Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Acta Biochim Biophys Sin (Shanghai) ; 54(9): 1234-1243, 2022 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-36148951

RESUMEN

Colon cancer is a common malignant tumor. However, its pathogenesis still needs further study. In this study, we explored the role of nucleosome assembly protein 1-like 1 (NAP1L1) in colon cancer and its underlying mechanism. Based on analysis of The Cancer Genome Atlas data, we found that NAP1L1 is augmented in colorectal cancer, and the elevated NAP1L1 expression is associated with a poor prognosis in patients with colon cancer. Immunohistochemistry staining results showed that upregulated NAP1L1 protein level is an unfavorable factor that stimulates colon cancer progression. To further investigate the role of NAP1L1 in colon cancer, we established a colon cancer cell line with NAP1L1 knockdown, and found that repressing NAP1L1 expression in colon cancer cells markedly reduces cell proliferation in vivo and in vitro by MTT assay, colony formation, EdU incorporation, and subcutaneous tumorigenesis in nude mice. Furthermore, we found that NAP1L1 binds to HDGF, recruits DDX5, and induces ß-catenin/CCND1 signaling, which promotes colon cancer cell proliferation. Finally, transfection with HDGF or DDX5restores cell growth in NAP1L1-knockdown colon cancer cells by upregulating DDX5/ß-catenin/CCND1 signaling. Our study demonstrates that NAP1L1 functions as a potential oncogene that promotes colon cancer tumorigenesis by binding to HDGF, which stimulates DDX5/ß-catenin/CCND1 signaling.


Asunto(s)
Neoplasias del Colon , beta Catenina , Animales , Ratones , beta Catenina/genética , beta Catenina/metabolismo , Ratones Desnudos , Transducción de Señal , Proliferación Celular , Neoplasias del Colon/genética , Carcinogénesis/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica
2.
J Transl Med ; 19(1): 387, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34503528

RESUMEN

OBJECTIVE: The recurrence and metastasis of nasopharyngeal cancer (NPC) may be mainly attributed to the persistence of cancer stem cells (CSCs); however, the linkage mechanism has yet to be fully elucidated. METHODS: The levels of miR-4721, FOXA1, and Nanog expression in NPC were detected by in situ hybridization and immunohistochemistry. In vivo and in vitro metastasis assays confirmed miR-4721 promotes cell migration and invasion. Tumor spheroid formation assay, side population (SP) assay, and ALDEFLUOR assay verified miR-4721 regulates cancer stem cell-like properties. Luciferase reporter assay showed that miR-4721 directly regulates FOXA1 and FOXA1 effects the promoter activity of miR-4721 and Nanog. Chromatin immunoprecipitation (ChIP) analysis and electrophoresis mobility shift assay (EMSA) revealed that FOXA1 combined the promoter region of human miR-4721 and Nanog and the possible mechanism was also analyzed. RESULTS: In this study, a new mechanism of NPC tumorigenesis related to miR-4721 was verified. We found that miR-4721, FOXA1 and Nanog control their expressions through a negative feedback loop and then activate the downstream regulator of stem cell signaling to promote the enrichment and metastasis of NPC stem cells. CONCLUSION: These findings elucidate that the feedback loop of miR-4721/FOXA1/Nanog can regulate stemness and metastasis in NPC and may provide an experimental theoretical basis for metastasis and treatment resistance in NPC.


Asunto(s)
MicroARNs , Neoplasias Nasofaríngeas , Línea Celular Tumoral , Movimiento Celular/genética , Retroalimentación , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito/genética , Humanos , MicroARNs/genética , Proteína Homeótica Nanog/genética , Neoplasias Nasofaríngeas/genética , Recurrencia Local de Neoplasia , Células Madre Neoplásicas
3.
Int J Cancer ; 146(2): 496-509, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31125123

RESUMEN

The biological role of vacuolar protein sorting 33B (VPS33B) has not been examined in colorectal cancer (CRC). We report that VPS33B was downregulated in dextran sulfate sodium/azoxymethane (DSS/AOM) -induced CRC mice models and nicotine-treated CRC cells via the PI3K/AKT/c-Jun pathway. Reduced VPS33B is an unfavorable factor promoting poor prognosis in human CRC patients. VPS33B overexpression suppressed CRC proliferation, intrahepatic metastasis and chemoresistance of cisplatin (DDP) in vivo and in vitro through modulating the epidermal growth factor receptor (EGFR)/RAS/ERK/c-Myc/p53/miR-133a-3p feedback loop and the downstream cell cycle or EMT-related factors. Furthermore, NESG1 as a newly identified tumor suppressor interacted with VPS33B via colocalization in the cytoplasm, and it was stimulated by VPS33B through the downregulation of RAS/ERK/c-Jun-mediated transcription. NESG1 also activated VPS33B expression via the RAS/ERK/c-Jun pathway. Suppression of NESG1 increased cell growth, migration and invasion via the reversion of the VPS33B-modulating signal in VPS33B-overexpressed cells. Taken together, VPS33B as a tumor suppressor is easily dysregulated by chemical carcinogens and it interacts with NESG1 to modulate the EGFR/RAS/ERK/c-Myc/p53/miR-133a-3p feedback loop and thus suppress the malignant phenotype of CRC.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Genes Supresores de Tumor/efectos de los fármacos , Nicotina/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas de Transporte Vesicular/genética , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Proteínas del Citoesqueleto/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Células HT29 , Humanos , Ratones , Transducción de Señal/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética
4.
Shock ; 61(6): 855-860, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38320215

RESUMEN

ABSTRACT: Background: With the advancement of medicine and the development of technology, the limiting factors of aeromedical evacuation are gradually decreasing, and the scope of indications is expanding. However, the hypobaric and hypoxic environments experienced by critically ill patients in flight can cause lung injury, leading to inflammation and hypoxemia, which remains one of the few limiting factors for air medical evacuation. This study aimed to examine the mechanism of secondary lung injury in rat models of acute lung injury that simulate aeromedical evacuation. Methods: An acute lung injury model was induced in SD rats by the administration of lipopolysaccharide (LPS) followed by exposure to a simulated aeromedical evacuation environment (equivalent to 8,000 feet above sea level) or a normobaric normoxic environment for 4 h. The expression of hypoxia-inducible factor 1α (HIF-1α) was stabilized by pretreatment with dimethyloxalylglycine. The reactive oxygen species levels and the protein expression levels of HIF-1α, Bcl-2-interacting protein 3 (BNIP3), and NIX in lung tissue were measured. Results: Simulated aeromedical evacuation exacerbated pathological damage to lung tissue and increased the release of inflammatory cytokines in serum as well as the reactive oxygen species levels and the protein levels of HIF-1α, BNIP3, and NIX in lung tissue. Pretreatment with dimethyloxalylglycine resulted in increases in the protein expression of HIF-1α, BNIP3, and NIX. Conclusion: Simulated aeromedical evacuation leads to secondary lung injury through mitophagy.


Asunto(s)
Lesión Pulmonar Aguda , Subunidad alfa del Factor 1 Inducible por Hipoxia , Proteínas de la Membrana , Mitofagia , Ratas Sprague-Dawley , Animales , Lesión Pulmonar Aguda/metabolismo , Ratas , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ambulancias Aéreas , Proteínas Mitocondriales/metabolismo , Modelos Animales de Enfermedad
5.
Int J Biol Sci ; 20(12): 4618-4634, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39309424

RESUMEN

Some colorectal cancer patients have experienced normal epithelial transformation into inflammatory and tumor states, but the molecular basis still needs to be further determined. The expression levels of SIX4 are gradually increased in dextran sodium sulfate (DSS) and azoxymethane (AOM)/DSS-induced colonic epithelial inflammation and tumors, respectively, in mice. Targeting SIX4 alleviates intestinal inflammation occurrence and reduces adenoma formation in mice. Clinical sample assays indicated that SIX4 is upregulated in inflammatory bowel disease (IBD) and colorectal cancer (CRC) tissues compared to normal colorectal tissues. In a subsequent study, we found that SIX4, transcriptionally activated by the proinflammatory IL-6/STAT3 signal, binds to c-Jun to transcribe IL-6, thus forming a positive IL-6/STAT3/SIX4/c-Jun feedback loop, which further induces intestinal inflammation occurrence. In addition, elevated SIX4 also induces the expression of DeltaNp63, rather than wild-type p63, by binding to its promoter and thus facilitates the activation of tumor stemness signals, which ultimately leads to the formation of colorectal cancer. Our study first observes that activated SIX4 in inflammation induction drives the transformation of colorectal epithelium into inflammation and tumor, which demonstrates SIX4 as a significant therapeutic target in IBD and colitis-associated colorectal cancer (CAC) and CRC pathogenesis.


Asunto(s)
Neoplasias Colorrectales , Inflamación , Transducción de Señal , Animales , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Ratones , Inflamación/metabolismo , Humanos , Interleucina-6/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Sulfato de Dextran , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Enfermedades Inflamatorias del Intestino/inducido químicamente , Ratones Endogámicos C57BL , Mucosa Intestinal/metabolismo , Azoximetano
6.
J Cancer ; 12(22): 6600-6609, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34659550

RESUMEN

Lung cancer is a serious threat to human health due to its high morbidity and mortality. microRNAs (miRNAs) are involved in the tumorigenesis and progression of lung cancer. In this study, we elucidated the role of miRNA-4507 (miR-4507) in the pathogenesis of non-small-cell lung cancer (NSCLC). miR-4507 is found to be upregulated in NSCLC cells (A549, H460). MTT, 5-ethynyl-2'-deoxyuridine (EdU), wound healing, and transwell assays were performed to evaluate NSCLC cell proliferation and migration. The results demonstrated that miR-4507 inhibition significantly decrease the proliferation and migration of NSCLC cells. Subsequently, a luciferase activity assay was conducted to verify the regulation of the predicted gene target of miR-4507, namely, TP53. Mechanism experiments show that miR-4507 activates the PI3K/AKT signal. Further, we co-transfected miR-4507 mimics and TP53 plasmids and found that TP53 overexpression could recover the effects of miR-4507 mimics on proliferation, migration, and the PI3K/AKT signal activation. These results suggested that miR-4507 targets TP53 to facilitate the proliferation and migration of lung cancer cells through PI3K/AKT signal and that miR-4507 could serve as a potential target for NSCLC treatment.

7.
Theranostics ; 11(16): 8112-8128, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34335983

RESUMEN

The coiled-coil domain containing protein members have been well documented for their roles in many diseases including cancers. However, the function of the coiled-coil domain containing 65 (CCDC65) remains unknown in tumorigenesis including gastric cancer. Methods: CCDC65 expression and its correlation with clinical features and prognosis of gastric cancer were analyzed in tissue. The biological role and molecular basis of CCDC65 were performed via in vitro and in vivo assays and a various of experimental methods including co-immunoprecipitation (Co-IP), GST-pull down and ubiquitination analysis et al. Finally, whether metformin affects the pathogenesis of gastric cancer by regulating CCDC65 and its-mediated signaling was investigated. Results: Here, we found that downregulated CCDC65 level was showed as an unfavourable factor in gastric cancer patients. Subsequently, CCDC65 or its domain (a.a. 130-484) was identified as a significant suppressor in GC growth and metastasis in vitro and in vivo. Molecular basis showed that CCDC65 bound to ENO1, an oncogenic factor has been widely reported to promote the tumor pathogenesis, by its domain (a.a. 130-484) and further promoted ubiquitylation and degradation of ENO1 by recruiting E3 ubiquitin ligase FBXW7. The downregulated ENO1 decreased the binding with AKT1 and further inactivated AKT1, which led to the loss of cell proliferation and EMT signal. Finally, we observed that metformin, a new anti-cancer drug, can significantly induce CCDC65 to suppress ENO1-AKT1 complex-mediated cell proliferation and EMT signals and finally suppresses the malignant phenotypes of gastric cancer cells. Conclusion: These results firstly highlight a critical role of CCDC65 in suppressing ENO1-AKT1 pathway to reduce the progression of gastric cancer and reveals a new molecular mechanism for metformin in suppressing gastric cancer. Our present study provides a new insight into the mechanism and therapy for gastric cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas de Unión al ADN/metabolismo , Glicoproteínas/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Gástricas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , China , Femenino , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Genes Supresores de Tumor/fisiología , Glicoproteínas/genética , Humanos , Masculino , Metformina/metabolismo , Metformina/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Oncogenes , Pronóstico , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
8.
Mol Ther Nucleic Acids ; 22: 557-571, 2020 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-33230457

RESUMEN

Nasopharyngeal carcinoma (NPC) is prevalent in East and Southeast Asia. In a previous study, Epstein-Barr virus (EBV)-miR-BART22 induces tumor metastasis and stemness and is significantly involved in NPC progression. In the present study, we observed that miR-4721 is induced by EBV-miR-BART22 through phosphatidylinositol 3-kinase (PI3K)/AKT/c-JUN/Sp1 signaling to promote its transcription. In a subsequent study, we observed that miR-4721 serves as a potential oncogenic factor promoting NPC cell cycle progression and cell proliferation in vitro and in vivo. Mechanism analysis indicated that miR-4721 directly targetes GSK3ß and reduces its expression, which therefore elevates ß-catenin intra-nuclear aggregation and activates its downstream cell cycle factors, including CCND1 and c-MYC. In clinical samples, miR-4721 and GSK3ß are respectively observed to be upregulated and downregulated in NPC progression. Elevated expression of miR-4721 is positively associated with clinical progression and poor prognosis. Our study first demonstrated that miR-4721 as an oncogene is induced by EBV-miR-BART22 via modulating PI3K/AKT/c-JUN/Sp1 signaling to target GSK3ß, which thus activates the WNT/ß-catenin-stimulated cell cycle signal and enhances the tumorigenic capacity in NPC. miR-4721 may be a potential biomarker or therapeutic target in NPC treatment in the future.

9.
EBioMedicine ; 48: 386-404, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31594754

RESUMEN

BACKGROUND: Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus (EBV)-related tumor. The role of EBV-encoding miR-BART22 is still unclear in NPC. This study aimed to identify the detailed mechanisms by which EBV-miR-BART22 functions as a tumor-promoting factor and evaluate the action of cinobufotalin in treating EBV-miR-BART22-overexpressing NPC cells. METHODS: Using real-time PCR, western blotting, immunohistochemistry, and In situ hybridization, we detected the expression of miR-BART22 and MAP2K4 in tissues and cells, as well as evaluated their clinical relevance in NPC patients. The effects of miR-BART22 on cell metastasis, stemness and DDP chemoresistance were examined by sphere formation assay, side population analysis, transwell, boyden, in vivo xenograft tumor mouse model et al. Western blotting, immunofluorescence staining, luciferase reporter assay, ChIP, EMSA and Co-IP assay et al. were performed to explore the detailed molecular mechanism of EBV-miR-BART22 in NPC. Finally, we estimated the effects and molecular basis of Cinobufotalin on EBV-miR-BART22-overexpressing NPC cells in vitro and in vivo assays. FINDINGS: We observed that EBV-miR-BART22 not only promoted tumor stemness and metastasis, but also enhanced the resistance to Cisplatin (DDP) in vitro and in vivo. Mechanistic analysis indicated that EBV-miR-BART22 directly targeted the MAP2K4 and upregulated non-muscle myosin heavy chain IIA (MYH9) expression by PI3K/AKT/c-Jun-induced transcription. Further, MYH9 interacted with glycogen synthase 3ß(GSK3ß) protein and induced its ubiquitin degradation by activating PI3K/AKT/c-Jun-induced ubiquitin transcription and the latter combined with increased TRAF6 E3 ligase, which further bound to GSK3ß protein. Reductions in the GSK3ß protein thus promoted ß-catenin expression and nuclear translocation, which induced tumor stemness and the epithelial-to-mesenchymal transition (EMT) signals. Furthermore, we observed that cinobufotalin, a new chemically synthesized compound, significantly suppressed EBV-miR-BART22-induced DDP chemoresistance by upregulating MAP2K4 to suppress MYH9/GSK3ß/ß-catenin and its downstream tumor stemness and EMT signals in NPC. Finally, clinical data revealed that increased miR-BART22 and reduced MAP2K4 expression caused the poor prognoses of NPC patients. INTERPRETATION: Our study provides a novel mechanism that cinobufotalin reversed the DDP chemoresistance and EMT induced by EBV-miR-BART22 in NPC.


Asunto(s)
Bufanólidos/farmacología , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Herpesvirus Humano 4/genética , MicroARNs/genética , ARN Viral , Línea Celular Tumoral , Glucógeno Sintasa/metabolismo , Humanos , MAP Quinasa Quinasa 4/genética , Cadenas Pesadas de Miosina/metabolismo , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/metabolismo , Transducción de Señal/efectos de los fármacos , Células Madre/metabolismo , beta Catenina/metabolismo
10.
Artículo en Inglés | MEDLINE | ID: mdl-31754475

RESUMEN

In this study, we present novel molecular mechanisms by which FOXO1 functions as a tumor suppressor to prevent the pathogenesis of nasopharyngeal carcinoma (NPC). First, we observed that FOXO1 not only controlled tumor stemness and metastasis, but also sensitized NPC cells to cisplatin (DDP) in vitro and in vivo. Mechanistic studies demonstrated that FOXO1-induced miR-200b expression through the GSK3ß/ß-catenin/TCF4 network-mediated stimulation of ZEB1, which reduced tumor stemness and the epithelial-mesenchymal transition (EMT) signal. Furthermore, we observed FOXO1 interaction with MYH9 and suppression of MYH9 expression by modulating the PI3K/AKT/c-Myc/P53/miR-133a-3p pathway. Decreased MYH9 expression not only reduced its interactions with GSK3ß, but also attenuated TRAF6 expression, which then decreased the ubiquitin-mediated degradation of GSK3ß protein. Increased GSK3ß expression stimulated the ß-catenin/TCF4/ZEB1/miR-200b network, which increased the downstream tumor stemness and EMT signals. Subsequently, we observed that chemically synthesized cinobufotalin (CB) strongly increased FOXO1-induced DDP chemosensitivity by reducing MYH9 expression, and the reduction in MYH9 modulated GSK3ß/ß-catenin and its downstream tumor stemness and EMT signal in NPC. In clinical samples, the combination of low FOXO1 expression and high MYH9 expression indicated the worst overall survival rates. Our studies demonstrated that CB potently induced FOXO1-mediated DDP sensitivity by antagonizing its binding partner MYH9 to modulate tumor stemness in NPC.

11.
J Mech Behav Biomed Mater ; 72: 182-191, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28499166

RESUMEN

Binary Zn-Cu alloy system is developed as potential biodegradable materials for cardiovascular implant application. The microstructure, tensile properties, in vitro corrosion behavior, cytotoxicity and antibacterial property of as-extruded Zn-xCu (x=1, 2, 3, and 4wt%) alloys are investigated systematically. It shows that as Cu content increases more CuZn5 phase precipitates. After extrusion, the CuZn5 phases are broken and the grains of Zn-xCu alloys are refined. Tensile test shows that Cu addition could significantly improve the mechanical properties of Zn-xCu alloys. Particularly, the elongation of the Zn-4Cu reaches 50.6±2.8%, which could facilitate the micro-tubes processing for stent fabrication. The micro-tubes of 3mm in outer diameter and 0.2mm in thickness as well as vascular stents have been fabricated successfully using the Zn-Cu binary alloy. The degradation rates of Zn-xCu alloys in c-SBF solution are quite low, which vary from 22.1±4.7 to 33.0±1.0µmyear-1. With increasing Cu concentration, the corrosion rates of the Zn-xCu alloys generally exhibit a little increase compared with pure Zn, which show no significant difference among Zn-xCu alloys. In vitro test shows that Zn-xCu alloys exhibit acceptable cytotoxicity to human endothelial cells and the antibacterial property (S. aureus) is perfect when Cu concentration is higher than 2wt%. Therefore, the newly developed Zn-xCu binary alloys could be promising candidates for biodegradable cardiovascular implant application due to their excellent combination of strength and ductility, low degradation rates, acceptable cytotoxicity and good antibacterial property.


Asunto(s)
Implantes Absorbibles , Aleaciones/química , Materiales Biocompatibles/química , Células Endoteliales/citología , Stents , Línea Celular , Cobre/química , Corrosión , Humanos , Ensayo de Materiales , Staphylococcus aureus , Difracción de Rayos X , Zinc/química
12.
Mater Sci Eng C Mater Biol Appl ; 69: 407-13, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27612729

RESUMEN

Zn-based alloys have been viewed as new potential materials for biodegradable implants, such as cardiovascular stents, mainly in consideration of their lower corrosion rate when compared with that of Mg alloys. In this study we developed a new Zinc-4wt.%Copper (Zn-4Cu) alloy as a biodegradable material. Hot extrusion was applied to Zn-4Cu to refine the microstructure and consequently improve its mechanical properties and corrosion resistance. After extrusion, dendritic CuZn5 phases were broken and distributed along the extrusion direction. The grains were refined obviously due to dynamical recrystallization. The yield strength (YS), ultimate tensile strength (UTS) and elongation of the as-extruded alloy are 250±10MPa, 270±10MPa and 51±2%, respectively. The corrosion rate of the as-extruded alloy in Hank's solution is about 9.41(±1.34)µmyear(-1). In vitro evaluation shows that Zn-4Cu presents acceptable toxicity to human endothelial cells, and could effectively inhibit bacteria adhesion and biofilm formation. The present study indicates that the as-extruded Zn-4Cu alloy exhibits excellent strength and ductility, uniform and slow degradation, good biocompatibility and significant antibacterial effect, which make it an excellent candidate material for biodegradable implants, especially for cardiovascular stents application.


Asunto(s)
Aleaciones/química , Materiales Biocompatibles/química , Cobre/química , Stents , Zinc/química , Implantes Absorbibles , Aleaciones/farmacología , Materiales Biocompatibles/metabolismo , Materiales Biocompatibles/farmacología , Biopelículas/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Corrosión , Humanos , Staphylococcus aureus/fisiología , Resistencia a la Tracción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA