Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 199
Filtrar
1.
Genes Dev ; 34(7-8): 495-510, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32139423

RESUMO

Obesity-induced diabetes affects >400 million people worldwide. Uncontrolled lipolysis (free fatty acid release from adipocytes) can contribute to diabetes and obesity. To identify future therapeutic avenues targeting this pathway, we performed a high-throughput screen and identified the extracellular-regulated kinase 3 (ERK3) as a hit. We demonstrated that ß-adrenergic stimulation stabilizes ERK3, leading to the formation of a complex with the cofactor MAP kinase-activated protein kinase 5 (MK5), thereby driving lipolysis. Mechanistically, we identified a downstream target of the ERK3/MK5 pathway, the transcription factor FOXO1, which promotes the expression of the major lipolytic enzyme ATGL. Finally, we provide evidence that targeted deletion of ERK3 in mouse adipocytes inhibits lipolysis, but elevates energy dissipation, promoting lean phenotype and ameliorating diabetes. Thus, ERK3/MK5 represents a previously unrecognized signaling axis in adipose tissue and an attractive target for future therapies aiming to combat obesity-induced diabetes.


Assuntos
Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatologia , Metabolismo Energético/genética , Lipólise/genética , Proteína Quinase 6 Ativada por Mitógeno/genética , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Obesidade/complicações , Células 3T3 , Tecido Adiposo/enzimologia , Animais , Diabetes Mellitus Tipo 2/tratamento farmacológico , Avaliação Pré-Clínica de Medicamentos , Proteína Forkhead Box O1/metabolismo , Deleção de Genes , Células HEK293 , Humanos , Hipoglicemiantes/uso terapêutico , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipase/genética , Lipase/metabolismo , Camundongos , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/genética
2.
Eur Spine J ; 33(5): 2056-2067, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38551688

RESUMO

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.


Assuntos
Apoptose , MicroRNAs , Neurônios , RNA Longo não Codificante , Traumatismos da Medula Espinal , Animais , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Apoptose/genética , Apoptose/fisiologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Camundongos , Neurônios/metabolismo , Proteína Quinase 6 Ativada por Mitógeno/genética , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Medula Espinal/metabolismo , Medula Espinal/patologia , Inflamação/genética , Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Masculino
3.
J Transl Med ; 21(1): 186, 2023 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-36895010

RESUMO

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.


Assuntos
Neoplasias da Mama , MicroRNAs , Fatores Associados à Proteína de Ligação a TATA , Neoplasias de Mama Triplo Negativas , Feminino , Humanos , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Hibridização in Situ Fluorescente , MicroRNAs/genética , RNA Circular/genética , Transdução de Sinais/genética , Fatores Associados à Proteína de Ligação a TATA/genética , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Proteína Quinase 6 Ativada por Mitógeno/metabolismo
4.
Proc Natl Acad Sci U S A ; 117(8): 4078-4087, 2020 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-32041890

RESUMO

Anthrax lethal toxin (LT) is a protease virulence factor produced by Bacillus anthracis that is required for its pathogenicity. LT treatment causes a rapid degradation of c-Jun protein that follows inactivation of the MEK1/2-Erk1/2 signaling pathway. Here we identify COP1 as the ubiquitin E3 ligase that is essential for LT-induced c-Jun degradation. COP1 knockdown using siRNA prevents degradation of c-Jun, ETV4, and ETV5 in cells treated with either LT or the MEK1/2 inhibitor, U0126. Immunofluorescence staining reveals that COP1 preferentially localizes to the nuclear envelope, but it is released from the nuclear envelope into the nucleoplasm following Erk1/2 inactivation. At baseline, COP1 attaches to the nuclear envelope via interaction with translocated promoter region (TPR), a component of the nuclear pore complex. Disruption of this COP1-TPR interaction, through Erk1/2 inactivation or TPR knockdown, leads to rapid COP1 release from the nuclear envelope into the nucleoplasm where it degrades COP1 substrates. COP1-mediated degradation of c-Jun protein, combined with LT-mediated blockade of the JNK1/2 signaling pathway, inhibits cellular proliferation. This effect on proliferation is reversed by COP1 knockdown and ectopic expression of an LT-resistant MKK7-4 fusion protein. Taken together, this study reveals that the nuclear envelope acts as a reservoir, maintaining COP1 poised for action. Upon Erk1/2 inactivation, COP1 is rapidly released from the nuclear envelope, promoting the degradation of its nuclear substrates, including c-Jun, a critical transcription factor that promotes cellular proliferation. This regulation allows mammalian cells to respond rapidly to changes in extracellular cues and mediates pathogenic mechanisms in disease states.


Assuntos
Antígenos de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Humanos , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 6 Ativada por Mitógeno/genética , Proteínas Nucleares/genética , Ubiquitina-Proteína Ligases/genética
5.
Plant J ; 107(4): 1084-1101, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34101285

RESUMO

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.


Assuntos
Proteína Quinase 6 Ativada por Mitógeno/genética , Oryza/microbiologia , Doenças das Plantas/microbiologia , Proteínas de Plantas/genética , Ácido Abscísico/metabolismo , Mapeamento Cromossômico , Ciclopentanos/metabolismo , Resistência à Doença , Regulação da Expressão Gênica de Plantas , Interações Hospedeiro-Patógeno/fisiologia , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Mutação , Oryza/genética , Oxilipinas/metabolismo , Filogenia , Doenças das Plantas/genética , Proteínas de Plantas/metabolismo , Plantas Geneticamente Modificadas , Xanthomonas/patogenicidade
6.
J Cell Physiol ; 237(4): 2271-2287, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35141958

RESUMO

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.


Assuntos
Proteína Forkhead Box O3/metabolismo , Transdução de Sinais , Animais , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Músculos , Proteínas Serina-Treonina Quinases/metabolismo
7.
Biochem Biophys Res Commun ; 612: 119-125, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35523049

RESUMO

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.


Assuntos
Diabetes Mellitus , Proteínas Serina-Treonina Quinases , Animais , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Obesidade
8.
Development ; 146(13)2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31118231

RESUMO

Asymmetric cell division is a key step in cellular differentiation in multicellular organisms. In plants, asymmetric zygotic division produces the apical and basal cells. The mitogen-activated protein kinase (MPK) cascade in Arabidopsis acts in asymmetric divisions such as zygotic division and stomatal development, but whether the effect on cellular differentiation of this cascade is direct or indirect following asymmetric division is not clear. Here, we report the analysis of a rice mutant, globular embryo 4 (gle4). In two- and four-cell-stage embryos, asymmetric zygotic division and subsequent cell division patterns were indistinguishable between the wild type and gle4 mutants. However, marker gene expression and transcriptome analyses showed that specification of the basal region was compromised in gle4 We found that GLE4 encodes MPK6 and that GLE4/MPK6 is essential in cellular differentiation rather than in asymmetric zygotic division. Our findings provide a new insight into the role of MPK in plant development. We propose that the regulation of asymmetric zygotic division is separate from the regulation of cellular differentiation that leads to apical-basal polarity.


Assuntos
Divisão Celular Assimétrica/genética , Proteína Quinase 6 Ativada por Mitógeno/fisiologia , Oryza , Zigoto/citologia , Divisão Celular/genética , Clonagem Molecular , Regulação Enzimológica da Expressão Gênica , Regulação da Expressão Gênica de Plantas , Proteína Quinase 6 Ativada por Mitógeno/genética , Oryza/embriologia , Oryza/enzimologia , Oryza/genética , Plantas Geneticamente Modificadas , Sementes/genética , Sementes/metabolismo
9.
BMC Plant Biol ; 22(1): 488, 2022 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-36229795

RESUMO

BACKGROUND: N-acyl-homoserine lactones (AHLs) are used as quorum-sensing signals by Gram-negative bacteria, but they can also affect plant growth and disease resistance. N-decanoyl-L-homoserine lactone (C10-HSL) is an AHL that has been shown to inhibit primary root growth in Arabidopsis, but the mechanisms underlying its effects on root architecture are unclear. Here, we investigated the signaling components involved in C10-HSL-mediated inhibition of primary root growth in Arabidopsis, and their interplay, using pharmacological, physiological, and genetic approaches. RESULTS: Treatment with C10-HSL triggered a transient and immediate increase in the concentrations of cytosolic free Ca2+ and reactive oxygen species (ROS), increased the activity of mitogen-activated protein kinase 6 (MPK6), and induced nitric oxide (NO) production in Arabidopsis roots. Inhibitors of Ca2+ channels significantly alleviated the inhibitory effect of C10-HSL on primary root growth and reduced the amounts of ROS and NO generated in response to C10-HSL. Inhibition or scavenging of ROS and NO neutralized the inhibitory effect of C10-HSL on primary root growth. In terms of primary root growth, the respiratory burst oxidase homolog mutants and a NO synthase mutant were less sensitive to C10-HSL than wild type. Activation of MPKs, especially MPK6, was required for C10-HSL to inhibit primary root growth. The mpk6 mutant showed reduced sensitivity of primary root growth to C10-HSL, suggesting that MPK6 plays a key role in the inhibition of primary root growth by C10-HSL. CONCLUSION: Our results indicate that MPK6 acts downstream of ROS and upstream of NO in the response to C10-HSL. Our data also suggest that Ca2+, ROS, MPK6, and NO are all involved in the response to C10-HSL, and may participate in the cascade leading to C10-HSL-inhibited primary root growth in Arabidopsis.


Assuntos
Arabidopsis , 4-Butirolactona/análogos & derivados , Acil-Butirolactonas/farmacologia , Bactérias , Proteína Quinase 6 Ativada por Mitógeno , Óxido Nítrico/farmacologia , Percepção de Quorum , Espécies Reativas de Oxigênio
10.
Cell Tissue Res ; 384(1): 113-127, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33404840

RESUMO

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.


Assuntos
Macrófagos/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Osteoartrite/terapia , Fator de Crescimento Transformador beta1/metabolismo , Animais , Modelos Animais de Doenças , Masculino , Células-Tronco Mesenquimais/citologia , Osteoartrite/genética , Osteoartrite/metabolismo , Osteoartrite/patologia , Ratos , Ratos Sprague-Dawley , Transfecção
11.
BMC Cancer ; 21(1): 155, 2021 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-33579235

RESUMO

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.


Assuntos
Movimento Celular , Regulação Neoplásica da Expressão Gênica , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Neoplasias Cutâneas/patologia , Fatores de Transcrição/metabolismo , Ativação Transcricional , Proteínas Supressoras de Tumor/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Proteína Quinase 6 Ativada por Mitógeno/genética , Fosforilação , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Proteínas rac1 de Ligação ao GTP/genética
12.
Biol Pharm Bull ; 44(11): 1662-1669, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34719643

RESUMO

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.


Assuntos
Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/toxicidade , Hipocampo/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Fármacos Neuroprotetores/farmacologia , Tiofenos/farmacologia , Animais , Western Blotting , Linhagem Celular , Relação Dose-Resposta a Droga , Imunofluorescência , Camundongos
13.
J Integr Plant Biol ; 63(10): 1815-1842, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34270159

RESUMO

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.


Assuntos
Resistência à Doença , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases , Oryza/enzimologia , Xanthomonas , Interações Hospedeiro-Patógeno , MAP Quinase Quinase 4/metabolismo , Proteína Quinase 6 Ativada por Mitógeno/metabolismo
14.
FASEB J ; 33(12): 14423-14439, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31670972

RESUMO

Ischemic stroke is one of the leading causes of long-term disability worldwide. It arises when the blood flow to the brain is severely impaired, causing brain infarction. The current therapies for ischemic stroke are tissue plasminogen activator and mechanical thrombectomy, which re-establishes blood circulation to the brain but offers no neuroprotective effects. Excitotoxicity, particularly through the N-methyl-d-aspartate receptor (NMDAR), has been heavily implicated in the pathophysiology of brain infarction resulting from ischemic stroke. Here we investigated the interaction between NMDAR and metabotropic glutamate receptor 1 (mGluR1) as a novel target to develop potential neuroprotective agents for ischemic stroke. Through coimmunoprecipitation and affinity binding assay, we revealed that the interaction is mediated through 2 distinct sites on the mGluR1 C terminus. We then found that the disruption of mGluR1-GluN2A subunit of NMDAR (GluN2A) protected the primary mouse hippocampal neurons against NMDAR-mediated excitotoxicity and reversed the NMDAR-mediated regulation of ERK1/2 in rat hippocampal slices. The same protection was also observed in an animal model of ischemic stroke, alleviating brain infarction and yielding better motor recovery. These findings confirmed the existence of a receptor-receptor interaction between NMDAR and mGluR1, implicating this interconnection as a potential treatment target site for ischemic stroke.-Lai, T. K. Y., Zhai, D. Su, P., Jiang, A., Boychuk, J., Liu, F. The receptor-receptor interaction between mGluR1 receptor and NMDA receptor: a potential therapeutic target for protection against ischemic stroke.


Assuntos
Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/prevenção & controle , Animais , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/citologia , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase 6 Ativada por Mitógeno/genética , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , N-Metilaspartato/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/genética , Receptores de N-Metil-D-Aspartato/genética
15.
Bioorg Med Chem Lett ; 30(22): 127551, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-32927028

RESUMO

Triazolo[4,5-d]pyrimidin-5-amines were identified from kinase selectivity screening as novel ERK3 inhibitors with sub-100 nanomolar potencies in a biochemical assay using MK5 as substrate and with an attractive kinase selectivity profile. ERK3 crystal structures clarified the inhibitor binding mode in the ATP pocket with impact on A-loop, GC-loop and αC-helix conformations suggesting a potential structural link towards MK5 interaction via the FHIEDE motif. The inhibitors also showed sub-100 nM potencies in a cellular ERK3 NanoBRET assay and with excellent correlation to the biochemical IC50s. This novel series provides valuable tool compounds to further investigate the biological function and activation mechanism of ERK3.


Assuntos
Proteína Quinase 6 Ativada por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Humanos , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Modelos Moleculares , Estrutura Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Relação Estrutura-Atividade
16.
Int J Mol Sci ; 21(21)2020 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-33114754

RESUMO

Extracellular signal-regulated kinase 3 (ERK3), known also as mitogen-activated protein kinase 6 (MAPK6), is an atypical member of MAPK kinase family, which has been poorly studied. Little is known regarding its function in biological processes, yet this atypical kinase has been suggested to play important roles in the migration and invasiveness of certain cancers. The lack of tools, such as a selective inhibitor, hampers the study of ERK3 biology. Here, we report the crystal structure of the kinase domain of this atypical MAPK kinase, providing molecular insights into its distinct ATP binding pocket compared to the classical MAPK ERK2, explaining differences in their inhibitor binding properties. Medium-scale small molecule screening identified a number of inhibitors, several of which unexpectedly exhibited remarkably high inhibitory potencies. The crystal structure of CLK1 in complex with CAF052, one of the most potent inhibitors identified for ERK3, revealed typical type-I binding mode of the inhibitor, which by structural comparison could likely be maintained in ERK3. Together with the presented structural insights, these diverse chemical scaffolds displaying both reversible and irreversible modes of action, will serve as a starting point for the development of selective inhibitors for ERK3, which will be beneficial for elucidating the important functions of this understudied kinase.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteína Quinase 6 Ativada por Mitógeno/química , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Sítios de Ligação , Cristalografia por Raios X , Humanos , Proteína Quinase 6 Ativada por Mitógeno/antagonistas & inibidores , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Bibliotecas de Moléculas Pequenas/química
17.
Int J Mol Sci ; 21(11)2020 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-32516969

RESUMO

Extracellular signal-regulated kinase 3 (ERK3) is an atypical member of the mitogen-activated protein kinase (MAPK) family. It harbors a kinase domain in the N-terminus and a long C-terminus extension. The C-terminus extension comprises a conserved in ERK3 and ERK4 (C34) region and a unique C-terminus tail, which was shown to be required for the interaction of ERK3 with the cytoskeletal protein septin 7. Recent studies have elucidated the role of ERK3 signaling in promoting the motility and invasiveness of cancer cells. However, little is known about the intramolecular regulation of the enzymatic activity and cellular functions of ERK3. In this study, we investigated the role of the elongated C-terminus extension in regulating ERK3 kinase activity and its ability to promote cancer cell migration and invasion. Our study revealed that the deletion of the C-terminus tail greatly diminishes the ability of ERK3 to promote the migration and invasion of lung cancer cells. We identified two molecular mechanisms underlying this effect. Firstly, the deletion of the C-terminus tail decreases the kinase activity of ERK3 towards substrates, including the oncogenic protein steroid receptor co-activator 3 (SRC-3), an important downstream target for ERK3 signaling in cancer. Secondly, in line with the previous finding that the C-terminus tail mediates the interaction of ERK3 with septin 7, we found that the depletion of septin 7 abolished the ability of ERK3 to promote migration, indicating that septin 7 acts as a downstream effector for ERK3-induced cancer cell migration. Taken together, the findings of this study advance our understanding of the molecular regulation of ERK3 signaling by unraveling the role of the C-terminus tail in regulating ERK3 kinase activity and functions in cancer cells. These findings provide useful insights for the development of therapeutic agents targeting ERK3 signaling in cancer.


Assuntos
Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Neoplasias/etiologia , Neoplasias/metabolismo , Domínios e Motivos de Interação entre Proteínas , Movimento Celular/genética , Ativação Enzimática , Humanos , Proteína Quinase 6 Ativada por Mitógeno/química , Proteína Quinase 6 Ativada por Mitógeno/genética , Neoplasias/patologia , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas/genética , Transdução de Sinais
18.
J Biol Chem ; 293(42): 16193-16205, 2018 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-30166347

RESUMO

ERK3 is an atypical mitogen-activated protein kinase (MAPK) that has recently gained interest for its role in promoting cancer cell migration and invasion. However, the molecular regulation of ERK3 functions in cancer cells is largely unknown. ERK3 has a single phospho-acceptor site (Ser189) in its activation motif rather than the TXY conserved in conventional MAPKs such as ERK1/2. Although dual phosphorylation of the TXY motif is known to be critical for the activation of conventional MAPKs, the role of Ser189 phosphorylation in ERK3 activity and its function in cancer cells remain elusive. In this study, we revealed that activation loop phosphorylation is important for ERK3 in promoting cancer cell invasiveness, as the S189A mutation greatly decreased the ability of ERK3 to promote migration and invasion of lung cancer cells. Interestingly, a catalytically inactive ERK3 mutant was still capable of increasing migration and invasion, although to a lesser extent compared with WT ERK3, suggesting that ERK3 promotes cancer cell invasiveness by both kinase-dependent and kinase-independent mechanisms. To elucidate how the S189A mutation reduces the invasiveness-promoting ability of ERK3, we tested its effect on the kinase activity of ERK3 toward steroid receptor coactivator 3 (SRC3), a recently identified substrate of ERK3 critical for cancer cell invasiveness. Compared with ERK3, ERK3-S189A exhibited a dramatic decrease in kinase activity toward SRC3 and a concomitantly reduced ability to stimulate matrix metalloproteinase expression. Taken together, our study unravels the importance of Ser189 phosphorylation for intramolecular regulation of ERK3 kinase activity and invasiveness-promoting ability in lung cancer cells.


Assuntos
Neoplasias Pulmonares/patologia , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Sítios de Ligação , Movimento Celular , Humanos , Invasividade Neoplásica , Coativador 3 de Receptor Nuclear/metabolismo , Fosforilação , Serina/metabolismo
19.
J Biol Chem ; 293(22): 8342-8361, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29674317

RESUMO

Epithelia contribute to physical barriers that protect internal tissues from the external environment and also support organ structure. Accordingly, establishment and maintenance of epithelial architecture are essential for both embryonic development and adult physiology. Here, using gene knockout and knockdown techniques along with gene profiling, we show that extracellular signal-regulated kinase 3 (ERK3), a poorly characterized atypical mitogen-activated protein kinase (MAPK), regulates the epithelial architecture in vertebrates. We found that in Xenopus embryonic epidermal epithelia, ERK3 knockdown impairs adherens and tight-junction protein distribution, as well as tight-junction barrier function, resulting in epidermal breakdown. Moreover, in human epithelial breast cancer cells, inhibition of ERK3 expression induced thickened epithelia with aberrant adherens and tight junctions. Results from microarray analyses suggested that transcription factor AP-2α (TFAP2A), a transcriptional regulator important for epithelial gene expression, is involved in ERK3-dependent changes in gene expression. Of note, TFAP2A knockdown phenocopied ERK3 knockdown in both Xenopus embryos and human cells, and ERK3 was required for full activation of TFAP2A-dependent transcription. Our findings reveal that ERK3 regulates epithelial architecture, possibly together with TFAP2A.


Assuntos
Neoplasias da Mama/patologia , Embrião não Mamífero/enzimologia , Células Epiteliais/química , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Fator de Transcrição AP-2/metabolismo , Xenopus laevis/fisiologia , Animais , Neoplasias da Mama/enzimologia , Sistemas CRISPR-Cas , Adesão Celular , Membrana Celular , Células Cultivadas , Embrião não Mamífero/citologia , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Feminino , Células Hep G2 , Humanos , Proteína Quinase 6 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 6 Ativada por Mitógeno/genética , Junções Íntimas , Fator de Transcrição AP-2/antagonistas & inibidores , Fator de Transcrição AP-2/genética , Xenopus laevis/embriologia
20.
J Cell Physiol ; 234(8): 13220-13232, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30569573

RESUMO

Mitogen-activated protein kinase 6 (MAPK6) represents an atypical MAPK also known as extracellular signal-regulated kinase 3 (ERK3), which has been shown to play roles in cell motility and metastasis. ERK3 promotes migration and invasion of lung cancer cells and head and neck cancer cells by regulating the expression and/or activity of proteins involved in cancer progression. For instance, ERK3 upregulates matrix metallopeptidases and thereby promotes cancer cell invasiveness, and it phosphorylates tyrosyl-DNA phosphodiesterase 2, thereby enhancing chemoresistance in lung cancer. Here we discovered that ERK3 plays a converse role in melanoma. We observed that BRAF, an oncogenic Ser/Thr kinase, upregulates ERK3 expression levels by increasing both ERK3 messenger RNA levels and protein stability. Interestingly, although BRAF's kinase activity was required for upregulating ERK3 gene transcription, BRAF stabilized ERK3 protein in a kinase-independent fashion. We further demonstrate that ERK3 inhibits the migration, proliferation and colony formation of melanoma cells. In line with this, high level of ERK3 predicted increased survival among patients with melanomas. Taken together, these results indicate that ERK3 acts as a potent suppressor of melanoma cell growth and invasiveness.


Assuntos
Melanoma/enzimologia , Melanoma/patologia , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Invasividade Neoplásica/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Xenoenxertos , Humanos , Camundongos , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA