Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.214
Filtrar
1.
Arch Insect Biochem Physiol ; 114(4): e22054, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37700521

RESUMEN

Signaling pathways regulate the transmission of signals during organism growth and development, promoting the smooth and accurate completion of numerous physiological and biochemical reactions. The extracellular signal-regulated kinase (ERK) signaling pathway is an essential pathway involved in regulating various physiological processes, such as cell proliferation, differentiation, adhesion, migration, and more. This pathway also contributes to several important physiological processes in silkworms, including protein synthesis, reproduction, and immune defense against pathogens. Organizing related studies on the ERK signaling pathway in silkworms can provide a better understanding of its mechanism in Lepidopterans and develop a theoretical foundation for improving cocoon production and new strategies for pest biological control.


Asunto(s)
Bombyx , Quinasas MAP Reguladas por Señal Extracelular , Lepidópteros , Animales , Bombyx/genética , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Transducción de Señal
2.
Neuropharmacology ; 202: 108835, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34648772

RESUMEN

Peripheral nerve injuries (PNIs) often result in persistent neuropathic pain, seriously affecting quality of life. Existing therapeutic interventions for PNI-induced neuropathic pain are far from satisfactory. Extracellular signal-regulated kinases (ERKs) and p38 have been found to participate in triggering and maintaining PNI-induced neuropathic pain. However, ERK and p38 also contribute to axonal regeneration and motor function recovery after PNI, making it difficult to inhibit ERK and p38 for therapeutic purposes. In this study, we simultaneously characterized neuropathic pain and motor function recovery in a mouse sciatic nerve crush injury model to identify the time window for therapeutic interventions. We further demonstrated that delayed delivery of a combination of ERK and p38 inhibitors at three weeks after PNI could significantly alleviate PNI-induced neuropathic pain without affecting motor function recovery. Additionally, the combined use of these two inhibitors could suppress pain markedly better than either inhibitor alone, possibly reducing the required dose of each inhibitor and alleviating the side effects and risks of the inhibitors when used individually.


Asunto(s)
Butadienos/farmacología , Butadienos/uso terapéutico , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Imidazoles/farmacología , Imidazoles/uso terapéutico , Neuralgia/tratamiento farmacológico , Neuralgia/etiología , Nitrilos/farmacología , Nitrilos/uso terapéutico , Traumatismos de los Nervios Periféricos/complicaciones , Traumatismos de los Nervios Periféricos/fisiopatología , Piridinas/farmacología , Piridinas/uso terapéutico , Nervio Ciático/lesiones , Nervio Ciático/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Axones/fisiología , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Ratones Endogámicos C57BL , Regeneración Nerviosa/genética , Neuralgia/genética , Recuperación de la Función , Resultado del Tratamiento , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
Front Immunol ; 12: 709229, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34691022

RESUMEN

Preterm labor (PTL) is a multifactorial syndrome that results in birth prior to 37 weeks of gestation. However, the specific molecular mechanisms underlying this condition have yet to be elucidated. Previous research demonstrated that the abnormal expression of IL-27, and its receptors, played a role in the pathophysiology of preterm labor. In the present study, we established a Lipopolysaccharide (LPS)-stimulated, infection-induced, preterm mouse model based on wild-type C57BL/6 mice and WSX-1-/-C57BL/6 mice. WSX-1 knockdown led to a significant delay in birth by 11.32 ± 2.157h. In addition, compared with wild-type C57B/6 mice, the expression levels of IFN-γ, IL-1ß, IL-6, TNF-α, and CXCL10, in the fetal membrane and myometrium of WSX-1-/-mice were significantly lower, particularly in the myometrium. We also confirmed similar pro-inflammatory effects arising from IL-27 in human amniotic cell line (WISH) and human myometrial smooth muscle cell line (HMSMC). Once stimulated by LPS, the pro-inflammatory action exhibited a synergistic effect and appeared to be time-dependent. Finally, we demonstrated that LY3214996, an inhibitor of the ERK pathway, significantly inhibited the pro-inflammatory effect mediated by IL-27. Overall, our data confirmed that the inflammatory effect mediated by the IL-27/IFN-r/ERK axis is involved in preterm labor. Our findings, therefore, provide an enhancement in our etiological understanding of the mechanisms underlying PTL.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/fisiología , Inflamación/etiología , Interleucina-27/fisiología , Trabajo de Parto Prematuro/inmunología , Animales , Células Cultivadas , Femenino , Humanos , Interferón gamma/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Embarazo , Receptores de Interleucina/fisiología , Útero/inmunología
4.
Mol Neurobiol ; 58(11): 5772-5789, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34406600

RESUMEN

Increasing evidence suggests that transmembrane protein 16A (TMEM16A) in nociceptive neurons is an important molecular component contributing to peripheral pain transduction. The present study aimed to evaluate the role and mechanism of TMEM16A in chronic nociceptive responses elicited by spared nerve injury (SNI). In this study, SNI was used to induce neuropathic pain. Drugs were administered intrathecally. The expression and cellular localization of TMEM16A, the ERK pathway, and NK-1 in the dorsal root ganglion (DRG) were detected by western blot and immunofluorescence. Behavioral tests were used to evaluate the role of TMEM16A and p-ERK in SNI-induced persistent pain and hypersensitivity. The role of TMEM16A in the hyperexcitability of primary nociceptor neurons was assessed by electrophysiological recording. The results show that TMEM16A, p-ERK, and NK-1 are predominantly expressed in small neurons associated with nociceptive sensation. TMEM16A is colocalized with p-ERK/NK-1 in DRG. TMEM16A, the MEK/ERK pathway, and NK-1 are activated in DRG after SNI. ERK inhibitor or TMEM16A antagonist prevents SNI-induced allodynia. ERK and NK-1 are downstream of TMEM16A activation. Electrophysiological recording showed that CaCC current increases and intrathecal application of T16Ainh-A01, a selective TMEM16A inhibitor, reverses the hyperexcitability of DRG neurons harvested from rats after SNI. We conclude that TMEM16A activation in DRG leads to a positive interaction of the ERK pathway with activation of NK-1 production and is involved in the development of neuropathic pain after SNI. Also, the blockade of TMEM16A or inhibition of the downstream ERK pathway or NK-1 upregulation may prevent the development of neuropathic pain.


Asunto(s)
Anoctaminas/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Ganglios Espinales/patología , Hiperalgesia/fisiopatología , Neuralgia/fisiopatología , Nervio Peroneo/lesiones , Receptores de Neuroquinina-1/fisiología , Células Receptoras Sensoriales/fisiología , Transducción de Señal/fisiología , Nervio Tibial/lesiones , Animales , Anoctaminas/antagonistas & inhibidores , Butadienos/farmacología , Dolor Crónico/etiología , Dolor Crónico/fisiopatología , Hiperalgesia/etiología , Ligadura , Masculino , Neuralgia/etiología , Nitrilos/farmacología , Nocicepción/fisiología , Pirimidinas/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Tiazoles/farmacología
5.
J Zhejiang Univ Sci B ; 22(6): 492-503, 2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34128372

RESUMEN

Dysregulated crosstalk between different signaling pathways contributes to tumor development, including resistance to cancer therapy. In the present study, we found that the mitogen-activated extracellular signal-regulated kinase (MEK) inhibitor trametinib failed to suppress the proliferation of PANC-1 and MGC803 cells by activating the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway, while the JAK2 inhibitor fedratinib failed to inhibit the growth of the PANC-1 cells upon stimulation of extracellular signal-regulated kinase (ERK) signaling. In particular, the most prominent enhancement of the anti-proliferative effect resulted from the concurrent blockage of the JAK2/STAT3 and ERK signaling pathways. Furthermore, the combination of the two inhibitors resulted in a reduced tumor burden in mice. Our evidence suggests novel crosstalk between JAK2/STAT3 and ERK signaling in gastric cancer (GC) and pancreatic ductal adenocarcinoma (PDAC) cells and provides a therapeutic strategy to overcome potential resistance in gastrointestinal cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Neoplasias Gastrointestinales/tratamiento farmacológico , Janus Quinasa 2/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Femenino , Neoplasias Gastrointestinales/patología , Humanos , Janus Quinasa 2/fisiología , Ratones , Ratones Endogámicos BALB C , Piridonas/uso terapéutico , Pirimidinonas/uso terapéutico , Pirrolidinas/uso terapéutico , Factor de Transcripción STAT3/fisiología , Transducción de Señal/efectos de los fármacos , Sulfonamidas/uso terapéutico , Microambiente Tumoral
6.
Transl Res ; 235: 129-143, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33894400

RESUMEN

Hereditary hemorrhagic telangiectasia (HHT) is a genetic disease characterized by vascular dysplasia. Mutations of the endoglin (ENG) gene that encodes a co-receptor of the transforming growth factor ß1 signaling pathway cause type I HHT. ENG is primarily expressed in endothelial cells (ECs), but its interaction with other key angiogenic pathways to control angiogenesis has not been well addressed. The aim of this study is to investigate ENG interplay with VEGFR2, FGFR1 and TIE2 in primary human ECs. ENG was knocked-down with siRNA in human umbilical vein ECs (HUVECs) and human lung microvascular ECs (HMVEC-L). Gene expression was measured by RT-qPCR and Western blotting. Cell signaling pathway activation was analyzed by detecting phosphor-ERK and phosphor-AKT levels. Cell migration and apoptosis were assessed using the Boyden chamber assay and the CCK-8 Kit, respectively. Loss of ENG in HUVECs led to significantly reduced expression of VEGFR2 but not TIE2 or FGFR1, which was also confirmed in HMVEC-L. HUVECs lacking ENG had significantly lower levels of active Rac1 and a substantial reduction of the transcription factor Sp1, an activator of VEGFR2 transcription, in nuclei. Furthermore, VEGF- but not bFGF- or angiopoietin-1-induced phosphor-ERK and phosphor-AKT were suppressed in ENG deficient HUVECs. Functional analysis revealed that ENG knockdown inhibited cell migratory but enhanced anti-apoptotic activity induced by VEGF. In contrast, bFGF, angiopoietin-1 and -2 induced HUVEC migration and anti-apoptotic activities were not affected by ENG knockdown. In conclusion, ENG deficiency alters the VEGF/VEGFR2 pathway, which may play a role in HHT pathogenesis.


Asunto(s)
Endoglina/fisiología , Células Endoteliales/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/fisiología , Receptor TIE-2/fisiología , Telangiectasia Hemorrágica Hereditaria/etiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Proteínas Proto-Oncogénicas c-akt/fisiología
7.
Mediators Inflamm ; 2021: 5582943, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33859537

RESUMEN

Inflammatory bowel disease (IBD), such as ulcerative colitis (UC) and Crohn's disease (CD), is an intractable intestinal inflammation associated with the disruption of the intestinal mucosa. We previously demonstrated that Lactobacillus brevis-derived long-chain polyphosphate (poly P) improved the intestinal barrier function by the upregulation of cell adhesion and relieved intestinal inflammation, thereby exerting a curing effect on colitis in vitro, in vivo, and in an investigator-initiated clinical study of UC. However, how poly P improves mucosal defects induced by intestinal inflammation has not been elucidated. In this study, we detected the accumulation of platelets in inflamed tissues induced by poly P in a dextran sulfate sodium- (DSS-) induced colitis mouse model. A light transmission aggregometry analysis and scanning electron microscopy showed that poly P promoted the platelet aggregation. An SRB assay and ki-67 staining showed that the supernatant of poly P-treated platelet-rich plasma (PRP) increased intestinal epithelial cell growth. A wound healing assay showed that the supernatant of poly P-treated PRP, but not poly P itself, accelerated wound healing. A Western blotting analysis indicated that mitogen-activated protein kinase activation was induced by the supernatant of poly P-treated human PRP in the epithelial cells and its wound healing effect was significantly decreased by the inhibition of ERK signaling. These data suggested that platelet-derived mediators induced by poly P improved intestinal inflammation through the promotion of epithelial cell growth by the activation of the ERK signaling pathway. The mechanism is a novel host-microbe interaction through mammalian platelet-derived mediators induced by bacterial molecules.


Asunto(s)
Mucosa Intestinal/efectos de los fármacos , Activación Plaquetaria/fisiología , Polifosfatos/farmacología , Probióticos/farmacología , Cicatrización de Heridas/efectos de los fármacos , Animales , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Ratones , Ratones Endogámicos BALB C , Agregación Plaquetaria/efectos de los fármacos
8.
J Invest Dermatol ; 141(11): 2646-2655.e6, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33865911

RESUMEN

IL-33 is a chromatin-associated multifunctional cytokine implicated in the pathogenesis of atopic dermatitis (AD), an inflammatory skin disorder characterized by skin barrier dysfunction. The previous reports show that IL-33 is highly detected in the nucleus of epidermal keratinocytes in AD lesions compared with that in unaffected or normal skin. However, it is unclear whether intracellular IL-33 directly contributes to the pathogenesis of AD. T helper type 2 cytokines IL-4 and IL-13 that are elevated in AD lesions suppress keratinocyte differentiation to impair skin barrier function. We investigated whether intracellular IL-33 is involved in IL-4 and IL-13 function. In monolayer culture and living skin equivalent analyses, IL-4 and IL-13 increased the expression of full-length IL-33 in the nucleus of keratinocytes by activating the MAPK/extracellular signal‒regulated kinase kinase/extracellular signal‒regulated kinase signaling pathway, which is necessary for the inhibition of differentiation markers FLG, LOR, keratin 1, and keratin 10. The nuclear IL-33 functions as a transcription cofactor of signal transducer and activator of transcription 3, increasing the binding of phosphorylated signal transducer and activator of transcription 3 to FLG promoter, thereby inhibiting its transcription, and it inhibits the expression of transcription factor RUNX1 by signal transducer and activator of transcription 3 and signal transducer and activator of transcription 6, thereby downregulating LOR, keratin 1, and keratin 10. Thus, the elevated nuclear IL-33 in the epidermis of AD lesions may be involved in the pathogenesis of AD by inhibiting keratinocyte differentiation and skin barrier function.


Asunto(s)
Dermatitis Atópica/etiología , Interleucina-13/farmacología , Interleucina-33/fisiología , Interleucina-4/farmacología , Queratinocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Unión al ADN/análisis , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Proteínas Filagrina/análisis , Proteínas Filagrina/genética , Humanos , Queratina-1/análisis , Queratina-10/análisis , Queratinocitos/química , Queratinocitos/citología , Sistema de Señalización de MAP Quinasas/fisiología , Factor de Transcripción STAT3/fisiología
9.
Diabetes ; 70(7): 1519-1535, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33906910

RESUMEN

In diabetic pathology, insufficiency in ß-cell mass, unable to meet peripheral insulin demand, and functional defects of individual ß-cells in production of insulin are often concurrently observed, collectively causing hyperglycemia. Here we show that the phosphorylation of ERK1/2 is significantly decreased in the islets of db/db mice as well as in those of a cohort of subjects with type 2 diabetes. In mice with abrogation of ERK signaling in pancreatic ß-cells through deletion of Mek1 and Mek2, glucose intolerance aggravates under high-fat diet-feeding conditions due to insufficient insulin production with lower ß-cell proliferation and reduced ß-cell mass, while in individual ß-cells dampening of the number of insulin exocytosis events is observed, with the molecules involved in insulin exocytosis being less phosphorylated. These data reveal bifunctional roles for MEK/ERK signaling in ß-cells for glucose homeostasis, i.e., in regulating ß-cell mass as well as in controlling insulin exocytosis in individual ß-cells, thus providing not only a novel perspective for the understanding of diabetes pathophysiology but also a potential clue for new drug development for diabetes treatment.


Asunto(s)
Glucemia/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Homeostasis , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Animales , Línea Celular , Dieta Alta en Grasa , Exocitosis , Humanos , Células Secretoras de Insulina/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Transducción de Señal
10.
J Invest Dermatol ; 141(10): 2391-2401.e13, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33857488

RESUMEN

Clinical studies have shown that diets enriched with omega-3 (also know as n-3) polyunsaturated fatty acids could relieve the symptoms of patients with psoriasis. However, the mechanisms involved remain poorly understood. The aim of this study was to investigate the effects of α-linolenic acid (ALA) on the proliferation and differentiation of psoriatic keratinocytes in a three-dimensional skin model. Skin models featuring healthy (healthy substitute) or psoriatic (psoriatic substitute) cells were engineered by the self-assembly method of tissue engineering using a culture medium supplemented with 10 µM ALA in comparison with the regular unsupplemented medium. ALA decreased keratinocyte proliferation and improved psoriatic substitute epidermal differentiation, as measured by decreased Ki67 staining and increased protein expression of FLG and loricrin. The added ALA was notably incorporated into the epidermal phospholipids and metabolized into long-chain n-3 polyunsaturated fatty acids, mainly eicosapentaenoic acid and n-3 docosapentaenoic acid. ALA supplementation led to increased levels of eicosapentaenoic acid derivatives (15-hydroxyeicosapentaenoic acid and 18-hydroxyeicosapentaenoic acid) as well as a decrease in levels of omega-6 (also know as n-6) polyunsaturated fatty acid lipid mediators (9-hydroxyoctadecadienoic acid, 12-hydroxyeicosatetraenoic acid, and leukotriene B4). Furthermore, the signal transduction mediators extracellular signal‒regulated kinases 1 and 2 were the kinases most activated after ALA supplementation. Taken together, these results show that ALA decreases the pathologic phenotype of psoriatic substitutes by normalizing keratinocyte proliferation and differentiation in vitro.


Asunto(s)
Queratinocitos/efectos de los fármacos , Psoriasis/tratamiento farmacológico , Ingeniería de Tejidos , Ácido alfa-Linolénico/farmacología , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/análisis , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Suplementos Dietéticos , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Queratinocitos/patología , Leucotrieno B4/análisis , Psoriasis/metabolismo , Psoriasis/patología , Ácido alfa-Linolénico/administración & dosificación
11.
Drug Dev Res ; 82(8): 1131-1143, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33818788

RESUMEN

Circular RNA FAT atypical cadherin 1 (circFAT1) has been reported to play vital roles in the progression of some cancers. However, the regulatory role and underlying mechanisms of circFAT1 in cervical cancer (CC) remain largely unknown. The expression of circFAT1, microRNA (miR)-409-3p and cyclin-dependent kinase 8 (CDK8) was detected using qRT-PCR and Western blot assays. Cell proliferation, apoptosis, migration and invasion in vitro were investigated using cell counting kit-8, colony formation, flow cytometry, and transwell assays, respectively. Western blot assay was used to determine the activation of ERK1/2 and p38 MAPK pathway. The interaction miR-409-3p and circFAT1 or CDK8 was confirmed by dual-luciferase reporter, pull-down or RIP assays. The effects of circFAT1 in vivo were determined using xenograft models. CircFAT1 was highly expressed in CC, and closely associated with poor prognosis. CircFAT1 knockdown resulted in the suppression of proliferation, migration and invasion, and promotion of apoptosis in CC cells via the inactivation of ERK1/2 and p38 MAPK pathway; also, circFAT1 silencing could inactivate this pathway and repressed CC tumor growth in vivo. Mechanistic analysis showed that circFAT1 directly sponged miR-409-3p and then relieved the repressive effect of miR-409-3p on its target CDK8. Furthermore, miR-409-3p inhibition reversed the effects of circFAT1 silencing on CC cells. Whereas, miR-409-3p overexpression impeded CC cell growth and motility, which was attenuated by CDK8. CircFAT1 promoted CC progression via activating ERK1/2 and p38 MAPK pathway through the miR-409-3p/CDK8 axis, suggesting a promising prognostic biomarker and therapeutic target for CC.


Asunto(s)
Cadherinas/genética , Quinasa 8 Dependiente de Ciclina/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , MicroARNs/fisiología , ARN Circular/fisiología , Neoplasias del Cuello Uterino/etiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Línea Celular Tumoral , Quinasa 8 Dependiente de Ciclina/genética , Progresión de la Enfermedad , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos BALB C
13.
Anticancer Res ; 41(3): 1219-1229, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33788713

RESUMEN

BACKGROUND: CD105 is highly expressed on human activated endothelial cells (ECs), is an important component of the TGF-ß1 receptor complex and is essential for angiogenesis. CD105 expression is up-regulated in activated ECs and is an important potential marker for cancer prognosis. MATERIALS AND METHODS: In vitro rat myoblasts transfected with the L-CD105 and S-CD105 transfectants. The transfectants were treated with TGF-ß1 for the angiogenesis study. RESULTS: L-CD105 affects cell proliferation in the presence and absence of TGF-ß1, and inhibits p-ERK1/2, p-MEK1/2 and p-c-Jun in L-CD105 transfectants compared to controls. The induction of phospho-ERK1/2 following treatment with TGF-ß1 remained significantly lower in L-CD105 transfectants compared to controls. CONCLUSION: L-CD105 inhibits the phosphorylation of ERK1/2, MEK1/2, c-Jun1/2/3, and associated signalling intermediates. CD105 modulates cell growth and TGF-ß1 induced cell signalling through ERK-c-Jun expression.


Asunto(s)
Endoglina/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias/prevención & control , Animales , Proliferación Celular , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Neoplasias/irrigación sanguínea , Neovascularización Patológica/etiología , Fosforilación , Ratas , Factor de Crecimiento Transformador beta1/farmacología
14.
Anticancer Res ; 41(3): 1231-1242, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33788714

RESUMEN

BACKGROUND/AIM: Successful therapy of EGFR-mutant NSCLC remains a challenging task despite initial benefits with the usage of EGFR tyrosine kinase inhibitors. Cancer immunotherapy has shown promising results in certain tumors, but response rate in EGFR-mutant NCLC is low, because these tumors are thought to have weak immunogenicity. MATERIALS AND METHODS: We used data from in vivo NSCLC databases as well as from in vitro cell culture experiments to investigate the regulation of CD73 by EGFR. RESULTS: EGFR expression was correlated with CD73 expression in patients' datasets, with EGFR-mutant tumors showing higher expression than their EGFR wildtype counterparts. Treatment of EGFR-mutant NSCLC cell lines with EGFR TKI reduced expression of CD73 at both mRNA and protein level. Among EGFR downstream signaling pathways, the Ras-Raf-ERK pathway was involved in the regulation of CD73 expression directly via ERK1/2 without the engagement of RSKs or MSKs. CONCLUSION: The results of this study may provide novel therapeutic strategies for the treatment of oncogene-driven NSCLC.


Asunto(s)
5'-Nucleotidasa/fisiología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Neoplasias Pulmonares/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas , 5'-Nucleotidasa/antagonistas & inhibidores , 5'-Nucleotidasa/genética , Carcinoma de Pulmón de Células no Pequeñas/fisiopatología , Línea Celular Tumoral , Receptores ErbB/fisiología , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/fisiología , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/fisiopatología , Sistema de Señalización de MAP Quinasas/fisiología , Transducción de Señal/fisiología
15.
Biochem J ; 478(4): 839-854, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33528492

RESUMEN

Aberrant differentiation of keratinocytes disrupts the skin barrier and causes a series of skin diseases. However, the molecular basis of keratinocyte differentiation is still poorly understood. In the present study, we examined the expression of C7ORF41 using tissue microarrays by immunohistochemistry and found that C7ORF41 is specifically expressed in the basal layers of skin epithelium and its expression is gradually decreased during keratinocytes differentiation. Importantly, we corroborated the pivotal role of C7ORF41 during keratinocyte differentiation by C7ORF41 knockdown or overexpression in TPA-induced Hacat keratinocytes. Mechanismly, we first demonstrated that C7ORF41 inhibited keratinocyte differentiation mainly through formatting a complex with IKKα in the cytoplasm, which thus blocked the nuclear translocation of IKKα. Furthermore, we also demonstrated that inhibiting the PKCα/ERK signaling pathway reversed the reduction in C7ORF41 in TPA-induced keratinocytes, indicating that C7ORF41 expression could be regulated by upstream PKCα/ERK signaling pathway during keratinocyte differentiation. Collectively, our study uncovers a novel regulatory network PKCα/ERK/C7ORF41/IKKα during keratinocyte differentiation, which provides potential therapeutic targets for skin diseases.


Asunto(s)
Epidermis/metabolismo , Quinasa I-kappa B/metabolismo , Queratinocitos/citología , Transducción de Señal/fisiología , Transporte Activo de Núcleo Celular , Diferenciación Celular , Línea Celular Transformada , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Queratinocitos/metabolismo , Proteína Quinasa C-alfa/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología
16.
J Surg Res ; 262: 224-239, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33039109

RESUMEN

BACKGROUND: Arteriovenous fistulae (AVF) are the preferred mode of vascular access for hemodialysis. Before use, AVF remodel by thickening and dilating to achieve a functional conduit via an adaptive process characterized by expression of molecular markers characteristic of both venous and arterial identity. Although signaling via EphB4, a determinant of venous identity, mediates AVF maturation, the role of its counterpart EphrinB2, a determinant of arterial identity, remains unclear. We hypothesize that EphrinB2 signaling is active during AVF maturation and may be a mechanism of venous remodeling. METHODS: Aortocaval fistulae were created or sham laparotomy was performed in C57Bl/6 mice, and specimens were examined on Days 7 or 21. EphrinB2 reverse signaling was activated with EphB4-Fc applied periadventitially in vivo and in endothelial cell culture medium in vitro. Downstream signaling was assessed using immunoblotting and immunofluorescence. RESULTS: Venous remodeling during AVF maturation was characterized by increased expression of EphrinB2 as well as Akt1, extracellular signal-regulated kinases 1/2 (ERK1/2), and p38. Activation of EphrinB2 with EphB4-Fc increased phosphorylation of EphrinB2, endothelial nitric oxide synthase, Akt1, ERK1/2, and p38 and was associated with increased diameter and wall thickness in the AVF. Both mouse and human endothelial cells treated with EphB4-Fc increased phosphorylation of EphrinB2, endothelial nitric oxide synthase, Akt1, ERK1/2, and p38 and increased endothelial cell tube formation and migration. CONCLUSIONS: Activation of EphrinB2 signaling by EphB4-Fc was associated with adaptive venous remodeling in vivo while activating endothelial cell function in vitro. Regulation of EphrinB2 signaling may be a new strategy to improve AVF maturation and patency.


Asunto(s)
Derivación Arteriovenosa Quirúrgica , Efrina-B2/fisiología , Remodelación Vascular/fisiología , Animales , Células Cultivadas , Células Endoteliales/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/fisiología , Receptor EphB4/farmacología , Transducción de Señal/fisiología , Venas/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
17.
Biomed Pharmacother ; 131: 110724, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32920518

RESUMEN

Stachydrine is a main active component of Leonurus japonicus (Chinese motherwort), which has traditionally been used to promote postpartum recovery and alleviate myocardial and cerebral ischemic injuries due to its pro-angiogenic effect. Our prior study demonstrated that stachydrine increased angiogenesis in zebrafish embryos, but its pro-angiogenic effect and underlying mechanisms on human umbilical vein endothelial cells (HUVECs) remain largely unknown. In the present study, we further investigated the role of stachydrine in sunitinib-injured HUVECs and its potential molecular mechanisms. The results showed that stachydrine exhibited a protective effect on sunitinib-injured HUVECs and significantly promoted their proliferation, migration, and tube formation, all central events of angiogenesis. In addition, stachydrine inhibited apoptosis and ROS production in sunitinib-injured HUVECs. Furthermore, our findings illustrated for the first time that stachydrine's molecular mechanisms for promoting angiogenesis might correlate with activation of the VEGFR2/MEK/ERK and inhibition of the mitochondrial-mediated apoptosis signaling pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Mitocondrias/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Neovascularización Fisiológica/efectos de los fármacos , Prolina/análogos & derivados , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Prolina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
18.
Biomed Pharmacother ; 131: 110750, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32942160

RESUMEN

Rhizoma Paris is a popular Chinese medicine in clinics. It contains four main saponins which are its major bioactive compounds. These saponins are Paris saponin I, II, VI and VII (PSI, PSII, PSVI and PSVII, respectively). Up to now, the research using HUVEC cells to evaluate the anti-angiogenic activity of four saponins is blank. The purpose of this study was to evaluate the anti-angiogenic properties (also known as angiotoxicity) of the four saponins in Rhizoma Paris on vascular endothelial cells-HUVEC cells, and to investigate the underlying mechanism, which has not been studied before. In this study, MTT assay, Lactate dehydrogenase (LDH) assay, wound healing experiments, transwell cell invasion assay, tubule formation experiment, DAPI staining, AV-PI double staining, and cell cycle analysis were used to determine the effects of Paris saponins. The results showed that, with increases in concentrations of PSI, PSII, PSVI and PSVII, the viability of HUVEC cells decreased significantly. In addition, four saponins dose-dependent enhanced LDH release and inhibited HUVEC cell migration, invasion, and angiogenesis. In terms of mechanism, PSI significantly inhibited protein expression in multiple signaling pathways. In particular, with the VEGF2 as the target, it activate the downstream PI3K / AKT / mTOR, SRC / eNOS, P38, PLCγ / ERK / MERK and JAK2/STAT3 signaling pathways. In conclusion, PSI, PSII, PSVI and PSVII can inhibit endothelial cell proliferation, migration and invasion, block endothelial cell cycle, induce endothelial cell apoptosis, act on protein expression in several anti-angiogenic signaling pathways, and finally inhibit angiogenesis in vitro. This study provides further data support for the clinical application of Paris saponins as antiangiogenic drugs.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Saponinas/farmacología , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Diosgenina/análogos & derivados , Diosgenina/farmacología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Janus Quinasa 2/fisiología , Óxido Nítrico Sintasa de Tipo III/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Fosfolipasa C gamma/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Factor de Transcripción STAT3/fisiología , Serina-Treonina Quinasas TOR/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Familia-src Quinasas/fisiología
19.
Br J Cancer ; 123(7): 1131-1144, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32606358

RESUMEN

BACKGROUND: Toward identifying new strategies to target gastric cancer stem-like cells (CSCs), we evaluated the function of the tumour suppressor CDK5 regulatory subunit-associated protein 3 (CDK5RAP3) in gastric CSC maintenance. METHODS: We examined the expression of CDK5RAP3 and CD44 in gastric cancer patients. The function and mechanisms of CDK5RAP3 were checked in human and mouse gastric cancer cell lines and in mouse xenograft. RESULTS: We show that CDK5RAP3 is weakly expressed in gastric CSCs and is negatively correlated with the gastric CSC marker CD44. CDK5RAP3 overexpression decreased expression of CSC markers, spheroid formation, invasion and migration, and reversed chemoresistance in gastric CSCs in vitro and vivo. CDK5RAP3 expression was found to be regulated by extracellular-related kinase (ERK) signalling. ERK inhibitors decreased spheroid formation, migration and invasion, and the expression of epithelial-to-mesenchymal transition (EMT)-related proteins in both GA cells and organoids derived from a genetically engineered mouse model of GA. Finally, CDK5RAP3 expression was associated with reduced lymph-node metastasis and better prognosis, even in the presence of high expression of the EMT transcription factor Snail, among patients with CD44-positive GA. CONCLUSIONS: Our results demonstrate that CDK5RAP3 is suppressed by ERK signalling and negatively regulates the self-renewal and EMT of gastric CSCs.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Neoplasias Gástricas/patología , Proteínas Supresoras de Tumor/fisiología , Animales , Proteínas de Ciclo Celular/análisis , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Humanos , Receptores de Hialuranos/análisis , Metástasis Linfática , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Células Madre Neoplásicas/fisiología , Proteínas Supresoras de Tumor/análisis
20.
FASEB J ; 34(9): 12289-12307, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32701200

RESUMEN

Given the rising evidence that gut malfunction including changes in the gut microbiota composition, plays a major role in the development of obesity and associated metabolic diseases, the exploring of novel probiotic bacteria with potential health benefits has attracted great attention. Recently Lactobacillus spp., exert potent anti-obesity effects by regulating key transcriptional and translational factors in adipose tissues. However, the molecular mechanism behind the anti-obesity effect of probiotics is not yet fully understood. Therefore, we investigated the effect of Lactobacillus plantarum A29 on the expression of adipogenic and lipogenic genes in 3T3-L1 adipocytes and high-fat diet (HFD)-fed mice. We observed that the treatment of 3T3-L1 adipocytes with the cell-free metabolites of L plantarum inhibited their differentiation and fat depositions via downregulating the key adipogenic transcriptional factors (PPAR-γ, C/EBP-α, and C/EBP-ß) and their downstream targets (FAS, aP2, ACC, and SREBP-1). Interestingly, supplementation with L plantarum reduced the fat mass and serum lipid profile concurrently with downregulation of lipogenic gene expression in the adipocytes, resulting in reductions in the bodyweight of HFD-fed obese mice. L plantarum treatment attenuated the development of obesity in HFD-fed mice via the activation of p38MAPK, p44/42, and AMPK-α by increasing their phosphorylation. Further analysis revealed that A29 modulated gut-associated microbiota composition. Thus, A 29 potential probiotic strain may alleviate the obesity development and its associated metabolic disorders via inhibiting PPARγ through activating the p38MAPK and p44/42 signaling pathways.


Asunto(s)
Disbiosis/terapia , Microbioma Gastrointestinal , Lactobacillus plantarum/fisiología , Obesidad/terapia , Probióticos/farmacología , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Diferenciación Celular , Dieta Alta en Grasa , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos ICR , Obesidad/metabolismo , Obesidad/microbiología , PPAR gamma/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...