using EGLN3 catalytically inactive knock-in mice indicate that inactivation of EGLN3 hydroxylase in host cells ameliorates LLC cancer growth through reprogramming the tumor microenvironment (TME). Adoptive transfer of macrophages with inactivated EGLN3 restrains tumor growth by mounting anti-tumor immunity and restricting angiogenesis. Administration of EGLN3 hydroxylase pharmacologic inhibitor to mice bearing LLC carcinoma impedes cancer growth by targeting the TME. LLC cells harboring inactivated EGLN3 exhibit reduced tumor burden via mitigating immunosuppressive milieu and inducing cancer senescence. This study provides novel insights into the role of CMA in regulating Erk3 stability and the mechanism behind EGLN3-enhanced stability of Erk3. This work demonstrates that inactivation of EGLN3 in malignant and stromal cells suppresses tumor by orchestrating reciprocal interplays between cancer cells and the TME. This work sheds new light on the role and mechanism for EGLN3 catalytic activity in regulating cancer growth. Manipulating EGLN3 activity holds promise for cancer treatment.


Subject(s)
Lung Neoplasms , Mitogen-Activated Protein Kinase 6 , Animals , Autophagy , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lysosomes/metabolism , Mice , Mitogen-Activated Protein Kinase 6/metabolism , Mixed Function Oxygenases/metabolism , Tumor Microenvironment
9.
J Cell Physiol ; 237(4): 2271-2287, 2022 04.
Article in English | MEDLINE | ID: mdl-35141958

ABSTRACT

The physiological functions and downstream effectors of the atypical mitogen-activated protein kinase extracellular signal-regulated kinase 3 (ERK3) remain to be characterized. We recently reported that mice expressing catalytically-inactive ERK3 (Mapk6KD/KD ) exhibit a reduced postnatal growth rate as compared to control mice. Here, we show that genetic inactivation of ERK3 impairs postnatal skeletal muscle growth and adult muscle regeneration after injury. Loss of MAPK-activated protein kinase 5 (MK5) phenocopies the muscle phenotypes of Mapk6KD/KD mice. At the cellular level, genetic or pharmacological inactivation of ERK3 or MK5 induces precocious differentiation of C2C12 or primary myoblasts, concomitant with MyoD activation. Reciprocally, ectopic expression of activated MK5 inhibits myogenic differentiation. Mechanistically, we show that MK5 directly phosphorylates FoxO3, promoting its degradation and reducing its association with MyoD. Depletion of FoxO3 rescues in part the premature differentiation of C2C12 myoblasts observed upon inactivation of ERK3 or MK5. Our findings reveal that ERK3 and its substrate MK5 act in a linear signaling pathway to control postnatal myogenic differentiation.


Subject(s)
Forkhead Box Protein O3/metabolism , Signal Transduction , Animals , Intracellular Signaling Peptides and Proteins , Mice , Mitogen-Activated Protein Kinase 6/metabolism , Muscles , Protein Serine-Threonine Kinases/metabolism
10.
Bioengineered ; 13(2): 4517-4527, 2022 02.
Article in English | MEDLINE | ID: mdl-35137672

ABSTRACT

Chronic kidney disease (CKD) in clinical is defined as a gradual loss of kidney function for more than 3 months. The pathologic course of CKD is characterized by extensive renal fibrosis; thus, preventing renal fibrosis is vital for the treatment of CKD. It has been reported that microRNA (miR)-374a-5p was under-expressed in renal venous blood samples from patients with CKD. In addition, it exhibited anti-apoptotic effects in renal tissues suggesting that miR-374a-5p may play an important role in CKD. However, it is not clear whether miR-374a-5p could be delivered to renal cells by exosomes and exerts anti-renal fibrosis effects. To mimic renal fibrosis in vitro, human renal tubular epithelial cell lines (HK-2 cells) were treated by transforming growth factor-ß (TGF-ß) 1. Reverse transcription-quantitative polymerase-chain reaction (RT-qPCR) or Western blot was carried out to evaluate the mechanism by which miR-374a-5p regulated the development of renal fibrosis. Next, exosomes were isolated using with ultracentrifugation method, and the relationship between miR-374a-5p and MAPK6 was evaluated using dual-Luciferase a reporter assay system. The results indicated TGF-ß1 significantly down-regulated the expression of miR-374a-5p in HK-2 cells and miR-374a-5p agomir remarkably inhibited the progression of fibrosis in vitro. In addition, exosomal miR-374a-5p could be internalized by HK-2 cells and obviously enhanced the level of miR-374a-5p in HK-2 cells. Furthermore, exosomal miR-374a-5p prevented the progression of renal fibrosis in vivo by regulating MAPK6/MK5/YAP axis. In conclusion, exosomal miR-374a-5p inhibited the progression of renal fibrosis by regulating MAPK6/MK5/YAP axis.


Subject(s)
Exosomes/chemistry , Kidney Diseases , Mesenchymal Stem Cells/cytology , Mitogen-Activated Protein Kinase 6/metabolism , Animals , Cell Line , Cells, Cultured , Exosomes/metabolism , Fibrosis , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Mesenchymal Stem Cells/chemistry , Mice , Mice, Inbred C57BL , MicroRNAs , Mitogen-Activated Protein Kinase 6/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics
11.
Exp Mol Med ; 54(1): 35-46, 2022 01.
Article in English | MEDLINE | ID: mdl-35022544

ABSTRACT

Extracellular signal-regulated kinase 3 (ERK3) is an atypical member of the mitogen-activated protein kinase (MAPK) family, members of which play essential roles in diverse cellular processes during carcinogenesis, including cell proliferation, differentiation, migration, and invasion. Unlike other MAPKs, ERK3 is an unstable protein with a short half-life. Although deubiquitination of ERK3 has been suggested to regulate the activity, its ubiquitination has not been described in the literature. Here, we report that FBXW7 (F-box and WD repeat domain-containing 7) acts as a ubiquitination E3 ligase for ERK3. Mammalian two-hybrid assay and immunoprecipitation results demonstrated that ERK3 is a novel binding partner of FBXW7. Furthermore, complex formation between ERK3 and the S-phase kinase-associated protein 1 (SKP1)-cullin 1-F-box protein (SCF) E3 ligase resulted in the destabilization of ERK3 via a ubiquitination-mediated proteasomal degradation pathway, and FBXW7 depletion restored ERK3 protein levels by inhibiting this ubiquitination. The interaction between ERK3 and FBXW7 was driven by binding between the C34D of ERK3, especially at Thr417 and Thr421, and the WD40 domain of FBXW7. A double mutant of ERK3 (Thr417 and Thr421 to alanine) abrogated FBXW7-mediated ubiquitination. Importantly, ERK3 knockdown inhibited the proliferation of lung cancer cells by regulating the G1/S-phase transition of the cell cycle. These results show that FBXW7-mediated ERK3 destabilization suppresses lung cancer cell proliferation in vitro.


Subject(s)
Lung Neoplasms , Mitogen-Activated Protein Kinase 6 , Animals , Cell Proliferation , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Lung Neoplasms/genetics , Mammals/metabolism , Mitogen-Activated Protein Kinase 6/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
12.
Int J Biol Sci ; 18(1): 112-123, 2022.
Article in English | MEDLINE | ID: mdl-34975321

ABSTRACT

Persistent infection with high-risk human papillomavirus (HPV) is the main risk factor for cervical cancer. Our mass spectrometry data showed that the Ras-associated binding protein Rab31 was upregulated by HPV; however, little is known regarding the role of Rab31 in the metastasis of cervical cancer cells. In this study, we showed that Rab31 was highly expressed in cervical cancer tissues and cells, and both HPV E6 and E7 promoted the expression of Rab31. Rab31 knockdown inhibited while Rab31 overexpression promoted the migration and invasion capabilities of cervical cancer cells. Additionally, Rab31 knockdown inhibited the epithelial-mesenchymal transition (EMT) and cytoskeletal rearrangement in cervical cancer cells. Furthermore, Rab31 interacted with mitogen-activated protein kinase 6 (MAPK6), and Rab31 knockdown inhibited the expression of MAPK6, which was mainly localized in the cytoplasm. More importantly, Rab31 knockdown promoted and Rab31 overexpression inhibited MAPK6 degradation. Accordingly, MAPK6 overexpression restored the decreased migration potential caused by Rab31 knockdown. Finally, a xenograft mouse model showed that Rab31 knockdown in cervical cancer cells led to reduced tumor growth and impaired lung and liver metastasis in vivo. In conclusion, Rab31 plays a crucial role in cervical cancer metastasis by inhibiting MAPK6 degradation. Thus, Rab31 may serve as a novel therapeutic target to manage cervical cancer.


Subject(s)
Mitogen-Activated Protein Kinase 6/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , rab GTP-Binding Proteins/metabolism , Animals , Cell Movement , Disease Progression , Female , HaCaT Cells , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Up-Regulation , Xenograft Model Antitumor Assays
13.
Biol Pharm Bull ; 44(11): 1662-1669, 2021.
Article in English | MEDLINE | ID: mdl-34719643

ABSTRACT

Glutamate differentially affects the levels extracellular signal-regulated kinase (ERK)1/2 and ERK3 and the protective effect of B355252, an aryl thiophene compound, 4-chloro-N-(naphthalen-1-ylmethyl)-5-(3-(piperazin-1-yl)phenoxy)thiophene-2-sulfonamide, is associated with suppression of ERK1/2. The objectives of this study were to further investigate the impact of B355252 on ERK3 and its downstream signaling pathways affected by glutamate exposure in the mouse hippocampal HT-22 neuronal cells. Murine hippocampal HT22 cells were incubated with glutamate and treated with B355252. Cell viability was assessed, protein levels of pERK3, ERK3, mitogen-activated protein kinase-activated protein kinase-5 (MAPKAPK-5), steroid receptor coactivator 3 (SRC-3), p-S6 and S6 were measured using Western blotting, and immunoreactivity of p-S6 was determined by immunocytochemistry. The results reveal that glutamate markedly diminished the protein levels of p-ERK3 and its downstream targets MK-5 and SRC-3 and increased p-S6, an indicator for mechanistic target of rapamycin (mTOR) activation. Conversely, treatment with B355252 protected the cells from glutamate-induced damage and prevented the glutamate-caused declines of p-ERK3, MK-5 and SRC-3 and increase of p-S6. Our study demonstrates that one of the mechanisms that glutamate mediates its cytotoxicity is through suppression of ERK3 and that B355252 rescues the cells from glutamate toxicity by reverting ERK3 level.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/toxicity , Hippocampus/drug effects , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 6/metabolism , Neuroprotective Agents/pharmacology , Thiophenes/pharmacology , Animals , Blotting, Western , Cell Line , Dose-Response Relationship, Drug , Fluorescent Antibody Technique , Mice
14.
Nat Commun ; 12(1): 5948, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34642330

ABSTRACT

Skeletal muscle is a highly adaptable tissue and remodels in response to exercise training. Using short RNA sequencing, we determine the miRNA profile of skeletal muscle from healthy male volunteers before and after a 14-day aerobic exercise training regime. Among the exercise training-responsive miRNAs identified, miR-19b-3p was selected for further validation. Overexpression of miR-19b-3p in human skeletal muscle cells increases insulin signaling, glucose uptake, and maximal oxygen consumption, recapitulating the adaptive response to aerobic exercise training. Overexpression of miR-19b-3p in mouse flexor digitorum brevis muscle enhances contraction-induced glucose uptake, indicating that miR-19b-3p exerts control on exercise training-induced adaptations in skeletal muscle. Potential targets of miR-19b-3p that are reduced after aerobic exercise training include KIF13A, MAPK6, RNF11, and VPS37A. Amongst these, RNF11 silencing potentiates glucose uptake in human skeletal muscle cells. Collectively, we identify miR-19b-3p as an aerobic exercise training-induced miRNA that regulates skeletal muscle glucose metabolism.


Subject(s)
DNA-Binding Proteins/genetics , Exercise/physiology , Glucose/metabolism , MicroRNAs/genetics , Protein Processing, Post-Translational , Adult , Animals , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Energy Metabolism/genetics , Healthy Volunteers , Humans , Kinesins/genetics , Kinesins/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 6/genetics , Mitogen-Activated Protein Kinase 6/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Oxygen Consumption/genetics , Phosphorylation , Physical Conditioning, Animal , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
15.
J Integr Plant Biol ; 63(10): 1815-1842, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34270159

ABSTRACT

Mitogen-activated protein kinase kinase kinase (MAPKKK) are the first components of MAPK cascades, which play pivotal roles in signaling during plant development and physiological processes. The genome of rice encodes 75 MAPKKKs, of which 43 are Raf-like MAPKKKs. The functions and action modes of most of the Raf-like MAPKKKs, whether they function as bona fide MAPKKKs and which are their downstream MAPKKs, are largely unknown. Here, we identified the osmapkkk43 mutant, which conferred broad-spectrum resistance to Xanthomonas oryzae pv. oryzae (Xoo), the destructive bacterial pathogen of rice. Oryza sativa (Os)MAPKKK43 encoding a Raf-like MAPKKK was previously known as Increased Leaf Angle 1 (OsILA1). Genetic analysis indicated that OsILA1 functioned as a negative regulator and acted upstream of the OsMAPKK4-OsMAPK6 cascade in rice-Xoo interactions. Unlike classical MAPKKKs, OsILA1 mainly phosphorylated the threonine 34 site at the N-terminal domain of OsMAPKK4, which possibly influenced the stability of OsMAPKK4. The N-terminal domain of OsILA1 is required for its homodimer formation and its full phosphorylation capacity. Taken together, our findings reveal that OsILA1 acts as a negative regulator of the OsMAPKK4-OsMAPK6 cascade and is involved in rice-Xoo interactions.


Subject(s)
Disease Resistance , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Oryza/enzymology , Xanthomonas , Host-Pathogen Interactions , MAP Kinase Kinase 4/metabolism , Mitogen-Activated Protein Kinase 6/metabolism
16.
Plant J ; 107(4): 1084-1101, 2021 08.
Article in English | MEDLINE | ID: mdl-34101285

ABSTRACT

Bacterial leaf streak (BLS) is a major bacterial disease of rice. Utilization of host genetic resistance has become one of the most important strategies for controlling BLS. However, only a few resistance genes have been characterized. Previously, a recessive BLS resistance gene bls1 was roughly mapped on chromosome 6. Here, we further delineated bls1 to a 21 kb region spanning four genes. Genetic analysis confirmed that the gene encoding a mitogen-activated protein kinase (OsMAPK6) is the target of the allelic genes BLS1 and bls1. Overexpression of BLS1 weakened resistance to the specific Xanthomonas oryzae pv. oryzicola (Xoc) strain JZ-8, while low expression of bls1 increased resistance. However, both overexpression of BLS1 and low expression of bls1 could increase no-race-specific broad-spectrum resistance. These results indicate that BLS1 and bls1 negatively regulate race-specific resistance to Xoc strain JZ-8 but positively and negatively control broad-spectrum resistance, respectively. Subcellular localization demonstrated that OsMAPK6 was localized in the nucleus. RGA4, which is known to mediate resistance to Xoc, is the potential target of OsMAPK6. Overexpression of BLS1 and low expression of bls1 showed increase in salicylic acid and induced expression of defense-related genes, simultaneously increasing broad-spectrum resistance. Moreover, low expression of bls1 showed increase an in jasmonic acid and abscisic acid, in company with an increase in resistance to Xoc strain JZ-8. Collectively, our study provides new insights into the understanding of BLS resistance and facilitates the development of rice host-resistant cultivars.


Subject(s)
Mitogen-Activated Protein Kinase 6/genetics , Oryza/microbiology , Plant Diseases/microbiology , Plant Proteins/genetics , Abscisic Acid/metabolism , Chromosome Mapping , Cyclopentanes/metabolism , Disease Resistance , Gene Expression Regulation, Plant , Host-Pathogen Interactions/physiology , Mitogen-Activated Protein Kinase 6/metabolism , Mutation , Oryza/genetics , Oxylipins/metabolism , Phylogeny , Plant Diseases/genetics , Plant Proteins/metabolism , Plants, Genetically Modified , Xanthomonas/pathogenicity
17.
BMC Cancer ; 21(1): 155, 2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33579235

ABSTRACT

BACKGROUND: p63, a member of the p53 gene family, is an important regulator for epithelial tissue growth and development. ∆Np63α is the main isoform of p63 and highly expressed in Non-melanoma skin cancer (NMSC). Extracellular signal-regulated kinase 3 (ERK3) is an atypical mitogen-activated protein kinase (MAPK) whose biochemical features and cellular regulation are distinct from those of conventional MAPKs such as ERK1/2. While ERK3 has been shown to be upregulated in lung cancers and head and neck cancers, in which it promotes cancer cell migration and invasion, little is known about the implication of ERK3 in NMSCs. METHODS: Fluorescent immunohistochemistry was performed to evaluate the expression levels of ΔNp63α and ERK3 in normal and NMSC specimens. Dunnett's test was performed to compare mean fluorescence intensity (MFI, indicator of expression levels) of p63 or ERK3 between normal cutaneous samples and NMSC samples. A mixed effects (ANOVA) test was used to determine the correlation between ΔNp63α and ERK3 expression levels (MFI). The regulation of ERK3 by ΔNp63α was studied by qRT-PCR, Western blot and luciferase assay. The effect of ERK3 regulation by ΔNp63α on cell migration was measured by performing trans-well migration assay. RESULTS: The expression level of ∆Np63α is upregulated in NMSCs compared to normal tissue. ERK3 level is significantly upregulated in AK and SCC in comparison to normal tissue and there is a strong positive correlation between ∆Np63α and ERK3 expression in normal skin and skin specimens of patients with AK, SCC or BCC. Further, we found that ∆Np63α positively regulates ERK3 transcript and protein levels in A431 and HaCaT skin cells, underlying the upregulation of ERK3 expression and its positive correlation with ∆Np63α in NMSCs. Moreover, similar to the effect of ∆Np63α depletion, silencing ERK3 greatly enhanced A431 cell migration. Restoration of ERK3 expression under the condition of silencing ∆Np63α counteracted the increase in cell migration induced by the depletion of ∆Np63α. Mechanistically, ERK3 inhibits the phosphorylation of Rac1 G-protein and the formation of filopodia of A431 skin SCC cells. CONCLUSIONS: ERK3 is positively regulated by ∆Np63α and mediates the role of ∆Np63α in suppressing cell migration in NMSC.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic , Mitogen-Activated Protein Kinase 6/metabolism , Skin Neoplasms/pathology , Transcription Factors/metabolism , Transcriptional Activation , Tumor Suppressor Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Line , Cell Line, Tumor , Humans , Mitogen-Activated Protein Kinase 6/genetics , Phosphorylation , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , rac1 GTP-Binding Protein/genetics
18.
Cell Cycle ; 20(2): 179-193, 2021 01.
Article in English | MEDLINE | ID: mdl-33459113

ABSTRACT

The function of a new long non-coding RNA GAS6-AS2 in non-small cell lung cancer (NSCLC) is not fully understood. In this study, GAS6-AS2 was identified, and its roles as well as mechanisms in regulating proliferation of NSCLCs cells were investigated. qRT-PCR was used to analyze GAS6-AS2, miR-144-3p, and MAPK6 expression. Protein expression was detected by Western blotting. Cell Counting Kit-8 (CCK8) assay was used to examine the cell proliferation ability. The interaction between GAS6-AS2 and miR-144-3p was confirmed by dual-luciferase reporter assay and RNA pull down assay. A xenograft model was constructed to monitor the mice NSCLC tumor growth in vivo. GAS6-AS2 was up-regulated, while miR-144-3p was suppressed in NSCLC cells compared with normal lung cells. GAS6-AS2 suppression could inhibit the progression of NSCLC cells, and miR-144-3p could attenuate the effect. GAS6-AS2 could function as a competitive endogenous RNA (ceRNA) via direct sponging miR-144-3p-3p, which further regulating the expression of MAPK6. The knockdown of GAS6-AS2 could greatly suppress the tumor growth of NSCLC in vivo. GAS6-AS2 up-regulated MAPK6 by sponging miR-144-3p in NSCLC tissues and cells. Thus, GAS6-AS2 is an effective therapeutic target in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cell Proliferation/genetics , Intercellular Signaling Peptides and Proteins/genetics , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 6/metabolism , Adult , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Middle Aged , Mitogen-Activated Protein Kinase 6/genetics , RNA, Long Noncoding/genetics , Xenograft Model Antitumor Assays/methods
19.
Cell Tissue Res ; 384(1): 113-127, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33404840

ABSTRACT

Osteoarthritis (OA) is the most common joint disease with an unsatisfactory therapy outcome and characterized by the degradation of articular cartilage and synovial inflammation. Here, we isolated bone marrow mesenchymal stem cells (BMSCs) from rat's bone marrow and BMSC-derived exosome (BMSCs-Exo) from BMSCs successfully. MiR-135b was proved to be highly expressed in TGF-ß1-stimulated BMSC-derived exosomes (BMSCs-ExoTGF-ß1). Then, our results demonstrated that BMSCs-ExoTGF-ß1 reduced OA-induced upregulation of pro-inflammatory factors in rat's serum and damage in cartilage tissues, which was then reversed by miR-135b decreasing. Subsequently, we found that the OA-resulted M1 polarization of synovial macrophages (SMs) was repressed by BMSCs-ExoTGF-ß1, this effect of BMSCs-ExoTGF-ß1 was limited by miR-135b decreasing. We also proved that M2 polarization of SMs can be induced by miR-135b mimics. Furthermore, we found that the promotory effect of miR-135b and BMSCs-ExoTGF-ß1 on M2 SMs polarization was reversed by increasing of MAPK6. Overall, our data showed that BMSCs-ExoTGF-ß1 attenuated cartilage damage in OA rats through carrying highly expressed miR-135b. Mechanistically, miR-135b promoted M2 polarization of SMs through targeting MAPK6, thus improving cartilage damage. Our study provided a novel regulatory mechanism of BMSCs-Exo in OA development and revealed a new potential treatment target of OA.


Subject(s)
Macrophages/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 6/metabolism , Osteoarthritis/therapy , Transforming Growth Factor beta1/metabolism , Animals , Disease Models, Animal , Male , Mesenchymal Stem Cells/cytology , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Rats , Rats, Sprague-Dawley , Transfection
20.
Mol Med Rep ; 23(2)2021 02.
Article in English | MEDLINE | ID: mdl-33300070

ABSTRACT

Pneumonia accounts for ~1.3 million mortalities in children per year worldwide. MicroRNAs are implicated in several diseases, including cancer and pneumonia; however, the role of let7f­5p in pneumonia is not completely understood. In the present study, lipopolysaccharide (LPS) was used to establish an in vitro pneumonia model in A549 and WI­38 cells. The reverse transcription­quantitative PCR (RT­qPCR) and western blotting results demonstrated that let7f­5p expression levels were significantly decreased, whereas MAPK6 expression levels were significantly increased in the peripheral venous blood of patients with pneumonia and in LPS­induced A549 and WI­38 cells compared with healthy volunteers and control cells, respectively. Furthermore, the dual­luciferase reporter assay demonstrated that let7f­5p targeted the 3'­untranslated region of MAPK6. The ELISA and RT­qPCR results demonstrated that let7f­5p mimic ameliorated LPS­induced inflammatory injury in A549 and WI­38 cells, as demonstrated by decreased expression levels of proinflammatory cytokines, including TNF­α and IL­6. In addition, the Cell Counting Kit­8 assay results indicated that let7f­5p mimic ameliorated LPS­induced reductions in cell viability, and the western blotting results demonstrated that let7f­5p mimic reversed LPS­induced activation of the STAT3 signaling pathway. Notably, the aforementioned let7f­5p­mediated effects were reversed by MAPK6 overexpression. Collectively, the results of the present study suggested that let7f­5p inhibited inflammation by targeting MAPK6 in the in vitro pneumonia model, thus let7f­5p may serve as a potential novel therapeutic target for pneumonia.


Subject(s)
Lung Injury/metabolism , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 6/metabolism , Models, Biological , Pneumonia/metabolism , A549 Cells , Child , Child, Preschool , Female , Humans , Infant , Inflammation , Lung Injury/genetics , Lung Injury/pathology , Male , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 6/genetics , Pneumonia/genetics , Pneumonia/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...