Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Cell Death Dis ; 14(12): 811, 2023 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-38071340

RESUMEN

Pancreatic cancer is highly lethal, of which 90% is pancreatic ductal adenocarcinoma (PDAC), with a 5-year survival rate of less than 12%, lacking effective treatment options and late diagnosis. Furthermore, the tumors show an intense resistance to cytotoxic chemotherapies. As autophagy is elevated in PDAC, targeting the autophagic pathway is regarded as a promising strategy for cancer treatment. Immunofluorescence and transmission electron microscopy were utilized to assess the autophagic flux. Label-free quantitative phosphoproteomics was used to figure out critically altered tyrosine phosphorylation of the proteins. Tumor-bearing mice were used to validate that SH2 TrM-(Arg)9 restrained the growth of tumor cells. SH2 TrM-(Arg)9 inhibited collagen-induced autophagy via blocking the DDR1/PYK2/ERK signaling cascades. SH2 TrM-(Arg)9 improved the sensitivity of PANC-1/GEM cells to gemcitabine (GEM). Inhibition of autophagy by SH2 TrM-(Arg)9 may synergized with chemotherapy and robusted tumor suppression in pancreatic cancer xenografts. SH2 TrM-(Arg)9 could enter into PDAC cells and blockade autophagy through inhibiting DDR1/PYK2/ERK signaling and may be a new treatment strategy for targeted therapy of PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Animales , Ratones , Quinasa 2 de Adhesión Focal/metabolismo , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/patología , Transducción de Señal , Autofagia , Línea Celular Tumoral , Receptor con Dominio Discoidina 1/metabolismo
2.
Theranostics ; 12(10): 4513-4535, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35832075

RESUMEN

Background: Idiopathic pulmonary fibrosis (IPF) is a progressive and irreversible fibrotic disease with high mortality. Currently, pirfenidone and nintedanib are the only approved drugs for IPF by the U.S. Food and Drug Administration (FDA), but their efficacy is limited. The activation of multiple phosphotyrosine (pY) mediated signaling pathways underlying the pathological mechanism of IPF has been explored. A Src homology-2 (SH2) superbinder, which contains mutations of three amino acids (AAs) of natural SH2 domain has been shown to be able to block phosphotyrosine (pY) pathway. Therefore, we aimed to introduce SH2 superbinder into the treatment of IPF. Methods: We analyzed the database of IPF patients and examined pY levels in lung tissues from IPF patients. In primary lung fibroblasts obtained from IPF patient as well as bleomycin (BLM) treated mice, the cell proliferation, migration and differentiation associated with pY were investigated and the anti-fibrotic effect of SH2 superbinder was also tested. In vivo, we further verified the safety and effectiveness of SH2 superbinder in multiple BLM mice models. We also compared the anti-fibrotic effect and side-effect of SH2 superbinder and nintedanib in vivo. Results: The data showed that the cytokines and growth factors pathways which directly correlated to pY levels were significantly enriched in IPF. High pY levels were found to induce abnormal proliferation, migration and differentiation of lung fibroblasts. SH2 superbinder blocked pY-mediated signaling pathways and suppress pulmonary fibrosis by targeting high pY levels in fibroblasts. SH2 superbinder had better therapeutic effect and less side-effect compare to nintedanib in vivo. Conclusions: SH2 superbinder had significant anti-fibrotic effects both in vitro and in vivo, which could be used as a promising therapy for IPF.


Asunto(s)
Fibrosis Pulmonar Idiopática , Animales , Bleomicina/farmacología , Proliferación Celular , Fibroblastos/metabolismo , Fibrosis , Fibrosis Pulmonar Idiopática/metabolismo , Ratones , Fosfotirosina/química , Fosfotirosina/metabolismo , Fosfotirosina/farmacología
3.
Clin Transl Med ; 11(3): e337, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33783993

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) exhibits the poorest prognosis of all solid tumors with a 5-year survival rate of less than 10% and a median survival of 6 months after diagnosis. Numerous targeted agents have been developed and evaluated to improve the survival benefit in patients with PDAC. Unfortunately, most agents have been proven futile mainly owing to the dense stroma and the sophisticated signaling pathways of PDAC. Here, we show the potent effectiveness of Aptamer-SH2 superbinder-(Arg)9 conjugate on the treatment of PDAC. In this conjugate, DNA aptamer selected against PDAC cell line confers the function of specifically recognizing and binding to the PDAC cells and activated pancreatic stellate cells (PSCs) in stroma; cell penetrating peptide (Arg)9 facilitates the intracellular delivery of fused proteins; SH2 superbinder conducts the drastic blockade of multiple phosphotyrosines (pY)-based signaling pathways in tumor cells. METHODS: PDAC-associated pY were reanalyzed by bioinformatics screen. XQ-2d and SH2 superbinder-(Arg)9 were crosslinked with BMH to form XQ-2d-SH2 CM-(Arg)9 conjugate. Immunofluorescence was utilized to assess the potency of the conjugate entering cells. MTT and wound healing assays were performed to evaluate the proliferation or migration of PANC-1 and BxPC-3 cells, respectively. Western blot and Pulldown assays revealed that conjugate influenced several pY-based signaling pathways. Tumor-bearing mice were used to validate XQ-2d-SH2 CM-(Arg)9, which restrained the growth and metastasis of cancer cells. RESULTS: XQ-2d-His-SH2 CM-(Arg)9 conjugate restrained proliferation, invasion, and metastasis of PDAC cells with potent efficacy via blocking the activity of several pY-related signaling cascades. XQ-2d-His-SH2 CM-(Arg)9 could eliminate the dense stroma of PDAC and then arrive at tumor tissues. CONCLUSIONS: XQ-2d-SH2 CM-(Arg)9 conjugate may efficiently destroy the pancreatic stroma and show potent antitumor efficacy with minimal toxic effect by regulating tumor cell proliferation and metastasis in vitro and in vivo, which makes it to be a promising targeted therapy of PDAC.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Aptámeros de Nucleótidos/uso terapéutico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Desnudos , Transducción de Señal/efectos de los fármacos
4.
J Exp Clin Cancer Res ; 38(1): 2, 2019 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-30606241

RESUMEN

BACKGROUND: Glioblastomas multiforme (GBM) is the most devastating primary intracranial malignancy lacking effective clinical treatments. Notch2 has been established to be a prognostic marker and probably involved in GBM malignant progression. N-acetylcysteine (NAC), a precursor of intracellular glutathione (GSH), has been widely implicated in prevention and therapy of several cancers. However, the role of NAC in GBM remains unclear and the property of NAC independent of its antioxidation is largely unknown. METHODS: The mRNA and protein levels of Notch family and other related factors were detected by RT-PCR and western blot, respectively. In addition, intracellular reactive oxygen species (ROS) was measured by flow cytometry-based DCFH-DA. Moreover, cell viability was assessed by CCK8 and cell cycle was analyzed by flow cytometry-based PI staining. The level of apoptosis was checked by flow cytometry-based Annexin V/PI. Cell migration and invasion were evaluated by wound healing and transwell invasion assays. At last, U87 Xenograft model was established to confirm whether NAC could restrain the growth of tumor. RESULTS: Our data showed that NAC could decrease the protein level of Notch2. Meanwhile, NAC had a decreasing effect on the mRNA and protein levels of its downstream targets Hes1 and Hey1. These effects caused by NAC were independent of cellular GSH and ROS levels. The mechanism of NAC-mediated Notch2 reduction was elucidated by promoting Notch2 degradation through Itch-dependent lysosome pathway. Furthermore, NAC could prevent proliferation, migration, and invasion and might induce apoptosis in GBM cells via targeting Notch2. Significantly, NAC could suppress the growth of tumor in vivo. CONCLUSIONS: NAC could facilitate Notch2 degradation through lysosomal pathway in an antioxidant-independent manner, thus attenuating Notch2 malignant signaling in GBM cells. The remarkable ability of NAC to inhibit cancer cell proliferation and tumor growth may implicate a novel application of NAC on GBM therapy.


Asunto(s)
Acetilcisteína/uso terapéutico , Antivirales/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Receptor Notch2/genética , Receptor Notch2/metabolismo , Acetilcisteína/farmacología , Animales , Antivirales/farmacología , Modelos Animales de Enfermedad , Glioblastoma/patología , Humanos , Ratones , Transducción de Señal , Transfección
5.
Anal Chim Acta ; 1032: 138-146, 2018 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-30143211

RESUMEN

Tyrosine phosphorylation, as a hallmark in cellular signal transduction, is important for a diverse array of cellular processes, such as proliferation, metabolism, motility, and survival. Aberrant tyrosine phosphorylation plays a causal role in many diseases, especially the cancer. Detecting protein phosphorylation status in the cancer cells or tissues is vital for assessing the pathological phase, discovering the cancer biomarkers, and identifying the drug targets. However, the common biochemical detection methods remain through anti-pTyr antibodies, which are known to have limited sensitivity, poor reproducibility and high cost. Recent studies have proved that superbinder SH2 domain is a good replacement of anti-pTyr antibodies for the specific enrichment of pTyr peptides in phosphoproteomics analysis. In this work, we exploited a series of affinity reagents based on superbinder SH2 derived from Src protein for detecting the pTyr-containing proteins to replace anti-pY antibodies in immunoblotting and immunofluorescence techniques. The excellent performance of HRP-sSH2 and EGFP-sSH2 was verified by the analysis of several different tumor cell samples and was compared with most commonly used commercial antibodies. EGFP-sSH2-(Arg)9 might be applied as the probe for direct fluorescence imaging in live cells via efficiently penetrating cell membranes and specifically binding with pTyr proteins. In summary, we have developed three novel, convenient, sensitive, and cost-effective affinity reagents that would have wide applications in protein tyrosine phosphorylation analysis for the tumor research and clinical diagnosis.


Asunto(s)
Arginina/química , Proteínas Fluorescentes Verdes/química , Neoplasias/química , Tirosina/metabolismo , Familia-src Quinasas/química , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Microscopía Fluorescente , Neoplasias/patología , Imagen Óptica , Fosforilación , Tirosina/química , Dominios Homologos src , Familia-src Quinasas/metabolismo
6.
J Exp Clin Cancer Res ; 37(1): 138, 2018 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-29976230

RESUMEN

BACKGROUND: Melanoma is a malignant tumor with high misdiagnosis rate and poor prognosis. The bio-targeted therapy is a prevailing method in the treatment of melanoma; however, the accompanying drug resistance is inevitable. SH2 superbinder, a triple-mutant of the Src Homology 2 (SH2) domain, shows potent antitumor ability by replacing natural SH2-containing proteins and blocking multiple pY-based signaling pathways. Polyarginine (Arg)9, a powerful vector for intracellular delivery of large molecules, could transport therapeutic agents across cell membrane. The purpose of this study is to construct (Arg)9-SH2 superbinder and investigate its effects on melanoma cells, expecting to provide potential new approaches for anti-cancer therapy and overcoming the unavoidable drug resistance of single-targeted antitumor agents. METHODS: (Arg)9 and SH2 superbinder were fused to form (Arg)9-SH2 superbinder via genetic engineering. Pull down assay was performed to identify that (Arg)9-SH2 superbinder could capture a wide variety of pY proteins. Immunofluorescence was used to detect the efficiency of (Arg)9-SH2 superbinder entering cells. The proliferation ability was assessed by MTT and colony formation assay. In addition, wound healing and transwell assay were performed to evaluate migration of B16F10, A375 and A375/DDP cells. Moreover, apoptosis caused by (Arg)9-SH2 superbinder was analyzed by flow cytometry-based Annexin V/PI. Furthermore, western blot revealed that (Arg)9-SH2 superbinder influenced some pY-related signaling pathways. Finally, B16F10 xenograft model was established to confirm whether (Arg)9-SH2 superbinder could restrain the growth of tumor. RESULTS: Our data showed that (Arg)9-SH2 superbinder had the ability to enter melanoma cells effectively and displayed strong affinities for various pY proteins. Furthermore, (Arg)9-SH2 superbinder could repress proliferation, migration and induce apoptosis of melanoma cells by regulating PI3K/AKT, MAPK/ERK and JAK/STAT signaling pathways. Importantly, (Arg)9-SH2 superbinder could significantly inhibit the growth of tumor in mice. CONCLUSIONS: (Arg)9-SH2 superbinder exhibited high affinities for pY proteins, which showed effective anticancer ability by replacing SH2-containing proteins and blocking diverse pY-based pathways. The remarkable ability of (Arg)9-SH2 superbinder to inhibit cancer cell proliferation and tumor growth might open the door to explore the SH2 superbinder as a therapeutic agent for cancer treatment.


Asunto(s)
Melanoma/metabolismo , Fosfotirosina/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Péptidos de Penetración Celular/farmacología , Modelos Animales de Enfermedad , Humanos , Melanoma/tratamiento farmacológico , Melanoma/patología , Melanoma Experimental , Ratones , Proteínas Recombinantes de Fusión/genética
7.
Nat Commun ; 6: 7721, 2015 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-26166433

RESUMEN

Although cell migration plays a central role in development and disease, the underlying molecular mechanism is not fully understood. Here we report that a phosphorylation-mediated molecular switch comprising deleted in liver cancer 1 (DLC1), tensin-3 (TNS3), phosphatase and tensin homologue (PTEN) and phosphoinositide-3-kinase (PI3K) controls the spatiotemporal activation of the small GTPases, Rac1 and RhoA, thereby initiating directional cell migration induced by growth factors. On epidermal growth factor (EGF) or platelet-derived growth factor (PDGF) stimulation, TNS3 and PTEN are phosphorylated at specific Thr residues, which trigger the rearrangement of the TNS3-DLC1 and PTEN-PI3K complexes into the TNS3-PI3K and PTEN-DLC1 complexes. Subsequently, the TNS3-PI3K complex translocates to the leading edge of a migrating cell to promote Rac1 activation, whereas PTEN-DLC1 translocates to the posterior for localized RhoA activation. Our work identifies a core signalling mechanism by which an external motility stimulus is coupled to the spatiotemporal activation of Rac1 and RhoA to drive directional cell migration.


Asunto(s)
Movimiento Celular/genética , Proteínas Activadoras de GTPasa/genética , Proteínas de Microfilamentos/genética , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasa/genética , Proteínas Supresoras de Tumor/genética , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Western Blotting , Línea Celular Tumoral , Factor de Crecimiento Epidérmico , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Inmunoprecipitación , Células MCF-7 , Microscopía Confocal , Microscopía Fluorescente , Mutagénesis Sitio-Dirigida , Fosforilación , Factor de Crecimiento Derivado de Plaquetas , Transducción de Señal , Tensinas , Imagen de Lapso de Tiempo , Proteínas de Unión al GTP rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA