Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Curr Issues Mol Biol ; 46(4): 3218-3235, 2024 Apr 07.
Article in English | MEDLINE | ID: mdl-38666932

ABSTRACT

The glycoprotein hormones LH, FSH, TSH and chorionic gonadotropin consist of a common α-subunit and a hormone-specific Ɵ-subunit. The α-subunit is expressed in the pituitary and the placental cells, and its expression is regulated by extracellular signal molecules. Much is known about the regulation of the α-subunit gene in the pituitary, but few studies have addressed the regulation of this gene in trophoblasts. The aim of this study was to characterize the molecular mechanism of stimulus-induced α-subunit gene transcription in JEG-3 cells, a cellular model for human trophoblasts, using chromatin-embedded reporter genes under the control of the α-subunit promoter. The results show that increasing the concentration of the second messengers cAMP or Ca2+, or expressing the catalytic subunit of cAMP-dependent protein kinase in the nucleus activated the α-subunit promoter. Similarly, the stimulation of p38 protein kinase activated the α-subunit promoter, linking α-subunit expression to stress response. The stimulation of a Gαq-coupled designer receptor activated the α-subunit promoter, involving the transcription factor CREB, linking α-subunit expression to hormonal stimulation and an increase in intracellular Ca2+. Deletion mutagenesis underscores the importance of a tandem cAMP response element within the glycoprotein hormone α-subunit promoter, which acts as a point of convergence for a multiple signaling pathway.

2.
FASEB J ; 36(1): e22103, 2022 01.
Article in English | MEDLINE | ID: mdl-34921695

ABSTRACT

Ubiquitination has been shown to provide an essential regulatory role in modulating the duration and amplitude of the signaling activity in angiogenesis. While successive enzymatic reactions mediated by three distinct types of enzymes commonly known as E1, E2, and E3 are required for ubiquitination, the role of E3s which govern the final step of ubiquitination has been extensively analyzed in angiogenesis. In contrast, the role of E2s, which determine the context and functional consequences of ubiquitination, remains largely unknown with respect to angiogenesis. To better elucidate the role of E2s in modulating endothelial behaviors during angiogenesis, we first systematically analyze the expression pattern of E2s in endothelial cells (ECs) using previously published scRNA-seq data and identify ubiquitin-conjugating enzyme variant 1 (UBE2V1), an unconventional E2 without innate catalytic activity, as one of the most abundantly expressed E2s in ECs. While ubiquitously expressed in diverse cell types, abrogation of UBE2V1Ā significantly impairs proliferation and viability of human umbilical vein endothelial cells (HUVECs) without affecting other cell types, suggesting that UBE2V1 is likely to possess nonredundant functions in ECs. Consistent with this idea, UBE2V1 appears to be critical for morphogenesis and migration of ECs during angiogenesis. Interestingly, we find that UBE2V1 is essential for fibroblast growth factor 2 (FGF2)-induced angiogenesis, but appears to have minor effects on vascular endothelial growth factor-A-induced angiogenesis in vitro as well as in vivo. Therefore, it seems that UBE2V1 could enable ECs to distinguish two related yet distinct angiogenic cues. Mechanistically, we show that UBE2V1 promotes ubiquitination of MEK kinase 1, a key mediator of FGF2Ā signaling, to enhance phosphorylation of extracellular signal-regulated kinase 1/2 in HUVECs. Taken together, our results illustrate the unique role of UBE2V1 as a key modulator for angiogenic behaviors in ECs.


Subject(s)
Cell Proliferation , Endothelium, Vascular/metabolism , Fibroblast Growth Factor 2/metabolism , MAP Kinase Signaling System , Transcription Factors/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 2/genetics , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , PC-3 Cells , Transcription Factors/genetics , Ubiquitin-Conjugating Enzymes/genetics
3.
Proc Natl Acad Sci U S A ; 117(35): 21308-21318, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817551

ABSTRACT

The MEKK1 protein is a pivotal kinase activator of responses to cellular stress. Activation of MEKK1 can trigger various responses, including mitogen-activated protein (MAP) kinases, NF-κB signaling, or cell migration. Notably, MEKK1 activity is triggered by microtubule-targeting chemotherapies, among other stressors. Here we show that MEKK1 contains a previously unidentified tumor overexpressed gene (TOG) domain. The MEKK1 TOG domain binds to tubulin heterodimers-a canonical function of TOG domains-but is unusual in that it appears alone rather than as part of a multi-TOG array, and has structural features distinct from previously characterized TOG domains. MEKK1 TOG demonstrates a clear preference for binding curved tubulin heterodimers, which exist in soluble tubulin and at sites of microtubule polymerization and depolymerization. Mutations disrupting tubulin binding decrease microtubule density at the leading edge of polarized cells, suggesting that tubulin binding may play a role in MEKK1 activity at the cellular periphery. We also show that MEKK1 mutations at the tubulin-binding interface of the TOG domain recur in patient-derived tumor sequences, suggesting selective enrichment of tumor cells with disrupted MEKK1-microtubule association. Together, these findings provide a direct link between the MEKK1 protein and tubulin, which is likely to be relevant to cancer cell migration and response to microtubule-modulating therapies.


Subject(s)
MAP Kinase Kinase Kinase 1/metabolism , Tubulin/metabolism , Humans , MAP Kinase Kinase Kinase 1/chemistry , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 1/ultrastructure , Neoplasms/genetics , Protein Domains
4.
Dev Growth Differ ; 60(6): 341-353, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29900546

ABSTRACT

Planarians have established a unique body pattern along the anterior-posterior (AP) axis, which consists of at least four distinct body regions arranged in an anterior to posterior sequence: head, prepharyngeal, pharyngeal (containing a pharynx), and tail regions, and possess high regenerative ability. How they reconstruct the regional continuity in a head-to-tail sequence after amputation still remains unknown. We use as a model planarian Dugesia japonica head regeneration from tail fragments, which involves dynamic rearrangement of the body regionality of preexisting tail tissues along the AP axis, and show here that RNA interference of the gene D.Ā japonica mek kinase 1 (Djmekk1) caused a significant anterior shift in the position of pharynx regeneration at the expense of the prepharyngeal region, while keeping the head region relatively constant in size, and accordingly led to development of a relatively longer tail region. Our data suggest that DjMEKK1 regulates anterior extracellular signal-regulated kinase (ERK) and posterior Ɵ-catenin signaling pathways in a positive and negative manner, respectively, to establish a proper balance resulting in the regeneration of planarian's scale-invariant trunk-to-tail patterns across individuals. Furthermore, we demonstrated that DjMEKK1 negatively modulates planarian Ɵ-catenin activity via its serine/threonine kinase domain, but not its PHD/RING finger domain, by testing secondary axis formation in Xenopus embryos. The data suggest that Djmekk1 plays an instructive role in the coordination between the establishment of the prepharyngeal region and posteriorizing of pharynx formation by balancing the two opposing morphogenetic signals along the AP axis during planarian regeneration.


Subject(s)
Helminth Proteins/metabolism , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Signaling System/physiology , Planarians/enzymology , Regeneration/physiology , Animals , Planarians/cytology
5.
Adv Exp Med Biol ; 1037: 97-131, 2017.
Article in English | MEDLINE | ID: mdl-29147906

ABSTRACT

The ability of DJ-1 to modulate signal transduction has significant effects on how the cell regulates normal processes such as growth, senescence, apoptosis, and autophagy to adapt to changing environmental stimuli and stresses. Perturbations of DJ-1 levels or function can disrupt the equilibrium of homeostatic signaling networks and set off cascades that play a role in the pathogenesis of conditions such as cancer and Parkinson's disease.DJ-1 plays a major role in various pathways. It mediates cell survival and proliferation by activating the extracellular signal-regulated kinase (ERK1/2) pathway and the phosphatidylinositol-3-kinase (PI3K)/Akt pathway. It attenuates cell death signaling by inhibiting apoptosis signal-regulating kinase 1 (ASK1) activation as well as by inhibiting mitogen-activated protein kinase kinase kinase 1 (MEKK1/MAP3K1) activation of downstream apoptotic cascades. It also modulates autophagy through the ERK, Akt, or the JNK/Beclin1 pathways. In addition, DJ-1 regulates the transcription of genes essential for male reproductive function, such as spermatogenesis, by relaying nuclear receptor androgen receptor (AR) signaling. In this chapter, we summarize the ways that DJ-1 regulates these pathways, focusing on how its role in signal transduction contributes to cellular homeostasis and the pathologic states that result from dysregulation.


Subject(s)
Neoplasms/metabolism , Parkinson Disease/metabolism , Protein Deglycase DJ-1/metabolism , Protein Interaction Maps , Signal Transduction , Animals , Humans , Models, Biological
6.
Int J Mol Sci ; 18(4)2017 Apr 19.
Article in English | MEDLINE | ID: mdl-28422048

ABSTRACT

SAK-HV is an anti-atherosclerosis recombinant fusion protein developed by our lab. Our study determined that SAK-HV promoted macrophage proliferation, of which the mechanism was explored by both RAW264.7 cells and primary macrophages. Mass spectrometric analysis and co-immunoprecipitation were combined to screen the SAK-HV-interacting proteins in RAW264.7 cells. Confocal microscopy was adopted to detect the localization of SAK-HV in cells. The results indicated that SAK-HV triggered macrophage proliferation via the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinases (ERK) and c-Jun N-terminal kinases (JNK) pathways by its SAK-mutant functional domain. We screened out Uba1 as the SAK-HV-interacting protein in the RAW264.7 cells and discovered their co-localization in the cytoplasm and nucleus. Inhibiting Uba1 significantly decreased the SAK-HV-induced macrophage proliferation. Thus, we postulated an attractive model of ubiquitination, in which the interactions between Uba1 and specific E2 enzymes are blocked by its interaction with SAK-HV. Based on this model, we detected the decreased self-ubiquitination of MEKK1 after SAK-HV treatment and concluded that SAK-HV inhibits the self-ubiquitination of MEKK1 via its SAK-mutant functional domain to activate MAPK/ERK and JNK pathways, promoting macrophage proliferation. This conclusion highly supported our hypothesized model of ubiquitination at the level of Uba1, which may represent a novel paradigm to promote macrophage proliferation by using the E1 enzyme (Uba1) as a switch.


Subject(s)
MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Macrophages/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Mice , Mutation , Phosphorylation , Protein Interaction Domains and Motifs/genetics , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Ubiquitination/drug effects
7.
Neurochem Res ; 41(12): 3308-3321, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27662850

ABSTRACT

The JNKs have been implicated in a variety of biological functions in mammalian cells, including apoptosis and the responses to stress. However, the physiological role of these pathways in the intracerebral hemorrhage (ICH) has not been fully elucidated. In this study, we identified a MAPK kinase kinase (MAPKKK), MEKK1, may be involved in neuronal apoptosis in the processes of ICH through the activation of JNKs. From the results of western blot, immunohistochemistry and immunofluorescence, we obtained a significant up-regulation of MEKK1 in neurons adjacent to the hematoma following ICH. Increasing MEKK1 level was found to be accompanied with the up-regulation of p-JNK 3, p53, and c-jun. Besides, MEKK1 co-localized well with p-JNK in neurons, indicating its potential role in neuronal apoptosis. What's more, our in vitro study, using MEKK1 siRNA interference in PC12 cells, further confirmed that MEKK1 might exert its pro-apoptotic function on neuronal apoptosis through extrinsic pathway. Thus, MEKK1 may play a role in promoting the brain damage following ICH.


Subject(s)
Apoptosis , Basal Ganglia/enzymology , Cerebral Hemorrhage/enzymology , MAP Kinase Kinase Kinase 1/metabolism , Neurons/enzymology , Animals , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/physiopathology , Male , Neurons/pathology , Rats, Sprague-Dawley
8.
Br J Nutr ; 115(9): 1547-55, 2016 05.
Article in English | MEDLINE | ID: mdl-26948765

ABSTRACT

The c-Jun N-terminal kinases (JNK) are members of the mitogen-activated protein kinase family and are activated by environmental stress. Se plays an important role in the biological pathways by forming selenoprotein. Selenoproteins have been shown to exhibit a variety of biological functions including antioxidant functions and maintaining cellular redox balance, and compromise of such important proteins would lead to oxidative stress and apoptosis. We examined the expression levels of JNK in Kashin-Beck disease (KBD) patients, tested the potential protective effects of sodium selenite on tert-butyl hydroperoxide (tBHP)-induced oxidative injury and apoptosis in human chondrocytes as well as its underlying mechanism in this study. We produced an oxidative damage model induced by tBHP in C28/I2 human chondrocytes to test the essential anti-apoptosis effects of Se in vitro. The results indicated that the expression level of phosphorylated JNK was significantly increased in KBD patients. Cell apoptosis was increased and molecule expressions of the JNK signalling pathway were activated in the tBHP-injured chondrocytes. Na2SeO3 protected against tBHP-induced oxidative stress and apoptosis in cells by increasing cell viability, reducing reactive oxygen species generation, increasing Glutathione peroxidase (GPx) activity and down-regulating the JNK pathway. These results demonstrate that apoptosis induced by tBHP in chondrocytes might be mediated via up-regulation of the JNK pathway; Na2SeO3 has an effect of anti-apoptosis by down-regulating the JNK signalling pathway.


Subject(s)
Chondrocytes/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Kashin-Beck Disease/metabolism , MAP Kinase Signaling System , Osteoarthritis/metabolism , Oxidative Stress/drug effects , Sodium Selenite/pharmacology , Antioxidants/metabolism , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Chondrocytes/metabolism , Down-Regulation , Glutathione Peroxidase/metabolism , Humans , Phosphorylation , Reactive Oxygen Species/metabolism , Selenium/pharmacology , Signal Transduction , Up-Regulation , tert-Butylhydroperoxide
9.
J Cell Biochem ; 116(12): 2903-14, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26018553

ABSTRACT

Previously, we showed that Mekk1 translocates to the nucleus, interacts with tumor suppressor protein p53, and co-represses PKD1 transcription via an atypical p53 binding site on the minimal PKD1 promoter (JBC 285:38,818-38,831, 2010). In this study, we report the mechanisms of Mekk1 nuclear transport and p53 binding. Using GFP-linked constitutively active-Mekk1 (CA-Mekk1) and a deletion strategy, we identified a nuclear localization signal (HRDVK) located at amino acid (aa) residues 1,349-1,353 in the C-terminal Mekk1 catalytic domain. Deletion of this sequence in CA-Mekk1 and full-length Mekk1 significantly reduced their nuclear translocation in both HEK293T and COS-1 cells. Using co-immunoprecipitation, we identified an adjacent sequence (GANLID, aa 1,354-1,360) in Mekk1 responsible for p53 binding. Deletion of this sequence markedly reduced the interaction of Mekk1 with p53. Mekk1 does not appear to affect phosphorylation of Ser15, located in the Mdm2 interaction site, or other Ser residues in p53. However, Mekk1 mediates p53 protein stability in the presence of Mdm2 and reduces p53 ubiquitination, suggesting an interference with Mdm2-mediated degradation of p53 by the ubiquitin-proteasome pathway.


Subject(s)
MAP Kinase Kinase Kinase 1/metabolism , Nuclear Localization Signals/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , TRPP Cation Channels/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , MAP Kinase Kinase Kinase 1/genetics , Nuclear Localization Signals/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Stability , Proto-Oncogene Proteins c-mdm2/genetics , TRPP Cation Channels/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitination/genetics
10.
J Exp Bot ; 65(3): 779-87, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24151303

ABSTRACT

As a signalling molecule, glutamate is best known for its role as a fast excitatory neurotransmitter in the mammalian nervous system, a role that requires the activity of a family of ionotropic glutamate receptors (iGluRs). The unexpected discovery in 1998 that Arabidopsis thaliana L. possesses a family of iGluR-related (GLR) genes laid the foundations for an assessment of glutamate's potential role as a signalling molecule in plants that is still in progress. Recent advances in elucidating the function of Arabidopsis GLR receptors has revealed similarities with iGluRs in their channel properties, but marked differences in their ligand specificities. The ability of plant GLR receptors to act as amino-acid-gated Ca(2+) channels with a broad agonist profile, combined with their expression throughout the plant, makes them strong candidates for a multiplicity of amino acid signalling roles. Although root growth is inhibited in the presence of a number of amino acids, only glutamate elicits a specific sequence of changes in growth, root tip morphology, and root branching. The recent finding that the MEKK1 gene is a positive regulator of glutamate sensitivity at the root tip has provided genetic evidence for the existence in plants of a glutamate signalling pathway analogous to those found in animals. This short review will discuss the most recent advances in understanding glutamate signalling in roots, considering them in the context of previous work in plants and animals.


Subject(s)
Arabidopsis/physiology , Glutamic Acid/metabolism , Plant Roots/physiology , Receptors, Glutamate/metabolism , Signal Transduction , Amino Acids/metabolism , Arabidopsis/genetics , Arabidopsis/growth & development , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Plant Roots/genetics , Plant Roots/growth & development , Receptors, Glutamate/genetics
11.
Appl Biochem Biotechnol ; 196(1): 1-17, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37097403

ABSTRACT

Asperulosidic acid (ASPA) is a plant-extracted iridoid terpenoid with tumor-suppressive and anti-inflammatory properties. At present, the antitumor function of ASPA and its related mechanisms in hepatocellular carcinoma (HCC) cells were explored. Human normal hepatocytes HL-7702 and HCC cells (Huh7 and HCCLM3) were treated with varying concentrations (0 to 200Ā Āµg/mL) of ASPA. Cell viability, proliferation, apoptosis, migration, and invasion were checked. The expression of proteins was detected by Western blot. Furthermore, the effect of ASPA (100Ā Āµg/mL) on the sensitivity of HCC cells to chemotherapeutic agents, including doxorubicin and cisplatin, was evaluated. A subcutaneous xenografted tumor model was set up in nude mice, and the antitumor effects of ASPA were evaluated. ASPA hindered HCC cells' proliferation, migration, and invasion, and amplified their apoptosis and sensitivity to chemotherapeutic agents. Additionally, ASPA inactivated the MEKK1/NF-κB pathway. Overexpression of MEKK1 increased HCC proliferation, migration, and invasion and facilitated chemoresistance. ASPA treatment alleviated the carcinogenic effect mediated by MEKK1 overexpression. MEKK1 knockdown slowed down HCC progression. However, ASPA could not exert additional antitumor effects in MEKK1 knockdown cells. In vivo results displayed that ASPA substantially curbed tumor growth and inactivated the MEKK1/NF-κB pathway in mice. All over, ASPA exerts antitumor effects in HCC by suppressing the MEKK1/NF-κB pathway.


Subject(s)
Carcinoma, Hepatocellular , Glycosides , Liver Neoplasms , Humans , Animals , Mice , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , NF-kappa B/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Mice, Nude , Cell Line, Tumor , Cell Proliferation , Apoptosis , Gene Expression Regulation, Neoplastic
12.
Cell Mol Gastroenterol Hepatol ; 15(2): 307-325, 2023.
Article in English | MEDLINE | ID: mdl-36244648

ABSTRACT

BACKGROUND & AIMS: The contribution of abnormal metabolic targets to hepatocellular carcinoma (HCC) progression and the associated regulatory mechanisms are attractive research areas. High-density lipoprotein binding protein (HDLBP) is an important transporter that protects cells from excessive cholesterol accumulation, but few studies have identified a role for HDLBP in HCC progression. METHODS: HDLBP expression was determined in HCC tissues and published datasets. The biological roles of HDLBP inĀ vitro and inĀ vivo were examined by performing a series of functional experiments. RESULTS: An integrated analysis confirmed that HDLBP expression was significantly elevated in HCC compared with noncancerous liver tissues. The knockdown or overexpression of HDLBP substantially inhibited or enhanced, respectively, HCC proliferation and sorafenib resistance. Subsequently, a mass spectrometry screen identified RAF1 as a potential downstream target of HDLBP. Mechanistically, when RAF1 was stabilized by HDLBP, MEKK1 continuously induced RAF1Ser259-dependent MAPK signaling. Meanwhile, HDLBP interacted with RAF1 by competing with the TRIM71 E3 ligase and inhibited RAF1 degradation through the ubiquitin-proteasome pathway. CONCLUSIONS: Our study reveals that HDLBP is an important mediator that stabilizes the RAF1 protein and maintains its activity, leading to HCC progression and sorafenib resistance. Thus, HDLBP might represent a potential biomarker and future therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Proto-Oncogene Proteins c-raf , Tripartite Motif Proteins , Humans , Biomarkers , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Cell Proliferation , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Sorafenib/pharmacology , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases , Proto-Oncogene Proteins c-raf/genetics , RNA-Binding Proteins/genetics
13.
Eur J Pharmacol ; 951: 175748, 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37149277

ABSTRACT

Resveratrol (RES), a natural polyphenolic compound found in red wine and grape skins, has attracted significant attention due to its cardioprotective properties. DJ-1, a multifunctional protein that participated in transcription regulation and antioxidant defense, was shown to provide a significant protective impact in cardiac cells treated with ischemia-reperfusion. We created a myocardial ischemia-reperfusion (I/R) model in vivo and in vitro by ligating the left anterior descending branch of rats and subjecting H9c2 cells to anoxia/reoxygenation (A/R) to investigate whether RES reduces myocardial ischemia-reperfusion injury by upregulating DJ-1. We discovered that RES dramatically enhanced cardiac function in rats with I/R. Subsequently, we found that RES prevented the rise in autophagy (P62 degradation and LC3-II/LC3-I increase) induced by cardiac ischemia-reperfusion in vitro and in vivo. Notably, the autophagic agonist rapamycin (RAPA) eliminated RES-induced cardioprotective effects. In addition, Further data showed that RES significantly increased the expression of DJ-1 in the myocardium with the treatment of I/R. At the same time, pretreatment with RES reduced phosphorylation of MAPK/ERK kinase kinase 1 (MEKK1) and Jun N-terminal Kinase (JNK) stimulated by cardiac ischemia-reperfusion, and Beclin-1 mRNA and protein levels while decreasing lactate dehydrogenase (LDH) and improving cell viability. However, the lentiviral shDJ-1 and JNK agonist anisomycin disrupted the effects of RES. In summary, RES could inhibit autophagy against myocardial ischemia-reperfusion injury through DJ-1 modulation of the MEKK1/JNK pathway, providing a novel therapeutic strategy for cardiac homeostasis.


Subject(s)
Myocardial Ischemia , Myocardial Reperfusion Injury , Rats , Animals , Myocardial Reperfusion Injury/metabolism , Resveratrol/therapeutic use , MAP Kinase Signaling System , MAP Kinase Kinase Kinases/metabolism , Autophagy , Myocytes, Cardiac , Apoptosis
14.
Inflammation ; 45(5): 2016-2026, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35460012

ABSTRACT

Berberine (BBR), a natural isoquinoline alkaloid exhibiting insulin sensitizing activity, has been applicated in the treatment of diabetes. However, until now, the exact target of BBR has not been well investigated. Here, primary hepatocytes pre-treated with TNF-α were used to evaluate the role of BBR on hepatic insulin sensitivity. Western blot and immunoprecipitation were used to investigate the effect of BBR on the crosstalk between TNF-α pathway and insulin signaling pathway. Molecular docking was used to verify the interactions between BBR and its potential targets. BBR inhibits the MEKK1 and MEK1/2, and thus suppresses the activation of their downstream ERK1/2. It attenuates the ERK1/2-induced serine phosphorylation of IRS-1 and thus enhances IRS-1 tyrosine phosphorylation and Akt activation. By molecular docking, BBR is proved to efficiently bind MEK1/2. MEKK1 is also considered as BBR target for its similarity in primary structure with MEK1/2. In conclusion, BBR ameliorates TNF-α-induced hepatic insulin resistance by targeting MEKK1 and MEK1/2.


Subject(s)
Berberine , Insulin Resistance , Berberine/pharmacology , Humans , Insulin/metabolism , Insulin Resistance/physiology , Isoquinolines , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Docking Simulation , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Serine/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tyrosine
15.
J Exp Clin Cancer Res ; 40(1): 200, 2021 Jun 21.
Article in English | MEDLINE | ID: mdl-34154626

ABSTRACT

BACKGROUND: MEK1/ERK signaling pathway plays an important role in most tumor progression, including colorectal cancer (CRC), however, MEK1-targeting therapy has little effective in treating CRC patients, indicating there may be a complex mechanism to activate MEK1/ERK signaling pathway except RAS activated mechanism. METHODS: To investigate the clinical significance of IMP3, we analyzed its expression levels in publicly available dataset and samples from Fudan University Shanghai Cancer Center. The effects of IMP3 on proliferation, migration, and invasion were determined by in vitro and in vivo experiments. To investigate the role of IMP3 in colon carcinogenesis, conditional IMP3 knockout C57BL/6 mice was generated. The IMP3/MEKK1/MEK/ERK signaling axis in CRC was screened and validated by RNA-sequencing, RNA immunoprecipitation, luciferase reporter and western blot assays. RESULTS: We find RNA binding protein IMP3 directly bind to MEKK1 mRNA 3'-UTR, which regulates its stability, promote MEKK1 expression and sequentially activates MEK1/ERK signaling. Functionally, IMP3 promote the malignant biological process of CRC cells via MEKK1/MEK1/ERK signaling pathway both in vitro and in vivo, Moreover, IMP3-/- mice show decreased the expression of MEKK1 as well as colorectal tumors compared with wild-type mice after treatment with azoxymethane/dextran sodium sulfate. Clinically, the expression of IMP3 and MEKK1 are positive correlated, and concomitant IMP3 and MEKK1 protein levels negatively correlate with metastasis in CRC patients. In addition, MEK1 inhibitor in combination with shRNA-IMP3 have a synergistic effect both in vitro and in vivo. CONCLUSION: Our study demonstrates that IMP3 regulates MEKK1 in CRC, thus activating the MEK1/ERK signaling in the progression of colorectal cancer, Furthermore, these results provide new insights into potential applications for combining MEK1 inhibitors with other target therapy such as IMP3 in preclinical trials for CRC patients.


Subject(s)
Colorectal Neoplasms/metabolism , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Signaling System , RNA, Messenger/metabolism , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Progression , Humans , Male , Mice , RNA, Messenger/genetics , Ribonucleoproteins, Small Nucleolar/genetics , Ribonucleoproteins, Small Nucleolar/metabolism
16.
Front Oncol ; 11: 609918, 2021.
Article in English | MEDLINE | ID: mdl-33868996

ABSTRACT

Breast tumors contain both transformed epithelial cells and non-transformed stroma cells producing secreted factors that can promote metastasis. Previously, we demonstrated that the kinase MEKK1 regulates cell migration and gene expression, and that transgene-induced breast tumor metastasis is markedly inhibited in MEKK1-deficient mice. In this report, we examined the role of MEKK1 in stroma cell gene expression and the consequent effect on breast tumor cell function. Using a heterotypic cell system to quantify the effect of stroma cells on breast tumor cell function, we discovered that MEKK1-/- fibroblasts are significantly less effective at inducing tumor cell invasion than MEKK1+/+ fibroblasts. Expression array analysis revealed that both baseline and tumor cell-induced expression of the chemokines CCL3, CCL4, and CCL5 were markedly reduced in MEKK1-/- mammary fibroblasts. By focusing on the role of MEKK1 in CCL5 regulation, we discovered that MEKK1 kinase activity promotes CCL5 expression, and inactive mutant MEKK1 strongly inhibits CCL5 transcription. CCL5 and the other MEKK1-dependent chemokines are ligands for the GPCR CCR5, and we show that the CCR5 antagonist Maraviroc strongly inhibits fibroblast-induced tumor cell migration. Finally, we report that fibroblast growth factor 5 (FGF-5) is secreted by MDA-MB 231 cells, that FGF-5 activates MEKK1 effectors ERK1/2 and NFκB in fibroblasts, and that chemical inhibition of NFκB inhibits CCL5 expression. Our results suggest that MEKK1 contributes to the formation of a breast tumor microenvironment that supports metastasis by promoting expression of stroma cell chemokine genes in response to tumor cell-induced paracrine signaling.

17.
Mol Cell Biol ; 41(10): e0008121, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34251884

ABSTRACT

Cullin-4 ubiquitin ligase (CRL4) complexes are differentially composed and highly dynamic protein assemblies that control many biological processes, including the global genome nucleotide excision repair (GG-NER) pathway. Here, we identified the kinase mitogen-activated protein kinase kinase kinase 1 (MEKK1) as a novel constitutive interactor of a cytosolic CRL4 complex that disassembles after DNA damage due to the caspase-mediated cleavage of MEKK1. The kinase activity of MEKK1 was important to trigger autoubiquitination of the CRL4 complex by K48- and K63-linked ubiquitin chains. MEKK1 knockdown prohibited DNA damage-induced degradation of the CRL4 component DNA-damage binding protein 2 (DDB2) and the CRL4 substrate p21 and also cell recovery and survival. A ubiquitin replacement strategy revealed a contribution of K63-branched ubiquitin chains for DNA damage-induced DDB2/p21 decay, cell cycle regulation, and cell survival. These data might also have implications for cancer, as frequently occurring mutations of MEKK1 might have an impact on genome stability and the therapeutic efficacy of CRL4-dependent immunomodulatory drugs such as thalidomide derivatives.


Subject(s)
DNA Repair/physiology , MAP Kinase Kinase Kinase 1/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Survival , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA/chemistry , DNA Damage/physiology , DNA Repair/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , MAP Kinase Kinase Kinase 1/genetics , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/physiology , Ubiquitination
18.
FEBS Lett ; 592(19): 3327-3334, 2018 10.
Article in English | MEDLINE | ID: mdl-30193004

ABSTRACT

The MEKK1 kinase is a key regulator of stress signaling in Arabidopsis; however, little is known about the regulation of its kinase activity. Here, we found that recombinant MEKK1, expressed in both mammalian HEK293 cells and Escherichia coli, shows a mobility shift in SDS-PAGE, and immunoblotting detected phosphorylation of serine, threonine, and tyrosine residues. N-terminal deletions, site-directed mutagenesis, and protein phosphatase treatment revealed that the mobility shift results from autophosphorylation of the kinase domain. We identified the tyrosine autophosphorylation sites in the N-terminal region of MEKK1. Tyrosine to phenylalanine mutations decrease phosphorylation of the substrate MKK1, suggesting the important role of this residue in the regulation of MEKK1 kinase activity. The present study is the first to show that plant MAPKKKs are regulated by tyrosine phosphorylation.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , MAP Kinase Kinase Kinases/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Binding Sites/genetics , Escherichia coli/genetics , HEK293 Cells , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinases/genetics , Mutation, Missense , Phenylalanine/genetics , Phenylalanine/metabolism , Phosphorylation , Recombinant Proteins/metabolism , Substrate Specificity , Tyrosine/genetics
19.
Biomed Pharmacother ; 102: 1111-1119, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29710529

ABSTRACT

OBJECTIVE: To determine the role of the MEKK1/SEK1/JNK1/AP-1 pathway in the action of Xihuang pill (XHP) in reducing regulatory T (Treg) cell numbers in the tumor microenvironment in a 4T1 mouse breast cancer model, and to clarify the anti-tumor mechanism of XHP in breast cancer. METHODS: We established a mouse 4T1 breast cancer model. Model mice were administered XHP for 2 weeks, and tumor tissues were then removed, weighed, sliced, and homogenized. Treg cells in the tumor microenvironment were isolated by magnetic cell sorting and analyzed by immunohistochemistry and flow cytometry. Treg cell apoptosis was detected by TdT-mediated dUTP nick end labeling. mRNA expression levels of MEKK1, SEK1, JNK1, and AP-1 in Treg cells in the tumor microenvironment were detected by quantitative real-time PCR and their protein expression levels were detected by immunofluorescence staining and western blot. RESULTS: Tumor weights were significantly lower in the XHP groups compared with the untreated control group. The overall number of Treg cells in the tumor microenvironment decreased while the number of apoptotic Treg cells increased with increasing doses of XHP. mRNA and protein expression levels of MEKK1, SEK1, JNK1, and AP-1 in Treg cells in the tumor microenvironment increased with increasing doses of XHP. CONCLUSION: XHP might promote Treg cell apoptosis in the tumor microenvironment and further inhibit the tumor growth of 4T1 mouse breast cancer. The mechanism of XHP may be related to upregulation of gene and protein expression of MEKK1, SEK1, JNK1, and AP-1 in Treg cells in the tumor microenvironment.


Subject(s)
Apoptosis , Drugs, Chinese Herbal/therapeutic use , MAP Kinase Signaling System , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/pathology , T-Lymphocytes, Regulatory/pathology , Tumor Microenvironment , Up-Regulation , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Separation , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Immunomagnetic Separation , Lymphocyte Count , MAP Kinase Signaling System/drug effects , Mammary Neoplasms, Animal/enzymology , Mammary Neoplasms, Animal/immunology , Mice, Inbred BALB C , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes, Regulatory/drug effects , Transcription Factor AP-1/metabolism , Tumor Burden/drug effects , Tumor Microenvironment/drug effects , Up-Regulation/drug effects
20.
Am J Transl Res ; 9(8): 3714-3722, 2017.
Article in English | MEDLINE | ID: mdl-28861162

ABSTRACT

MicroRNAs (miRNAs) play critical roles in the tumorigenesis of prostate cancer, while the biological function of miR-4735-3p is unknown. Mitogen-activated protein kinase kinase kinase 1 (MEKK1) has been shown to induce androgen receptor (AR)-dependent apoptosis in prostate cancer cells, but the regulation of MEKK1 in prostate cancer cells remains poorly defined. Here, we showed that miR-4735-3p was a MEKK1-targeting miRNA, and was highly expressed in AR+ prostate cancer specimens. Moreover, the levels of miR-4735-3p and MEKK1 inversely correlated. MiR-4735-3p-low subjects had a better overall survival, compared to miR-4735-3p-high subjects. MiR-4735-3p targeted the 3'-UTR of MEKK1 mRNA to inhibit its protein translation. Overexpression of miR-4735-3p inhibited MEKK1-mediated cell apoptosis upon docetaxel treatment, while depletion of miR-4735-3p enhanced it. Together, our data suggest that miR-4735-3p may suppress MEKK1-mediated prostate cancer cell apoptosis during chemotherapy. Inhibition of miR-4735-3p may improve the outcome of chemotherapy for some prostate cancers.

SELECTION OF CITATIONS
SEARCH DETAIL