Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Photochem Photobiol B ; 257: 112948, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38833786

RESUMEN

Autophagy participates in the regulation of ferroptosis. Among numerous autophagy-related genes (ATGs), ATG5 plays a pivotal role in ferroptosis. However, how ATG5-mediated ferroptosis functions in UVB-induced skin inflammation is still unclear. In this study, we unveil that the core ferroptosis inhibitor GPX4 is significantly decreased in human skin tissue exposed to sunlight. We report that ATG5 deletion in mouse keratinocytes strongly protects against UVB-induced keratinocyte ferroptosis and skin inflammation. Mechanistically, ATG5 promotes the autophagy-dependent degradation of GPX4 in UVB-exposed keratinocytes, which leads to UVB-induced keratinocyte ferroptosis. Furthermore, we find that IFN-γ secreted by ferroptotic keratinocytes facilitates the M1 polarization of macrophages, which results in the exacerbation of UVB-induced skin inflammation. Together, our data indicate that ATG5 exacerbates UVB-induced keratinocyte ferroptosis in the epidermis, which subsequently gives rise to the secretion of IFN-γ and M1 polarization. Our study provides novel evidence that targeting ATG5 may serve as a potential therapeutic strategy for the amelioration of UVB-caused skin damage.


Asunto(s)
Proteína 5 Relacionada con la Autofagia , Ferroptosis , Interferón gamma , Queratinocitos , Macrófagos , Rayos Ultravioleta , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Queratinocitos/citología , Proteína 5 Relacionada con la Autofagia/metabolismo , Proteína 5 Relacionada con la Autofagia/genética , Animales , Ratones , Interferón gamma/metabolismo , Macrófagos/metabolismo , Macrófagos/efectos de la radiación , Macrófagos/citología , Humanos , Piel/efectos de la radiación , Piel/metabolismo , Piel/patología , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Autofagia/efectos de la radiación , Inflamación/metabolismo , Inflamación/patología
2.
J Photochem Photobiol B ; 238: 112613, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36495669

RESUMEN

Engagement of regulated cell death in keratinocytes plays a crucial role in the pathogenesis and development of skin disorders associated with UV radiation. However, it remains unclear how microRNAs (miRNAs) participate in the regulation of UV-caused keratinocyte death. In this study, we found that miR-133a-3p was decreased in the epidermis of UVB-challenged mice and UVB-irradiated keratinocyte cell line HaCaT cells. The intradermal injection of agomir miR-133a-3p ameliorated skin damage of UVB-challenged mice, especially epidermal necrosis. Meanwhile, the injection inhibited apoptosis indicator PARP cleavage and pyroptosis indicator GSDME cleavage in the epidermis. In UVB-challenged HaCaT cells, transfection of miR-133a-3p mimic or inhibitor alleviated or aggravated UVB-induced apoptosis and GSDME-mediated pyroptosis respectively. miR-133a-3p was also involved in the effects of metformin treatment on alleviating skin damage in UVB-challenged mice and on inhibiting apoptosis and GSDME-mediated pyroptosis in UVB-irradiated HaCaT cells. We confirmed that CYLD is a target gene of miR-133a-3p and participates in the protective effects of miR-133a-3p on inhibiting UVB-caused apoptosis and GSDME-mediated pyroptosis in keratinocytes. This study indicates a pivotal role for miR-133a-3p of keratinocytes in UVB-caused skin damage. Alleviating skin photodamage by restoring the decrease of miR-133a-3p can be considered a potential therapeutic approach.


Asunto(s)
Gasderminas , MicroARNs , Animales , Ratones , Apoptosis , Queratinocitos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Piroptosis , Rayos Ultravioleta , Nanopartículas del Metal , Oro
3.
Cell Death Discov ; 8(1): 451, 2022 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-36344541

RESUMEN

Keratinocyte necroptosis (with proinflammatory characteristic) is required for epidermal damage in contact hypersensitivity (CHS). In DNCB-induced CHS mice model, we observed the aggravated keratinocyte death and increased phosphorylation level of MLKL, RIPK3 and RIPK1. However, CHS skin lesion did not present in keratinocyte-specific Mlkl knockout mice. We validated that MLKL-mediated keratinocyte necroptosis is required for epidermal damage in response to immune microenvironment in CHS. Moreover, MLKL-mediated necroptosis deficiency or inhibition resulted in blocking recruitment and activation of inflammatory cells in CHS via reducing HMGB1 release in keratinocytes. This study suggests that MLKL-mediated keratinocyte necroptosis functions as a self-amplified actor in inflammatory responses and could be considered as an effective therapeutic target. It proposes an innovative prospective that inhibiting keratinocyte necroptosis can prevent the development of epidermal damage in CHS.

4.
Cell Death Dis ; 13(10): 841, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36182937

RESUMEN

Gasdermin E (GSDME)-mediated pyroptosis is induced in keratinocytes of UVB-challenged skin. The role of GSDME in UVB-caused skin damage remains unknown. To explore the role of GSDME in UVB-induced skin inflammation. We compared differences in skin appearance, histological features, keratinocyte death modalities, infiltration of immune cells, and levels of some inflammatory cytokines between Gsdme-/- mice and wild type (WT) mice after UVB exposure. We explored whether keratinocytes contribute to GSDME deficiency-caused aggravation of UVB-induced skin inflammation in GSDME knockdown keratinocyte cultured in vitro and keratinocyte-specific Gsdme conditional knockout mice. We used anti-Ly6G antibody to deplete neutrophils and explore their role in UVB-caused skin damage. Skin damage and neutrophils infiltration were aggravated in UVB-challenged Gsdme-/- mice, compared with UVB-challenged WT mice. Apoptosis and necroptosis, which were initiated together with GSDME-mediated pyroptosis in UVB-challenged WT mice, were not enhanced in UVB-challenged Gsdme-/- mice. Neutrophils activation indicators and its recruiting cytokines were increased in skin tissue of UVB-challenged Gsdme-/- mice. However, GSDME knockdown did not lead to the further increase of mRNA and secretion of TNF-α and IL-6 in UVB-challenged keratinocytes. Skin damage was not aggravated in UVB-challenged Gsdme cKO mice. Neutrophils depletion alleviated UVB-caused skin damage in WT mice and Gsdme-/- mice, and eliminated its aggravation in Gsdme-/- mice. This study demonstrates that GSDME plays a restrictive role in UVB-induced skin damage through inhibiting excessive recruitment and activation of neutrophils in the immune microenvironment in UVB-caused skin inflammation. However, keratinocytes might not contribute to this restrictive function.


Asunto(s)
Dermatitis , Neutrófilos , Animales , Citocinas , Inflamación/etiología , Interleucina-6 , Queratinocitos , Ratones , Ratones Noqueados , Proteínas Citotóxicas Formadoras de Poros , ARN Mensajero , Piel , Factor de Necrosis Tumoral alfa , Rayos Ultravioleta/efectos adversos
5.
J Exp Clin Cancer Res ; 41(1): 246, 2022 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-35964097

RESUMEN

BACKGROUND: Malignant transformation of the epidermis is an essential process in the pathogenesis of cutaneous squamous-cell carcinoma (cSCC). Although evidence has demonstrated that CD147 plays key roles in various tumors, the role of CD147 in epidermal malignant transformation in vivo remains unclear. METHODS: Epidermal CD147-overexpression or knockout (EpiCD147-OE or EpiCD147-KO) transgenic mouse models were generated for in vivo study. RNA-sequencing and q-PCR were performed to identify the differentially expressed genes. Immunohistochemistry and flow cytometry were performed to investigate the role of CD147 in regulating myeloid-derived suppressor cells (MDSCs). Immunoprecipitation, EMSA and ChIP assays were performed to investigate the mechanism of CD147 in cell transformation. RESULTS: We found that specific overexpression of CD147 in the epidermis (EpiCD147-OE) induces spontaneous tumor formation; moreover, a set of chemokines and cytokines including CXCL1, which play essential function in MDSC recruitment, were significantly upregulated in EpiCD147-OE transgenic mice. As expected, overexpression of CD147 in the epidermis remarkably facilitated tumorigenesis by increasing the rate of tumor initiation and the number and size of tumors in the DMBA/TPA mouse model. Interestingly, the expression of CXCL1 and the infiltration of MDSCs were dramatically increased in EpiCD147-OE transgenic mice. Our findings also showed that knockdown of CD147 attenuated EGF-induced malignant transformation as well as CXCL1 expression in HaCaT cells. Consistently, CD147 was found overexpressed in cutaneous squamous cell carcinoma (cSCC), and positively related with the expression of CD33, a myeloid-associated marker. We further identified RSK2, a serine/threonine kinase, as an interacting partner of CD147 at the binding site of CD147D207-230. The interaction of CD147 and RSK2 activated RSK2, thus enhancing AP-1 transcriptional activation. Furthermore, EMSAs and ChIP assays showed that AP-1 could associate with the CXCL1 promoter. Importantly, RSK2 inhibitor suppressed the tumor growth in DMBA/TPA mouse model by inhibiting the recruitment of MDSCs. CONCLUSION: Our findings demonstrate that CD147 exerts a key function in epidermal malignant transformation in vivo by activating keratinocytes and recruiting MDSCs via the RSK2/AP-1 pathway.


Asunto(s)
Basigina/metabolismo , Carcinoma de Células Escamosas , Neoplasias Cutáneas , Animales , Carcinoma de Células Escamosas/genética , Transformación Celular Neoplásica/metabolismo , Epidermis/metabolismo , Epidermis/patología , Ratones , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Neoplasias Cutáneas/genética , Factor de Transcripción AP-1/genética
6.
J Dermatol Sci ; 102(1): 25-35, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33642112

RESUMEN

BACKGROUND: Excessive inflammation and cell death induced by ultraviolet (UV) cause skin photodamage. Metformin possesses anti-inflammatory and cytoprotective effects. However, whether metformin inhibits inflammation and cell death in UVB-induced acute skin damage is unclear. OBJECTIVE: To evaluate the anti-inflammatory and cytoprotective effects of metformin in vitro and in vivo. Furthermore, its potential mechanism has been explored. METHODS: Transcriptome sequencing and multiplex cytokines analysis were used to evaluate the validity of in vitro UVB-induced acute damage keratinocyte model and anti-inflammatory effects of metformin. We also determined the expression and nuclear translocation of CCAAT/enhancer-binding protein beta (C/EBPß), an important transcriptional factor of Interleukin-1beta (IL-1ß). Cell viability and cell death of keratinocytes were evaluated upon UVB irradiation in the presence or absence of metformin. 0.6% metformin cream was applied on UVB-irradiated mice to explore its pharmacological effects in vivo. RESULTS: Transcriptional landscape of 50 mJ/cm2 UVB-irradiated HaCaT cells is typical of UVB-induced acute damage keratinocyte model in vitro. Metformin alleviated transcription and secretion of IL-1ß, Tumor Necrosis Factor-alpha, and Fibroblast Growth Factor 2, expression and nuclear translocation of C/EBPß in this model. Metformin also protected keratinocytes from cell death caused by UVB-induced cellular secretions, which contributed to its cytoprotective effects. Topical administration of 0.6% metformin cream alleviated UVB-induced skin damage in mice. CONCLUSION: We proved the protective roles of metformin in UVB-challenged keratinocytes and UVB-irradiated mice, which indicated the potential value of metformin in topical therapy against skin photodamage.


Asunto(s)
Antiinflamatorios/farmacología , Queratinocitos/efectos de los fármacos , Metformina/farmacología , Piel/efectos de los fármacos , Rayos Ultravioleta/efectos adversos , Administración Cutánea , Animales , Antiinflamatorios/uso terapéutico , Carcinogénesis/efectos de los fármacos , Carcinogénesis/patología , Carcinogénesis/efectos de la radiación , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Células HaCaT , Humanos , Queratinocitos/patología , Queratinocitos/efectos de la radiación , Metformina/uso terapéutico , Ratones , Especies Reactivas de Oxígeno , Piel/patología , Piel/efectos de la radiación , Envejecimiento de la Piel/efectos de los fármacos , Envejecimiento de la Piel/efectos de la radiación , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/prevención & control , Quemadura Solar/etiología , Quemadura Solar/patología , Quemadura Solar/prevención & control
9.
Biomark Res ; 8: 41, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32944244

RESUMEN

Long non-coding RNAs (lncRNAs) represent an important class of RNAs comprising more than 200 nucleotides, which are produced by RNA polymerase II. Although lacking an open reading framework and protein-encoding activity, lncRNAs can mediate endogenous gene expression by serving as chromatin remodeler, transcriptional or post-transcriptional modulator, and splicing regulator during gene modification. In recent years, increasing evidence shows the significance of lncRNAs in many malignancies, with vital roles in tumorigenesis and cancer progression. Moreover, lncRNAs were also considered potential diagnostic and prognostic markers in cancer. The lncRNA small nuclear RNA host gene 16 (SNHG16), found on chromosome 17q25.1, represents a novel tumor-associated lncRNA. SNHG16 was recently found to exhibit dysregulated expression in a variety of malignancies. There are growing evidence of SNHG16's involvement in characteristics of cancer, including proliferation, apoptosis, together with its involvement in chemoresistance. In addition, SNHG16 has been described as a promising diagnostic and prognostic biomarker in cancer patients. The current review briefly summarizes recently reported findings about SNHG16 and discuss its expression, roles, mechanisms, and diagnostic and prognostic values in human cancers.

10.
Int J Oncol ; 57(5): 1203-1213, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32901832

RESUMEN

EphA2 (EPH receptor A2) (erythropoietin­producing hepatocellular receptor tyrosine kinase subtype A2) plays a crucial role in human cancers, and is a promising target for the development of new anticancer drugs. In this study, we showed that the interaction of Annexin A1 (ANXA1) and EphA2 increased EphA2 stability by inhibiting its proteasome degradation in gastric cancer (GC) and colon cancer (CC) cells, and the amino acid residues 20­30 and 28­30 of ANXA1 N terminal were responsible for binding and stabilizing EphA2. Based on the amino acid residues of ANXA1 responsible for binding EphA2, we developed ANXA1­derived 3 amino acid­long (SKG) and 11 amino acid­long peptides (EYVQTVKSSKG) in fusion to cell­penetrating peptide, named as A1(28­30) and A1(20­30) respectively, and found that A1(28­30) and A1(20­30) blocked the binding of ANXA1 with EphA2, targeted EphA2 degradation, and suppressed the growth of GC and CC cells in vitro and in mice. Our data demonstrated that ANXA1 was able to bind and stabilize EphA2 in GC and CC cells, and disruption of ANXA1­EphA2 interaction by the two ANXA1­derived peptides inhibited the growth of GC and CC cells by targeting EphA2 degradation, presenting a potential strategy for treating GC and CC with these peptides.


Asunto(s)
Anexina A1/farmacología , Neoplasias del Colon/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Receptor EphA2/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
11.
J Hematol Oncol ; 13(1): 114, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32811512

RESUMEN

Eph receptors and the corresponding Eph receptor-interacting (ephrin) ligands jointly constitute a critical cell signaling network that has multiple functions. The tyrosine kinase EphA2, which belongs to the family of Eph receptors, is highly produced in tumor tissues, while found at relatively low levels in most normal adult tissues, indicating its potential application in cancer treatment. After 30 years of investigation, a large amount of data regarding EphA2 functions have been compiled. Meanwhile, several compounds targeting EphA2 have been evaluated and tested in clinical studies, albeit with limited clinical success. The present review briefly describes the contribution of EphA2-ephrin A1 signaling axis to carcinogenesis. In addition, the roles of EphA2 in resistance to molecular-targeted agents were examined. In particular, we focused on EphA2's potential as a target for cancer treatment to provide insights into the application of EphA2 targeting in anticancer strategies. Overall, EphA2 represents a potential target for treating malignant tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Efrina-A2/antagonistas & inhibidores , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Vacunas contra el Cáncer , Ensayos Clínicos como Asunto , Terapia Combinada , Células Dendríticas/inmunología , Sistemas de Liberación de Medicamentos , Inducción Enzimática/efectos de los fármacos , Efrina-A1/fisiología , Efrina-A2/fisiología , Humanos , Inmunoconjugados/uso terapéutico , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoterapia Adoptiva , Ratones , Ratones Desnudos , Nanocápsulas , Proteínas de Neoplasias/fisiología , Neoplasias/enzimología , Neoplasias/terapia , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Receptor EphA2 , Proteínas Recombinantes de Fusión/metabolismo , Especificidad por Sustrato , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Res ; 80(20): 4386-4398, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32737118

RESUMEN

Overexpression of ANXA1 and EphA2 has been linked to various cancers and both proteins have attracted considerable attention for the development of new anticancer drugs. Here we report that ANXA1 competes with Cbl for binding EphA2 and increases its stability by inhibiting Cbl-mediated EphA2 ubiquitination and degradation in nasopharyngeal carcinoma (NPC). Binding of ANXA1 to EphA2 promoted NPC cell growth and metastasis in vitro and in vivo by elevating EphA2 levels and increasing activity of EphA2 oncogenic signaling (pS897-EphA2). Expression of ANXA1 and EphA2 was positively correlated and both were significantly higher in NPC tissues than in the normal nasopharyngeal epithelial tissues. Patients with high expression of both proteins presented poorer disease-free survival and overall survival relative to patients with high expression of one protein alone. Furthermore, amino acid residues 20-30aa and 28-30aa of the ANXA1 N-terminus bound EphA2. An 11 amino acid-long ANXA1-derived peptide (EYVQTVKSSKG) was developed on the basis of this N-terminal region, which disrupted the connection of ANXA1 with EphA2, successfully downregulating EphA2 expression and dramatically suppressing NPC cell oncogenicity in vitro and in mice. These findings suggest that ANXA1 promotes NPC growth and metastasis via binding and stabilization of EphA2 and present a strategy for targeting EphA2 degradation and treating NPC with a peptide. This therapeutic strategy may also be extended to other cancers with high expression of both proteins. SIGNIFICANCE: These findings show that EphA2 is a potential target for NPC therapeutics and an ANXA1-derived peptide suppresses NPC growth and metastasis. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/20/4386/F1.large.jpg.


Asunto(s)
Anexina A1/metabolismo , Efrina-A2/metabolismo , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/patología , Animales , Anexina A1/química , Anexina A1/genética , Sitios de Unión , Unión Competitiva , Línea Celular Tumoral , Efrina-A2/química , Efrina-A2/genética , Humanos , Masculino , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/mortalidad , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/mortalidad , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Receptor EphA2 , Ubiquitina/metabolismo
13.
Cell Death Dis ; 11(8): 709, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32848131

RESUMEN

EphA2 is an important oncogenic protein and emerging drug target, but the oncogenic role and mechanism of ligand-independent phosphorylation of EphA2 at tyrosine 772 (pY772-EphA2) is unclear. In this study, we established nasopharyngeal carcinoma (NPC) cell lines with stable expression of exogenous EphA2 and EphA2-Y772A (phosphorylation inactivation) using endogenous EphA2-knockdown cells, and observed that pY772A EphA2 was responsible for EphA2-promoting NPC cell proliferation and anchorage-independent and in vivo growth in mice. Mechanistically, EphA2-Y772A mediated EphA2-activating Shp2/Erk-1/2 signaling pathway in the NPC cells, and Gab1 (Grb2-associated binder 1) and Grb2 (growth factor receptor-bound protein 2) were involved in pY772-EphA2 activating this signaling pathway. Our results further showed that Shp2/Erk-1/2 signaling mediated pY772-EphA2-promoting NPC cell proliferation and anchorage-independent growth. Moreover, we observed that EphA2 tyrosine kinase inhibitor ALW-II-41-27 inhibited pY772-EphA2 and EphA2-Y772A decreased the inhibitory effect of ALW-II-41-27 on NPC cell proliferation. Collectively, our results demonstrate that pY772-EphA2 is responsible for EphA2-dependent NPC cell growth in vitro and in vivo by activating Shp2/Erk-1/2 signaling pathway, and is a pharmacologic target of ALW-II-41-27, suggesting that pY772-EphA2 can serve as a therapeutic target in NPC and perhaps in other cancers.


Asunto(s)
Efrina-A2/genética , Carcinoma Nasofaríngeo/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , China , Efrina-A2/metabolismo , Proteína Adaptadora GRB2/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Desnudos , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/patología , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Receptor EphA2/genética , Receptor EphA2/metabolismo , Transducción de Señal/genética , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell Death Dis ; 11(5): 322, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32376822

RESUMEN

HDAC7 plays a crucial role in cancers, and is the main drug target of several HDAC inhibitors. However, the role and mechanism of HDAC7 in nasopharyngeal carcinoma (NPC) are still unclear. In this study, we observed that HDAC7 was significantly upregulated in the NPC tissues relative to normal nasopharyngeal mucosa (NNM) tissues, HDAC7 expression levels were positively correlated with NPC progression and negatively correlated with patient prognosis, and HDAC7 knockdown dramatically inhibited the in vitro proliferation, migration, and invasion of NPC cells, and the growth of NPC xenografts in mice, indicating the HDAC7 promotes the oncogenicity of NPC. Mechanistically, HDAC7 promoted the in vitro proliferation, migration, and invasion of NPC cells by upregulating EphA2, in which miR-4465 mediated HDAC7-regulating EphA2, a direct target gene of miR-4465. We further showed that miR-4465 was significantly downregulated in the NPC tissues relative to NNM tissues, and inhibited the in vitro proliferation, migration, and invasion of NPC cells by targeting EphA2 expression. Moreover, we observed that the expressions of HDAC7, miR-4465, and EphA2 in NPC tissues were correlated. The results suggest that HDAC7 promotes the oncogenicity of NPC by downregulating miR-4465 and subsequently upregulating EphA2, highlighting HDAC7 as a potential therapeutic target for NPC.


Asunto(s)
Efrina-A2/metabolismo , Histona Desacetilasas/metabolismo , MicroARNs/genética , Animales , Apoptosis/genética , Carcinoma/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Efrina-A2/genética , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasas/genética , Humanos , Ratones , MicroARNs/metabolismo , Carcinoma Nasofaríngeo/genética , Neoplasias Nasofaríngeas/patología , Pronóstico , Receptor EphA2 , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Oncogene ; 39(21): 4183-4197, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32291412

RESUMEN

CD147, also known as extracellular matrix metalloproteinase inducer (EMMPRIN), is a transmembrane glycoprotein that is highly expressed in tumor cells, particularly melanoma cells, and plays critical roles in tumor cell metastasis through the regulation of matrix metalloprotease (MMP) expression. In this study, we identified Fyn as a novel interacting protein of CD147. Fyn is a member of the Src family of nonreceptor tyrosine kinases that regulates diverse physiological processes, such as T lymphocyte differentiation, through the TCR signaling pathway. Our findings demonstrated that Fyn directly phosphorylates CD147 at Y140 and Y183. Two phosphospecific antibodies against Y140 and Y183 were developed to validate the phosphorylation of CD147 by Fyn. Moreover, the CD147-FF (Y140F/Y183F) mutation impaired the interaction between CD147 and GnT-V, leading to decreased CD147 glycosylation and membrane recruitment. In addition, CD147-FF significantly blocked MMP-9 expression as well as cell migration. Moreover, we found that Fyn is overexpressed in clinical melanoma tissues as well as in melanoma cell lines. Knockdown of Fyn expression markedly attenuated the malignant phenotype of melanoma cells in vitro and in vivo through downregulation of CD147 phosphorylation, indicating that Fyn/CD147 is a potential target molecule in melanoma treatment. Finally, through virtual screening, we identified amodiaquine as a potential inhibitor targeting the Fyn/CD147 axis. Amodiaquine treatment dramatically inhibited the phosphorylation of CD147 by Fyn, thus attenuating melanoma cell growth and invasion in vitro and in vivo, suggesting that amodiaquine is a promising inhibitor for melanoma treatment.


Asunto(s)
Basigina/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Transducción de Señal , Sustitución de Aminoácidos , Basigina/genética , Línea Celular Tumoral , Glicosilación , Humanos , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/genética , Melanoma/genética , Melanoma/patología , Mutación Missense , Metástasis de la Neoplasia , Fosforilación/genética , Proteínas Proto-Oncogénicas c-fyn/genética
16.
Cancer Lett ; 444: 162-174, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30583071

RESUMEN

Our phosphoproteomics identified that phosphorylation of EphA2 at serine 897 (pS897-EphA2) was significantly upregulated in the high metastatic nasopharyngeal carcinoma (NPC) cells relative to non-metastatic NPC cells. However, the role and underlying mechanism of pS897-EphA2 in cancer metastasis and stem properties maintenance remain poorly understood. In this study, we established NPC cell lines with stable expression of exogenous EphA2 and EphA2-S897A using endogenous EphA2 knockdown cells, and observed that pS897-EphA2 maintained EphA2-dependent NPC cell in vitro migration and invasion, in vivo metastasis and cancer stem properties. Using phospho-kinase antibody array to identify signaling downstream of pS897-EphA2, we found that AKT/Stat3 signaling mediated pS897-EphA2-promoting NPC cell invasion, metastasis and stem properties, and Sox-2 and c-Myc were the effectors of pS897-EphA2. Immunohistochemistry showed that pS897-EphA2 was positively correlated with NPC metastasis and negatively correlated with patient overall survival. Moreover, ERK/RSK signaling controlled serum-induced pS897-EphA2 in NPC cells. Collectively, our results demonstrate that pS897-EphA2 is indispensable for EphA2-dependent NPC cell invasion, metastasis and stem properties by activating AKT/Stat3/Sox-2 and c-Myc signaling pathway, suggesting that pS897-EphA2 can serve as a therapeutic target in NPC and perhaps in other cancers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Nasofaríngeas/patología , Células Madre Neoplásicas/patología , Receptor EphA2/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Metástasis Linfática , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Invasividad Neoplásica , Células Madre Neoplásicas/metabolismo , Fosforilación , Pronóstico , Receptor EphA2/genética , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Int J Oncol ; 58(1): 133-134, 2019 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-33899924

RESUMEN

Following the publication of the above article, the authors have realized that one of the data panels featured in Fig. 5D was selected incorrectly. Specifically, the wrong image was selected for the A1 (28­30), HCT116 experiment. The authors have revisited their original sources to identify the correct data panel, and can confirm that the error arose unintentionally during the process of compiling the figure. The correct version of Fig. 5, featuring corrected data panel for Fig. 5D, is shown on the next page. The authors confirm that this error did not affect the conclusions reported in this study, and are grateful to the Editor of International Journal of Oncology for allowing them the opportunity to publish this corrigendum. Furthermore, the authors apologize to the readership of the Journal for any inconvenience caused. [the original article was published in International Journal of Oncology 57: 1203­1213, 2020; DOI: 10.3892/ijo.2020.5119].

18.
Cell Death Dis ; 9(12): 1154, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30459351

RESUMEN

Annexin A1 (ANXA1) is dysregulated in the various tumors. However, the role and mechanism of ANXA1 in the cancers are poorly understood. In this study, we first showed a clinically positive correlation between ANXA1 and autophagy-associated protein SQSTM1 expression in nasopharyngeal carcinoma (NPC) and ANXA1-regulating SQSTM1 expression through autophagy, and further demonstrated that ANXA1 inhibited BECN1 and ATG5-dependent autophagy in the NPC cells. Using phospho-kinase antibody array to identify signaling through which ANXA1 regulated NPC cell autophagy, we found that ANXA1-suppressed autophagy was associated with PI3K/AKT signaling activation. We also showed that ANXA1 expression was significantly increased in the NPCs with metastasis relative to NPCs without metastasis and positively correlated with lymphonode and distant metastasis; high ANXA1 expression in the NPC cells promoted in vitro tumor cell migration and invasion and in vivo metastasis. Lastly, we showed that inhibition of autophagy restored the ability of tumor cell migration and invasion, epithelial-mesenchymal transition (EMT)-like alterations and in vivo metastasis in the ANXA1 knockdown NPC cells with autophagy activation; ANXA1-suppresed autophagy induced EMT-like alterations possibly by inhibiting autophagy-mediated degradation of Snail. Our data suggest that ANXA1-suppressed autophagy promotes NPC cell migration, invasion and metastasis by activating PI3K/AKT signaling pathway, highlighting that the activation of autophagy may inhibit metastasis of NPC with high ANXA1 expression.


Asunto(s)
Anexina A1/genética , Autofagia/genética , Carcinoma Nasofaríngeo/genética , Proteína Sequestosoma-1/genética , Proteína 5 Relacionada con la Autofagia/genética , Beclina-1/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Humanos , Carcinoma Nasofaríngeo/patología , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Metástasis de la Neoplasia , Fosfatidilinositol 3-Quinasas/genética , Proteolisis , Proteínas Proto-Oncogénicas c-akt/genética , Factores de Transcripción de la Familia Snail/genética
19.
Cell Death Dis ; 9(12): 1148, 2018 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-30451832

RESUMEN

RACK1 is upregulated in the various types of human cancers, and considered to play a role in the development and progression of human cancer. However, the role and mechanism of RACK in the colon cancer are poorly understood. In this study, we detected RACK1 expression in 63 normal colonic mucosa, 60 colonic inflammatory polyps, 60 colonic adenomas, 180 colon adenocarcinomas, and 40 lymph node metastases by immunohistochemistry, and observed that RACK1 expression was progressively elevated in the carcinogenic process of human colonic epithelium, and RACK1 expressional levels were positively correlated with the malignant degree and lymph node metastasis of colon cancers, and negatively correlated with the patient survival. With a combination of loss-of-function and gain-of-function approaches, we observed that RACK1 promoted colon cancer cell proliferation, inhibited colon cancer cell apoptosis, and enhanced the anchorage-independent and xenograft growth of colon cancer cells. Moreover, we found that RACK1-induced autophagy of colon cancer cells; RACK1-induced autophagy promoted colon cancer cell proliferation and inhibited colon cancer cell apoptosis. Our data suggest that RACK1 acts as an oncogene in colon cancer, and RACK1-induced autophagy promotes proliferation and survival of colon cancer, highlighting the therapeutic potential of autophagy inhibitor in the colon cancer with high RACK1 expression.


Asunto(s)
Carcinogénesis/genética , Neoplasias del Colon/genética , Proteínas de Neoplasias/genética , Receptores de Cinasa C Activada/genética , Apoptosis/genética , Autofagia/genética , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias del Colon/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA