Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Biomed Pharmacother ; 161: 114460, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36870282

RESUMEN

Myricetin is a typical flavonol with various pharmacological effects which shows favorable biological activities in cancer. However, the underlying mechanisms and potential targets of myricetin in NSCLC (non-small cell lung cancer) cells remain unclear. First, we demonstrated that myricetin not only inhibited the proliferation, migration and invasion, but also induced apoptosis in A549 and H1299 cells in a dose-dependent manner. Then, we confirmed myricetin may play an anti-NSCLC effect through modulating MAPK-related functions and signaling pathway by Network pharmacology. Furthermore, MKK3 (MAP Kinase Kinase 3) was identified and confirmed as a potential target of myricetin by biolayer interferometry (BLI) and molecular docking, revealing that myricetin directly bound to MKK3. Moreover, three mutations (D208, L240, and Y245) of key amino acids predicted by molecular docking obviously decreased the affinity between myricetin and MKK3. Finally, enzyme activity assay was utilized to determine the effect of myricetin on MKK3 activity in vitro, and the result showed that myricetin attenuated MKK3 activity. Subsequently, myricetin decreased the phosphorylation of p38 MAPK. Furthermore, knockdown of MKK3 reduced the susceptibility of A549 and H1299 cells to myricetin. These results suggested that myricetin inhibited the growth of NSCLC cells via targeting MKK3 and influencing the downstream p38 MAPK signaling pathway. The findings revealed that MKK3 is a potential target of myricetin in the NSCLC and myricetin is considered to be a small-molecular inhibitor of MKK3, which can improve comprehension of the molecular mechanisms of myricetin pharmacological effects in cancer and further development of MKK3 inhibitors.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , MAP Quinasa Quinasa 3/genética , MAP Quinasa Quinasa 3/metabolismo , Simulación del Acoplamiento Molecular , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
J Orthop Surg Res ; 17(1): 455, 2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-36243801

RESUMEN

BACKGROUND: Apart from the current understanding of enzyme function, the mechanism of ectonucleotide pyrophosphatase/phosphodiesterase 1 (Enpp1) deficiency-associated osteoporosis is unknown. We aimed to explore the changes in the expression of signaling pathways of bone tissues involved in Enpp1 deficiency. METHODS: The body weights and morphology and histology of the bones of male Enpp1 knockout (KO) and wild-type (WT) mice were assessed. The humeri of WT and Enpp1 KO mice at 12 weeks of age were subjected to high-throughput quantitative molecular measurements, and bioinformatics analysis was performed. Proteins from humeri and calvarial pre-osteoblasts (Pobs) were used to verify the differentially expressed signaling pathways and to explain the mechanism of Enpp1 deficiency-associated osteoporosis. RESULTS: Enpp1 KO mice had significantly lower body weight and trabecular bone mass in the hindlimbs than WT mice. Proteomics and immunoblotting showed that Enpp1 deletion downregulated the expression of the p38 mitogen-activated protein kinase (MAPK) signaling pathway in bones. Lysophosphatidic acid (LPA) was involved in activating the MKK3/p38 MAPK/PCNA pathway and proliferating Pobs in Enpp1 KO mice, whereas a p38 MAPK inhibitor suppressed the LPA-induced pro-proliferation phenotype (p < 0.05). CONCLUSION: The inhibition of MKK3/p38 MAPK/PCNA pathway plays an important role in the development of osteoporosis caused by Enpp1 deficiency, and LPA partially rescued the proliferation of pre-osteoblasts via the MKK3/p38 MAPK/PCNA pathway.


Asunto(s)
Osteoporosis , Proteínas Quinasas p38 Activadas por Mitógenos , Animales , Masculino , Ratones , MAP Quinasa Quinasa 3/genética , MAP Quinasa Quinasa 3/metabolismo , Ratones Noqueados , Osteoporosis/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Pirofosfatasas/genética , Pirofosfatasas/metabolismo , Transducción de Señal/genética
3.
Int J Mol Sci ; 22(22)2021 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-34830095

RESUMEN

Compared to other ethnicities, Hispanic children incur the highest rates of leukemia, and most cases are diagnosed as Acute Lymphoblastic Leukemia (ALL). Despite improved treatment and survival for ALL, disproportionate health outcomes in Hispanics persist. Thus, it is essential to identify oncogenic mutations within this demographic to aid in the development of new strategies to diagnose and treat ALL. Using whole-exome sequencing, five single nucleotide polymorphisms within mitogen-activated protein kinase 3 (MAP2K3) were identified in an ALL cancer patient library from the U.S./Mexico border. MAP2K3 R26T and P11T are located near the substrate-binding site, while R65L and R67W localized to the kinase domain. Truncated-MAP2K3 mutant Q73* was also identified. Transfection in HEK293 cells showed that the quadruple-MEK3 mutant (4M-MEK3) impacted protein stability, inducing degradation and reducing expression. The expression of 4M-MEK3 could be rescued by cysteine/serine protease inhibition, and proteasomal degradation of truncated-MEK3 occurred in a ubiquitin-independent manner. MEK3 mutants displayed reduced auto-phosphorylation and enzymatic activity, as seen by decreases in p38 phosphorylation. Furthermore, uncoupling of the MEK3/p38 signaling pathway resulted in less suppressive activity on HEK293 cell viability. Thus, disruption of MEK3 activation may promote proliferative signals in ALL. These findings suggest that MEK3 represents a potential therapeutic target for treating ALL.


Asunto(s)
Proliferación Celular/genética , MAP Quinasa Quinasa 3 , Sistema de Señalización de MAP Quinasas/genética , Mutación , Proteínas de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras , Proteolisis , Células HEK293 , Células Hep G2 , Humanos , MAP Quinasa Quinasa 3/genética , MAP Quinasa Quinasa 3/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
4.
World Neurosurg ; 150: e696-e704, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33774214

RESUMEN

OBJECTIVE: Von Hippel-Lindau (VHL) syndrome is a rare disease that occurs in an autosomal-dominant genetic pattern. Due to the high genetic variability of VHL diseases, current studies have limited clinical value. Moreover, casual genetic variations in patients with VHL syndrome are still unclear. METHODS: Here, we performed whole-exome sequencing of 25 individuals to identify reliable disease-related variations. Systemic computational analysis was performed for variant detection, and Sanger sequencing was used to validate detected mutations. RESULTS: Most of the known mutations in the VHL gene were observed in the studied population. In addition, a large fragment deletion in VHL exon 2 in the immediate family members of the last family was detected. This had not been reported earlier. Moreover, we identified 3 novel mutation sites in the MAP2K3 gene that may be involved in the occurrence and development of the VHL disease. CONCLUSIONS: These results demonstrated that the heterogeneous nature of VHL syndrome and novel mutational signatures may help to improve the diagnostic ability of VHL syndrome.


Asunto(s)
MAP Quinasa Quinasa 3/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/genética , Análisis Mutacional de ADN , Femenino , Humanos , Masculino , Mutación , Linaje , Secuenciación del Exoma
5.
Mol Oncol ; 15(5): 1566-1583, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33660414

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is one of the most refractory malignancies worldwide. Mitogen-activated protein kinase 3 (MAP2K3) has a contradictory role in tumor progression, and the function and expression patterns of MAP2K3 in ESCC remain to be determined. We found that MAP2K3 expression to be downregulated in ESCC, and MAP2K3 downregulation correlated with clinically poor survival. MAP2K3 inhibited ESCC cell proliferation and invasion in vitro and in vivo. MAP2K3 suppressed STAT3 expression and activation. Mechanistically, MAPSK3 interacted with MDM2 to promote STAT3 degradation via the ubiquitin-proteasome pathway. Furthermore, exosomal miR-19b-3p derived from the plasma of patients with ESCC could suppress MAP2K3 expression to promote ESCC tumorigenesis. STAT3 was found to bind to the MIR19B promoter and increased the expression of miR-19b-3p in ESCC cells. In summary, our results demonstrated that the miR-19b-3p-MAP2K3-STAT3 feedback loop regulates ESCC tumorigenesis and elucidates the potential of therapeutically targeting this pathway in ESCC.


Asunto(s)
Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/patología , MAP Quinasa Quinasa 3/fisiología , MicroARNs/fisiología , Factor de Transcripción STAT3/fisiología , Adulto , Anciano , Animales , Estudios de Casos y Controles , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago/genética , Retroalimentación Fisiológica/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , MAP Quinasa Quinasa 3/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Persona de Mediana Edad , Invasividad Neoplásica , Factor de Transcripción STAT3/genética
6.
J Integr Plant Biol ; 63(7): 1324-1340, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33605510

RESUMEN

Mitogen-activated protein kinases (MPKs) play essential roles in guard cell signaling, but whether MPK cascades participate in guard cell ethylene signaling and interact with hydrogen peroxide (H2 O2 ), nitric oxide (NO), and ethylene-signaling components remain unclear. Here, we report that ethylene activated MPK3 and MPK6 in the leaves of wild-type Arabidopsis thaliana as well as ethylene insensitive2 (ein2), ein3, nitrate reductase1 (nia1), and nia2 mutants, but this effect was impaired in ethylene response1 (etr1), nicotinamide adenine dinucleotide phosphate oxidase AtrbohF, mpk kinase1 (mkk1), and mkk3 mutants. By contrast, the constitutive triple response1 (ctr1) mutant had constitutively active MPK3 and MPK6. Yeast two-hybrid, bimolecular fluorescence complementation, and pull-down assays indicated that MPK3 and MPK6 physically interacted with MKK1, MKK3, and the C-terminal region of EIN2 (EIN2 CEND). mkk1, mkk3, mpk3, and mpk6 mutants had typical levels of ethylene-induced H2 O2 generation but impaired ethylene-induced EIN2 CEND cleavage and nuclear translocation, EIN3 protein accumulation, NO production in guard cells, and stomatal closure. These results show that the MKK1/3-MPK3/6 cascade mediates ethylene-induced stomatal closure by functioning downstream of ETR1, CTR1, and H2 O2 to interact with EIN2, thereby promoting EIN3 accumulation and EIN3-dependent NO production in guard cells.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Proteínas de Unión al ADN/metabolismo , Etilenos/farmacología , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 3/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estomas de Plantas/efectos de los fármacos , Estomas de Plantas/metabolismo , Receptores de Superficie Celular/metabolismo , Factores de Transcripción/metabolismo , Arabidopsis/efectos de los fármacos , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Unión al ADN/genética , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 3/genética , Proteínas Quinasas Activadas por Mitógenos/genética , Receptores de Superficie Celular/genética , Factores de Transcripción/genética
7.
Cell Death Dis ; 11(8): 713, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32873775

RESUMEN

Esophageal cancer is one of the most common cancer with limited therapeutic strategies, thus it is important to develop more effective strategies to against it. Sulforaphene (SFE), an isothiocyanate isolated from radish seeds, was proved to inhibit esophageal cancer progression in the current study. Flow cytometric analysis showed SFE induced cell apoptosis and cycle arrest in G2/M phase. Also, scrape motility and transwell assays presented SFE reduced esophageal cancer cell metastasis. Microarray results showed the influence of SFE on esophageal cancer cells was related with stearoyl-CoA desaturase (SCD), cadherin 3 (CDH3), mitogen-activated protein kinase kinase 3 (MAP2K3) and growth arrest and DNA damage inducible beta (GADD45B). SCD and CDH3 could promote esophageal cancer metastasis via activating the Wnt pathway, while the latter one was involved in a positive feedback loop, GADD45B-MAP2K3-p38-p53, to suppress esophageal cancer growth. GADD45B was known to be the target gene of p53, and we proved in this study, it could increase the phosphorylation level of MAP2K3 in esophageal cancer cells, activating p38 and p53 in turn. SFE treatment elevated MAP2K3 and GADD45B expression and further stimulated this feedback loop to better exert antitumor effect. In summary, these results demonstrated that SFE had the potential for developing as a chemotherapeutic agent because of its inhibitory effects on esophageal cancer metastasis and proliferation.


Asunto(s)
Cadherinas/metabolismo , Isotiocianatos/farmacología , Estearoil-CoA Desaturasa/metabolismo , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Cadherinas/fisiología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Femenino , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Isotiocianatos/metabolismo , MAP Quinasa Quinasa 3/genética , MAP Quinasa Quinasa 3/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Desnudos , Estearoil-CoA Desaturasa/fisiología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Proc Natl Acad Sci U S A ; 117(33): 19994-20003, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32747557

RESUMEN

The transcriptional regulator YAP, which plays important roles in the development, regeneration, and tumorigenesis, is activated when released from inhibition by the Hippo kinase cascade. The regulatory mechanism of YAP in Hippo-low contexts is poorly understood. Here, we performed a genome-wide RNA interference screen to identify genes whose loss of function in a Hippo-null background affects YAP activity. We discovered that the coatomer protein complex I (COPI) is required for YAP nuclear enrichment and that COPI dependency of YAP confers an intrinsic vulnerability to COPI disruption in YAP-driven cancer cells. We identified MAP2K3 as a YAP regulator involved in inhibitory YAP phosphorylation induced by COPI subunit depletion. The endoplasmic reticulum stress response pathway activated by COPI malfunction appears to connect COPI and MAP2K3. In addition, we provide evidence that YAP inhibition by COPI disruption may contribute to transcriptional up-regulation of PTGS2 and proinflammatory cytokines. Our study offers a resource for investigating Hippo-independent YAP regulation as a therapeutic target for cancers and suggests a link between YAP and COPI-associated inflammatory diseases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína Coat de Complejo I/metabolismo , MAP Quinasa Quinasa 3/metabolismo , Neoplasias/metabolismo , Interferencia de ARN , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular Tumoral , Proteína Coat de Complejo I/genética , Regulación Neoplásica de la Expresión Génica , Genoma , Vía de Señalización Hippo , Humanos , MAP Quinasa Quinasa 3/genética , Ratones , Neoplasias/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Proteínas Señalizadoras YAP
10.
Theranostics ; 10(17): 7906-7920, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32685028

RESUMEN

Background: Capsaicin is an active compound found in plants of the Capsicum genus; it has a range of therapeutic benefits, including anti-tumor effects. Here we aimed to delineate the inhibitory effects of capsaicin on nasopharyngeal carcinoma (NPC). Methods: The anti-cancer effects of capsaicin were confirmed in NPC cell lines and xenograft mouse models, using CCK-8, clonogenic, wound-healing, transwell migration and invasion assays. Co-immunoprecipitation, western blotting and pull-down assays were used to determine the effects of capsaicin on the MKK3-p38 axis. Cell proliferation and EMT marker expression were monitored in MKK3 knockdown (KD) or over-expression NPC cell lines treated with or without capsaicin. Finally, immunohistochemistry was performed on NPC specimens from NPC patients (n = 132) and the clinical relevance was analyzed. Results: Capsaicin inhibited cell proliferation, mobility and promoted apoptosis in NPC cells. Then we found that capsaicin directly targets p38 for dephosphorylation. As such, MKK3-induced p38 activation was inhibited by capsaicin. Furthermore, we found that capsaicin-induced inhibition of cell motility was mediated by fucokinase. Xenograft models demonstrated the inhibitory effects of capsaicin treatment on NPC tumor growth in vivo, and analysis of clinical NPC samples confirmed that MKK3 phosphorylation was associated with NPC tumor growth and lymphoid node metastasis. Conclusions: The MKK3-p38 axis represents a potential therapeutic target for capsaicin. MKK3 phosphorylation might serve as a biomarker to identify NPC patients most likely to benefit from adjunctive capsaicin treatment.


Asunto(s)
Capsaicina/farmacología , MAP Quinasa Quinasa 3/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Carcinoma Nasofaríngeo/tratamiento farmacológico , Neoplasias Nasofaríngeas/tratamiento farmacológico , Animales , Capsaicina/uso terapéutico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , MAP Quinasa Quinasa 3/genética , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Persona de Mediana Edad , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/patología , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Fosforilación/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Int J Mol Sci ; 21(10)2020 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-32429593

RESUMEN

The activation of p38 mitogen-activated protein kinases (MAPKs) through a phosphorylation cascade is the canonical mode of regulation. Here, we report a novel activation mechanism for p38α. We show that Arg49 and Arg149 of p38α are methylated by protein arginine methyltransferase 1 (PRMT1). The non-methylation mutations of Lys49/Lys149 abolish the promotive effect of p38α on erythroid differentiation. MAPK kinase 3 (MKK3) is identified as the major p38α upstream kinase and MKK3-mediated activation of the R49/149K mutant p38α is greatly reduced. This is due to a profound reduction in the interaction of p38α and MKK3. PRMT1 can enhance both the methylation level of p38α and its interaction with MKK3. However, the phosphorylation of p38α by MKK3 is not a prerequisite for methylation. MAPK-activated protein kinase 2 (MAPKAPK2) is identified as a p38α downstream effector in the PRMT1-mediated promotion of erythroid differentiation. The interaction of MAPKAPK2 with p38α is also significantly reduced in the R49/149K mutant. Together, this study unveils a novel regulatory mechanism of p38α activation via protein arginine methylation on R49/R149 by PRMT1, which impacts partner interaction and thus promotes erythroid differentiation. This study provides a new insight into the complexity of the regulation of the versatile p38α signaling and suggests new directions in intervening p38α signaling.


Asunto(s)
Arginina/metabolismo , Eritropoyesis/genética , MAP Quinasa Quinasa 3/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , MAP Quinasa Quinasa 3/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Espectrometría de Masas , Metilación , Mutación , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Represoras/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
12.
Gene ; 724: 144146, 2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-31634561

RESUMEN

miRNA mediated genetic regulation is widely involved in carcinogenesis of cervical cancer. In this study, miR-214 was confirmed to directly regulate MKK3 via imperfect base-pairing to its 3'UTR, resulting down-regulation of its expression level. Compared to normal tissues, a down-regulated level of miR-214 was observed in cervical cancer, while MKK3 was up-regulated. Next, we demonstrated that over-expression of miR-214 or knockdown of MKK3 can inhibit the growth, proliferation, invasion and migration of cervical cancer in vitro and in vivo. Moreover, the effects of miR-214 in HeLa cells were rescued by the restoration of MKK3. In conclusion, our results laid new foundations for investigating the pathogenesis and diagnosis of cervical cancer.


Asunto(s)
MAP Quinasa Quinasa 3/genética , MicroARNs/genética , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Regiones no Traducidas 3' , Animales , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , MAP Quinasa Quinasa 3/metabolismo , Ratones Desnudos , MicroARNs/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cell Death Dis ; 10(11): 842, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31695024

RESUMEN

Colorectal cancer (CRC) is one of the most common malignant tumors worldwide and understanding its underlying molecular mechanisms is crucial for the development of therapeutic strategies. The mitogen-activated protein kinase-kinase 3 (MKK3) is a specific activator of p38 MAP kinases (p38 MAPKs), which contributes to the regulation of several cellular functions, such as proliferation, differentiation, apoptosis as well as response to drugs. At present, the exact MKK3/p38 MAPK pathway contribution in cancer is heavily debated because of its pleiotropic function. In this work, we retrospectively explored the prognostic and pathobiologic relevance of MKK3 in a cohort of CRC patients and assessed MKK3 molecular functions in a panel of CRC lines and colonocytes primary cultures. We found increased MKK3 levels in late-stage CRC patients which correlated with shorter overall survival. Herein, we report that the MKK3 targeting by inducible RNA interference univocally exerts antitumor effects in CRC lines but not in primary colonocytes. While MKK3 depletion per se affects growth and survival by induction of sustained autophagy and death in some CRC lines, it potentiates response to chemotherapeutic drug 5-fluorouracil (5-FU) in all of the tested CRC lines in vitro. Here, we demonstrate for the first time that in CRC the MKK3 specifically activates p38delta MAPK isoform to sustain prosurvival signaling and that such effect is exacerbated upon 5-FU challenge. Indeed, p38delta MAPK silencing recapitulates MKK3 depletion effects in CRC cells in vitro and in vivo. Overall, our data identified a molecular mechanism through which MKK3 supports proliferation and survival signaling in CRC, further supporting MKK3 as a novel and extremely attractive therapeutic target for the development of promising strategies for the management of CRC patients.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , MAP Quinasa Quinasa 3/genética , Proteína Quinasa 13 Activada por Mitógenos/genética , Autofagia/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Supervivencia sin Enfermedad , Femenino , Fluorouracilo/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HT29 , Xenoinjertos , Humanos , Masculino , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos
14.
Mol Med Rep ; 20(2): 1797-1807, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31257538

RESUMEN

Melanoma is an aggressive skin carcinoma with poor prognosis, and is prevalent worldwide. It was demonstrated that microRNA (miR)­21 and mitogen­activated protein kinase kinase 3 (MKK3) both participated in the occurrence and development of various tumors; however, their detailed roles in the progression of melanoma remain unclear. Reverse transcription­quantitative PCR (RT­qPCR) and western blot analyses were conducted to examine the expression levels of miR­21 and MKK3 in clinical specimens of patients with melanoma and melanoma cell lines. A dual­luciferase reporter assay was performed to verify the target interaction between miR­21 and MKK3. The mRNA and protein expressions of MKK3 were measured using RT­qPCR and western blot analysis, respectively, following transfection with miR­21 mimics and inhibitor. Subsequently, Cell Counting Kit­8 and colony formation assays, and flow cytometry were conducted to assess the effects of miR­21 and MKK3 on the cell growth of melanoma. Cell migration and invasion experiments were performed to evaluate the effects of miR­21 and MKK3 on the cell metastasis of melanoma. It was revealed that MKK3 was upregulated, and miR­21 was downregulated in patients with melanoma and melanoma cell lines. MKK3 was demonstrated to be a direct target of miR­21. Furthermore, it was demonstrated that upregulated miR­21 expression and downregulated MKK3 expression suppressed cell proliferation and colony formation, promoted apoptosis, delayed the cell cycle, and inhibited cell migration and invasion. The present findings suggested that miR­21 could inhibit the cell growth and metastasis of melanoma by negatively regulating MKK3.


Asunto(s)
Carcinogénesis/genética , MAP Quinasa Quinasa 3/genética , Melanoma/genética , MicroARNs/genética , Adulto , Anciano , Apoptosis/genética , Movimiento Celular/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Melanoma/patología , Persona de Mediana Edad , Metástasis de la Neoplasia , Fosfohidrolasa PTEN/genética
15.
J Biol Chem ; 294(15): 5867-5878, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30760523

RESUMEN

Endothelial dysfunction is induced by inflammatory mediators including multiple G protein-coupled receptor (GPCR) agonists. However, the GPCR signaling pathways that promote endothelial dysfunction are incompletely understood. We previously showed that thrombin promotes endothelial barrier disruption through autophosphorylation and activation of p38 mitogen-activated protein kinase (MAPK) via a non-canonical transforming growth factor-ß-activated protein kinase-1-binding protein-1 (TAB1) and TAB2-dependent pathway rather than the canonical three-tiered kinase cascade. Here, we sought to determine whether other GPCR agonists stimulate p38 MAPK activation via this non-canonical pathway in human endothelial cells derived from different vascular beds. Using primary human umbilical vein endothelial cells (HUVECs), HUVEC-derived EA.hy926 cells, and human dermal microvascular endothelial cells (HDMECs), we found that both non-canonical and canonical p38 activation pathways components are expressed in these various endothelial cell types, including TAB3, a structurally-related TAB2 homolog. Moreover, multiple GPCRs agonists, including thrombin, histamine, prostaglandin E2, and ADP, stimulated robust p38 autophosphorylation, whereas phosphorylation of the upstream MAPKs MAP kinase kinase 3 (MKK3) and MKK6, was virtually undetectable, indicating that non-canonical p38 activation may exist for other GPCRs. Indeed, in EA.hy926 cells, thrombin- and histamine-stimulated p38 activation depended on TAB1-TAB2, whereas in primary HUVECs, both TAB1-TAB2 and TAB1-TAB3 were required for p38 activation. In HDMECs, thrombin-induced p38 activation depended on TAB1-TAB3, but histamine-induced p38 activation required TAB1-TAB2. Moreover, thrombin- and histamine-stimulated interleukin-6 production required both TAB1-TAB2 and TAB1-TAB3 in HUVEC. We conclude that multiple GPCR agonists utilize non-canonical TAB1-TAB2 and TAB1-TAB3-dependent p38 activation to promote endothelial inflammatory responses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Adenosina Difosfato/genética , Adenosina Difosfato/metabolismo , Línea Celular , Dinoprostona/genética , Dinoprostona/metabolismo , Histamina/genética , Histamina/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interleucina-6/biosíntesis , Interleucina-6/genética , MAP Quinasa Quinasa 3/genética , MAP Quinasa Quinasa 3/metabolismo , Fosforilación/genética , Trombina/genética , Trombina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética
16.
Cell Death Dis ; 10(3): 149, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30770795

RESUMEN

The c-Jun N-terminal kinase (JNK) pathway plays essential roles in regulating a variety of physiological processes including cell migration and invasion. To identify critical factors that regulate JNK-dependent cell migration, we carried out a genetic screen in Drosophila based on the loss-of-cell polarity-triggered cell migration in the wing epithelia, and identified MKK3 licorne (lic) as an essential regulator of JNK-mediated cell migration and invasion. We found that loss of lic suppressed ptc > scrib-IR or ptc > Egr triggered cell migration in the wing epithelia, and Rasv12/lgl-/- induced tumor invasion in the eye discs. In addition, ectopic expression of Lic is sufficient to induce JNK-mediated but p38-independent cell migration, and cooperate with oncogenic Ras to promote tumor invasion. Consistently, Lic is able to activate JNK signaling by phosphorylating JNK, which up-regulates the matrix metalloproteinase MMP1 and integrin, characteristics of epithelial-mesenchymal transition (EMT). Moreover, lic is required for physiological JNK-mediate cell migration in thorax development. Finally, expression of human MKK3 in Drosophila is able to initiate JNK-mediated cell migration, cooperates with oncogenic Ras to trigger tumor invasion, and rescue loss-of-lic induced thorax closure defect. As previous studies suggest that MKK3 specifically phosphorylates and activates p38MAPK, our data provide the first in vivo evidence that MKK3 regulates JNK-dependent cell migration and invasion, a process evolutionarily conserved from flies to human.


Asunto(s)
Movimiento Celular/genética , Proteínas de Drosophila/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa 3/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Proteínas Quinasas/metabolismo , Animales , Animales Modificados Genéticamente , Polaridad Celular/genética , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/genética , Transición Epitelial-Mesenquimal/genética , Evolución Molecular , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Cadenas beta de Integrinas/metabolismo , MAP Quinasa Quinasa 3/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Invasividad Neoplásica/genética , Proteínas Quinasas/genética
17.
Biochem Biophys Res Commun ; 494(1-2): 358-364, 2017 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-28988112

RESUMEN

Acute pancreatitis is a common threat to human health. Caerulin provokes severe inflammations, causing injuries to surrounding pancreatic cells. TNF receptor-associated factor 3 (TRAF3) is a highly versatile regulator of immune response. The current study aims to understand the potential effect of TRAF3 on caerulin-induced pro-inflammatory responses. In the primary-cultured mouse bone marrow-derived macrophages (BMDMs), caerulin induced TRAF3 protein stabilization, which formed a complex with mitogen-activated protein kinase kinase 3 (MKK3) to mediate downstream p38 activation. Lentiviral shRNA-mediated TRAF3 stable knockdown significantly attenuated caerulin-induced MKK3-p38 activation and production of several key pro-inflammatory cytokines, including interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α) and IL-17. Remarkably, TRAF3 knockdown in caerulin-stimulated BMDMs also alleviated cytotoxicity to Panc02 and primary mouse pancreatic cells. Thus, TRAF3 is required for caerulin-induced p38 activation and macrophage-mediated pro-inflammatory responses. TRAF3 expression in macrophages could be a novel therapeutic target protein for the treatment of acute pancreatitis.


Asunto(s)
Ceruletida/farmacología , Células Epiteliales/efectos de los fármacos , Macrófagos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor 3 Asociado a Receptor de TNF/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Animales , Técnicas de Cocultivo , Células Epiteliales/citología , Células Epiteliales/inmunología , Regulación de la Expresión Génica , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Lentivirus/genética , Lentivirus/inmunología , MAP Quinasa Quinasa 3/genética , MAP Quinasa Quinasa 3/inmunología , Macrófagos/citología , Macrófagos/inmunología , Ratones , Páncreas/citología , Páncreas/efectos de los fármacos , Páncreas/inmunología , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología , Factor 3 Asociado a Receptor de TNF/antagonistas & inhibidores , Factor 3 Asociado a Receptor de TNF/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
18.
Apoptosis ; 22(11): 1362-1379, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28836036

RESUMEN

Cancer cells often have faulty apoptotic pathways resulting in sustenance of survivability, tumour metastasis and resistance to anticancer drugs. Alternate strategies are sought to improve therapeutic efficacy and therefore HepG2 cells were treated with S-allyl-cysteine (SAC) and berberine (BER) to analyze their mechanistic impact upon necroptosis along with its interacting relationship to apoptosis. In the present study we observed that SAC and BER exposure reduced NFκß nuclear translocation through adenylate cyclase-cAMP-protein kinaseA axis and eventually evaded c-FLIP inhibition. Effective RIP1 k63-polyubiquitination and persistent MKK3/MKK6 expression during drug treatment potentiated caspase8 activity via p53-DISC conformation. Resultant tBid associated lysosomal protease mediated AIF truncation induced DNA fragmentation and persuaded effector caspase mediated scramblase activation resulting induction of necroptosis in parallel to apoptotic events. SAC+BER effectively reduced Rb-phosphorylation resulting insignificant nuclear E2F presence led to ending of cell proliferation. Therefore necroptosis augmented the drug response and may be targeted alongside cell proliferation inhibition in formation of efficient therapeutics against liver cancer.


Asunto(s)
Adenilil Ciclasas/genética , Berberina/farmacología , Cisteína/análogos & derivados , MAP Quinasa Quinasa 3/genética , MAP Quinasa Quinasa 6/genética , Transducción de Señal , Adenilil Ciclasas/metabolismo , Apoptosis/efectos de los fármacos , Factor Inductor de la Apoptosis/genética , Factor Inductor de la Apoptosis/metabolismo , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Línea Celular , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Cisteína/farmacología , Fragmentación del ADN/efectos de los fármacos , Combinación de Medicamentos , Regulación de la Expresión Génica , Células Hep G2 , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , MAP Quinasa Quinasa 3/metabolismo , MAP Quinasa Quinasa 6/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Necrosis/inducido químicamente , Necrosis/genética , Necrosis/metabolismo , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Transporte de Proteínas , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
19.
J Biol Chem ; 292(39): 16161-16173, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28739872

RESUMEN

The kinase p38α MAPK (p38α) plays a pivotal role in many biological processes. p38α is activated by canonical upstream kinases that phosphorylate the activation region. The purpose of our study was to determine whether such activation may depend on redox-sensing cysteines within p38α. p38α was activated and formed a disulfide-bound heterodimer with MAP2K3 (MKK3) in rat cardiomyocytes and isolated hearts exposed to H2O2 This disulfide heterodimer was sensitive to reduction by mercaptoethanol and was enhanced by the thioredoxin-reductase inhibitor auranofin. We predicted that Cys-119 or Cys-162 of p38α, close to the known MKK3 docking domain, were relevant for these redox characteristics. The C119S mutation decreased whereas the C162S mutation increased the dimer formation, suggesting that these two Cys residues act as vicinal thiols, consistent with C119S/C162S being incapable of sensing H2O2 Similarly, disulfide heterodimer formation was abolished in H9C2 cells expressing both MKK3 and p38α C119S/C162S and subjected to simulated ischemia and reperfusion. However, the p38α C119S/C162S mutants did not exhibit appreciable alteration in activating dual phosphorylation. In contrast, the anti-inflammatory agent 10-nitro-oleic acid (NO2-OA), a component of the Mediterranean diet, reduced p38α activation and covalently modified Cys-119/Cys-162, probably obstructing MKK3 access. Moreover, NO2-OA reduced the dephosphorylation of p38α by hematopoietic tyrosine phosphatase (HePTP). Furthermore, steric obstruction of Cys-119/Cys-162 by NO2-OA pretreatment in Langendorff-perfused murine hearts prevented the p38-MKK3 disulfide dimer formation and attenuated H2O2-induced contractile dysfunction. Our findings suggest that cysteine residues within p38α act as redox sensors that can dynamically regulate the association between p38 and MKK3.


Asunto(s)
Cistina/metabolismo , Ventrículos Cardíacos/enzimología , MAP Quinasa Quinasa 3/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Modelos Moleculares , Miocitos Cardíacos/enzimología , Estrés Oxidativo , Sustitución de Aminoácidos , Animales , Línea Celular , Células Cultivadas , Cisteína/química , Cisteína/metabolismo , Cistina/química , Activación Enzimática , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Humanos , Técnicas In Vitro , MAP Quinasa Quinasa 3/química , MAP Quinasa Quinasa 3/genética , Masculino , Ratones Endogámicos C57BL , Proteína Quinasa 14 Activada por Mitógenos/química , Proteína Quinasa 14 Activada por Mitógenos/genética , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Conformación Proteica , Multimerización de Proteína , Ratas Wistar , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo
20.
EMBO Mol Med ; 9(9): 1244-1262, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28674080

RESUMEN

Atherosclerosis, the major cause of cardiovascular disease, is a chronic inflammatory disease characterized by the accumulation of lipids and inflammatory cells in the artery wall. Aberrant expression of microRNAs has been implicated in the pathophysiological processes underlying the progression of atherosclerosis. Here, we define the contribution of miR-21 in hematopoietic cells during atherogenesis. Interestingly, we found that miR-21 is the most abundant miRNA in macrophages and its absence results in accelerated atherosclerosis, plaque necrosis, and vascular inflammation. miR-21 expression influences foam cell formation, sensitivity to ER-stress-induced apoptosis, and phagocytic clearance capacity. Mechanistically, we discovered that the absence of miR-21 in macrophages increases the expression of the miR-21 target gene, MKK3, promoting the induction of p38-CHOP and JNK signaling. Both pathways enhance macrophage apoptosis and promote the post-translational degradation of ABCG1, a transporter that regulates cholesterol efflux in macrophages. Altogether, these findings reveal a major role for hematopoietic miR-21 in atherogenesis.


Asunto(s)
Apoptosis , Aterosclerosis/fisiopatología , Macrófagos/inmunología , MicroARNs/inmunología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/inmunología , Animales , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Vasos Sanguíneos/inmunología , Femenino , Humanos , MAP Quinasa Quinasa 3/genética , MAP Quinasa Quinasa 3/inmunología , Macrófagos/citología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Necrosis/genética , Necrosis/inmunología , Necrosis/patología , Necrosis/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA