Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Small ; : e2312288, 2024 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-38431966

RESUMO

The development of capable of simultaneously modulating the sluggish electrochemical kinetics, shuttle effect, and lithium dendrite growth is a promising strategy for the commercialization of lithium-sulfur batteries. Consequently, an elaborate preparation method is employed to create a host material consisting of multi-channel carbon microspheres (MCM) containing highly dispersed heterostructure Fe3 O4 -FeTe nanoparticles. The Fe3 O4 -FeTe@MCM exhibits a spontaneous built-in electric field (BIEF) and possesses both lithophilic and sulfophilic sites, rendering it an appropriate host material for both positive and negative electrodes. Experimental and theoretical results reveal that the existence of spontaneous BIEF leads to interfacial charge redistribution, resulting in moderate polysulfide adsorption which facilitates the transfer of polysulfides and diffusion of electrons at heterogeneous interfaces. Furthermore, the reduced conversion energy barriers enhanced the catalytic activity of Fe3 O4 -FeTe@MCM for expediting the bidirectional sulfur conversion. Moreover, regulated Li deposition behavior is realized because of its high conductivity and remarkable lithiophilicity. Consequently, the battery exhibited long-term stability for 500 cycles with 0.06% capacity decay per cycle at 5 C, and a large areal capacity of 7.3 mAh cm-2 (sulfur loading: 9.73 mg cm-2 ) at 0.1 C. This study provides a novel strategy for the rational fabrication of heterostructure hosts for practical Li-S batteries.

2.
Clin Pharmacol Ther ; 115(5): 1085-1091, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38159264

RESUMO

Episil is a bioadhesive barrier-forming liquid gel that can relieve mucositis caused by radiotherapy (RT) and effectively relieve pain. The purpose of this trial is to compare the efficacy and safety of Episil in improving acute radiation dermatitis (ARD) in patients with breast cancer. This study included patients who met the criteria for postoperative RT for breast cancer. The primary end point was the grade of RD during treatment. A total of 102 patients were included in this study. The patients were grouped in a 2:1 ratio using the randomized number table method: 67 patients received Episil combined with conventional skin care (the Episil group), whereas the remaining 35 patients served as the control group and received conventional skin care only (the control group). According to the grading criteria of the Radiation Therapy Oncology Group (RTOG), the skin reaction rate and severity were significantly better in the Episil group than the control group (24.62%, 72.31%, 3.08, 0, 0 vs. 0, 85.71%, 14.29%, 0, 0, 0) across grades 0 to 4 (P < 0.05). The itchiness score exhibited s significant reduction in the Episil group as compared with the control group (P < 0.05). The results of the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-C30) showed that the overall health (z = -5.855, P < 0.001) and overall quality of life (z = -6.583, P < 0.001) were better in the Episil group than the control group after RT. Overall, in patients with breast cancer receiving RT, the topical application of Episil may significantly reduce the grading of ARD, alleviate patient symptoms, and improve the patient's overall quality of life.


Assuntos
Neoplasias da Mama , Radiodermite , Elastômeros de Silicone , Humanos , Feminino , Neoplasias da Mama/radioterapia , Neoplasias da Mama/cirurgia , Qualidade de Vida , Radiodermite/tratamento farmacológico , Dor
3.
Nano Lett ; 23(17): 7805-7814, 2023 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-37651260

RESUMO

Lithium (Li) metal is regarded as the "Holy Grail" of anodes for high-energy rechargeable lithium batteries by virtue of its ultrahigh theoretical specific capacity and the lowest redox potential. However, the Li dendrite impedes the practical application of Li metal anodes. Herein, lithiophilic three-dimensional Cu-CuSn porous framework (3D Cu-CuSn) was fabricated by a vapor phase dealloying strategy via the difference in saturated vapor pressure between different metals and the Kirkendall effect. CuSn alloy sites were converted into LiSn alloy sites through the molten Li infusion method, and composite Li metal anodes (3D Cu-LiSn-Li) are achieved. Alloyed tin, as the bridge between the porous copper substrate and metallic Li, plays a critical role in optimizing Li nucleation and enhancing the fast lithium migration kinetics. This work demonstrates that lithiophilic binary copper alloys are an effective way to achieve room-temperature high rate performance and satisfied low-temperature cycling stability for Li metal batteries.

4.
Small ; 19(5): e2205525, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36433827

RESUMO

Lithium-sulfur battery suffers from sluggish kinetics at low temperatures, resulting in serious polarization and reduced capacity. Here, this work introduces medium-entropy-alloy FeCoNi as catalysts and carbon nanofibers (CNFs) as hosts. FeCoNi nanoparticles are in suit synthesized in cotton-derived CNFs. FeCoNi with atomic-level mixing of each element can effectively modulate lithium polysulfides (LiPSs), multiple components making them promising to catalyze more LiPSs species. The higher configurational entropy endows FeCoNi@CNFs with extraordinary electrochemical activity, corrosion resistance, and mechanical properties. The fractal structure of CNFs provides a large specific surface area, leaving room for volume expansion and Li2 S accumulation, facilitating electrolyte wetting. The unique 3D conductive network structure can suppress the shuttle effect by physicochemical adsorption of LiPSs. This work systematically evaluates the performance of the obtained Li2 S6 /FeCoNi@CNFs electrode. The initial discharge capacity of Li2 S6 /FeCoNi@CNFs reaches 1670.8 mAh g-1 at 0.1 C under -20 °C. After 100 cycles at 0.2 C, the capacity decreases from 1462.3 to 1250.1 mAh g-1 . Notably, even under -40 °C at 0.1 C, the initial discharge capacity of Li2 S6 /FeCoNi@CNFs still reaches 1202.8 mAh g-1 . After 100 cycles at 0.2 C, the capacity retention rate is 50%. This work has important implications for the development of low-temperature Li-S batteries.

5.
J Laparoendosc Adv Surg Tech A ; 31(7): 783-789, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32915105

RESUMO

Purpose: To compare the relative clinical efficacies of radioactive and normal stent insertion methods as a means of treating patients suffering from malignant esophageal obstruction (MEO). Materials and Methods: The Pubmed, Embase, and Cochrane Library databases were searched for relevant randomized controlled trials (RCTs) from the date of inception through to July 2020. RevMan v5.3 was used for all data analyses. Results: This meta-analysis included six RCTs that included a total of 194 patients who had undergone radioactive stent insertion and 209 who had normal stent insertion. There were no significant differences in pooled improvement of dysphagia scores (P = .40), rates of stent restenosis (24.7% versus 28.7%, P = .35), stent migration (3.3% versus 4.4%, P = .61), severe chest pain (22.8% versus 20.3%, P = .61), hemorrhage (11.0% versus 9.8%, P = .80), or fistula formation (6.1% versus 4.2%, P = .55) between two groups. The pooled time to restenosis (P < .00001) and survival (P < .00001) were significant longer in the radioactive stent group. Significant heterogeneity was detected in the endpoint of improvement of dysphagia score (I2 = 89%; P = .0002). Funnel plot analyses did not detect any evidence of publication bias pertaining to the selected study endpoints. Conclusions: Our meta-analysis demonstrated that radioactive stent insertion can prolong stent patency and survival for patients with MEO compared with normal stent insertion.


Assuntos
Braquiterapia/instrumentação , Neoplasias Esofágicas/radioterapia , Estenose Esofágica/radioterapia , Implantação de Prótese/métodos , Stents , Braquiterapia/métodos , Transtornos de Deglutição/etiologia , Transtornos de Deglutição/radioterapia , Neoplasias Esofágicas/complicações , Estenose Esofágica/etiologia , Esofagoscopia , Fluoroscopia , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento
6.
Cancer Control ; 27(1): 1073274820956619, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32951452

RESUMO

At present, concurrent chemoradiotherapy (CRT) is considered the standard treatment of limited-stage small cell lung cancer (LS-SCLC). However, LS-SCLC is highly heterogeneous in the T stage, N stage, and prognosis. Increasing evidence has shown that individual treatment should be considered when treating LS-SCLC patients. The aim of the present study was to explore the optimal combination model of thoracic radiotherapy (TRT) and chemotherapy in N3 LS-SCLC. We retrospectively analyzed 93 N3 LS-SCLC patients treated in the Department of Oncology of Binzhou Medical University Hospital (Shandong, China) between March 2010 and October 2015. A total of 52 (52/93; 55.9%) patients received sequential CRT, and 41 (41/93; 44.1%) patients received concurrent CRT. All patients received 4-6 cycles of chemotherapy and TRT (50-60 Gy). The median follow-up time was 25.4 months (range was 6-65 months).The overall response rate was 88.5% in the sequential CRT group (9.6% complete response rate and 78.8% partial response rate) and 90.2% in the concurrent CRT group (14.6% complete response rate and 75.6% partial response rate). The PFS and OS were 15.4 months and 19.1 months in sequential CRT group, and 16.9 months and 20.5 months in concurrent CRT group. There was no significant difference in treatment response rate, PFS, and OS between sequential and concurrent CRT patients. The most common treatment-related toxicities were nausea/vomiting, neutropenia, and esophagitis. In conclusion, when concurrent CRT is performed in N3 LS-SCLC patients, tolerance to treatment should be fully considered. In our study, sequential CRT and concurrent CRT showed the same efficacy, and sequential CRT demonstrated better tolerance. However, these results require confirmation in future follow-up studies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Quimiorradioterapia/mortalidade , Neoplasias Pulmonares/terapia , Carcinoma de Pequenas Células do Pulmão/terapia , Cisplatino/administração & dosagem , Etoposídeo/administração & dosagem , Feminino , Seguimentos , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Carcinoma de Pequenas Células do Pulmão/patologia , Taxa de Sobrevida
7.
Aging (Albany NY) ; 12(9): 8680-8701, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32396871

RESUMO

Several of the thousands of long noncoding RNAs (lncRNAs) have been functionally characterized in various tumors. In this study, we aimed to explore the function and possible molecular mechanism of lncRNA KTN1 antisense RNA 1 (KTN1-AS1) involved in non-small cell lung cancer (NSCLC). We identified a novel NSCLC-related lncRNA, KTN1 antisense RNA 1 (KTN1-AS1) which was demonstrated to be distinctly highly expressed in NSCLC. KTN1-AS1 upregulation was induced by STAT1. Clinical study also suggested that higher levels of KTN1-AS1 were associated with advanced clinical progression and a shorter five-year overall survival. Functionally, loss-of-function assays with in vitro and in vivo experiments revealed that KTN1-AS1 promoted the proliferation, migration, invasion and EMT progress of NSCLC cells, and suppressed apoptosis. Mechanistic studies indicated that miR-23b was a direct target of KTN1-AS1, which functioned as a ceRNA to subsequently facilitate miR-23b's target gene DEPDC1 expression in NSCLC cells. Rescue experiments confirmed that KTN1-AS1 overexpression could increase the colony formation and migration ability suppressed by miR-23b upregulation in NSCLC cells. Overall, our findings imply that STAT1-induced upregulation of KTN1-AS1 display tumor-promotive roles in NSCLC progression via regulating miR-23b/DEPDC1 axis, suggesting that KTN1-AS1 may be a novel biomarker and therapeutic target for NSCLC patients.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/genética , Proteínas de Membrana/metabolismo , RNA Longo não Codificante/genética , Fator de Transcrição STAT1/metabolismo , Células A549 , Animais , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Movimento Celular/genética , Proliferação de Células/genética , Progressão da Doença , Feminino , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Invasividade Neoplásica/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fator de Transcrição STAT1/genética , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Cell Physiol ; 234(4): 3675-3684, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30256389

RESUMO

Hepatocyte nuclear factor 6 (HNF6), as a transcription factor, has been reported to be involved in cell proliferation, carcinogenesis, and tumor metastasis. Here, we demonstrated the role of HNF6 in tumor growth and liver metastasis in colorectal cancer (CRC). Through bioinformatics and clinical samples analysis, we found HNF6 messenger RNA was upregulated both in CRC primary sites and liver metastases, and its high expression indicated poor survival in CRC patients. In vitro studies confirmed that HNF6 promoted cell proliferation and colony formation. What is more, in mouse models, the xenografts grew significantly faster and liver metastasis rate was nearly 45% higher in mice injected with HNF6-overexpressing cells. Further mechanism exploration showed that HNF6 expression affected cell adhesion and conferred resistance to anoikis in CRC cells. Taken together, HNF6 expression was upregulated in CRC and closely correlated with poor survival. HNF6 promoted CRC cell proliferation and tumor growth, and may contribute to liver metastasis via conferring cell resistance to anoikis.


Assuntos
Movimento Celular , Proliferação de Células , Neoplasias Colorretais/metabolismo , Fator 6 Nuclear de Hepatócito/metabolismo , Neoplasias Hepáticas/metabolismo , Animais , Anoikis , Adesão Celular , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , Fator 6 Nuclear de Hepatócito/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/secundário , Camundongos Endogâmicos BALB C , Camundongos Nus , Transdução de Sinais , Carga Tumoral , Regulação para Cima
9.
Oncol Rep ; 40(5): 2778-2787, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30132532

RESUMO

The brain expressed x­linked gene 1 (BEX1) is a member of the BEX family and is aberrantly expressed in many cancers. However, the clinical significance of BEX1 expression level and its role in the pathology of esophageal squamous cell cancer (ESCC) remain unknown. In the present study, we determined BEX1 expression in the tumor and adjacent normal tissues from 118 ESCC patients by immunohistochemistry and determined the proliferation and growth of ESCC cells following ectopic overexpression of BEX1 in cultured cells and in mouse­ESCC xenografts. We observed that BEX1 was downregulated in ESCC tissues compared to adjacent normal tissues, and low BEX1 expression was significantly associated with larger ESCC tumor volume (P<0.001), advanced T stage (P=0.011) and advanced clinical stage (P=0.039). Additionally, survival analysis revealed that low expression of BEX1 significantly predicted poor prognosis in patients with ESCC (P<0.001). Multivariate analysis revealed that low BEX1 expression was an independent prognostic factor of poor survival (P=0.039). In vitro analysis revealed that overexpression of BEX1 inhibited ESCC cell proliferation and colony formation. Furthermore, in vivo tumorigenesis assays revealed that ectopic overexpression of BEX1 suppressed ESCC tumor growth in mice. Further immunoblotting analysis demonstrated that BEX1 upregulation led to reduced expression and phosphorylation of NF­κB p65, indicating inhibition of the NF­κB signaling pathway by BEX1. Our findings indicated that low BEX1 expression may be an independent prognostic marker for poor survival and may serve as a potential target for ESCC therapy.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/patologia , Proteínas do Tecido Nervoso/metabolismo , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/mortalidade , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/mortalidade , Carcinoma de Células Escamosas do Esôfago , Esôfago/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Metástase Linfática , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Fosforilação , Prognóstico , Análise de Sobrevida , Fator de Transcrição RelA/metabolismo , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J BUON ; 23(3): 814-819, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30003756

RESUMO

PURPOSE: To investigate the effects of microRNA-210 (miRNA- 210) on the biological behaviors (proliferation and invasion) of EC109 cells of highly metastatic human esophageal cancer (EC). METHODS: The EC109 genomic DNA of human EC was used as a template to amplify the precursor sequence of miRNA-210 by polymerase chain reaction (PCR). The precursor sequence of miRNA-210 was sub-cloned into the eukaryotic expression vector pcDNA3.1(-) via double digestion by BamH I and Hind III restriction enzymes. Then the pcDNA3.1 (-)-pri-miRNA-210 vector (named as p-miRNA-210) that was constructed successfully was transiently transfected into EC109 cells of human EC in vitro. Quantitative real-time PCR (qRT-PCR) was used to detect the expression level of mature miR-210. 3-(4,5-dimethyl-2-thiazolyl)-2,5- diphenyl-2-H-tetrazolium bromide (MTT) assay and scratch method were adopted to detect the proliferation and in vitro migration of EC109 cells, and flow cytometry was performed to detect the degree of cell apoptosis. RESULTS: The eukaryotic expression vector carrying miRNA- 210 was constructed successfully. Compared with that in the blank group (Mock) and the control group (P-Blank), miRNA-210 was overexpressed in the transfected EC109 cells. The cell apoptosis was significantly increased compared with that in the control group (p<0.05); the inhibition of proliferation of EC109 cells in the p-miRNA-210 vector transfected group was remarkably elevated (p<0.05), and wound healing ability was also significantly increased (p<0.05). CONCLUSION: The overexpression of miRNA-210 can significantly inhibit the proliferation of EC109 cells of human EC and accelerate the migration ability and the rate of apoptosis, providing a potential strategy for the treatment of EC.


Assuntos
Apoptose/genética , Proliferação de Células/genética , Neoplasias Esofágicas/genética , MicroRNAs/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Transfecção/métodos
11.
Oncol Res ; 25(7): 1039-1046, 2017 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-28244854

RESUMO

Hepatocyte cell adhesion molecule (hepaCAM), a new type of CAM, belongs to the immunoglobulin superfamily. Recently, hepaCAM was reported to be implicated in cancer development, and many researchers investigated its biological function in the tumorigenesis of various cancers. However, what kind of role hepaCAM plays in colorectal cancer (CRC) remains unknown. In this study, we found that hepaCAM was downregulated in CRC tissues and cell lines. Overexpression of hepaCAM inhibited CRC cell proliferation, migration, and invasion in vitro. Furthermore, the tumorigenesis assay showed that increased expression of hepaCAM suppressed CRC tumor growth and metastasis in vivo. We also demonstrated that overexpression of hepaCAM reduced the protein expression levels of ß-catenin, cyclin D1, and c-Myc, indicating its inhibitory effect on the Wnt/ß-catenin signaling pathway. In conclusion, our study results suggest hepaCAM as a promising therapeutic target for CRC and provide a better understanding for the molecular basis of CRC progression.


Assuntos
Neoplasias Colorretais/genética , Proteínas/genética , Animais , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Metástase Neoplásica , Ensaios Antitumorais Modelo de Xenoenxerto
12.
PLoS One ; 9(3): e91782, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24626299

RESUMO

An enhanced anti-apoptotic capacity of tumor cells plays an important role in the process of breakpoint cluster region/Abelson tyrosine kinase gene (BCR/ABL)-independent imatinib resistance. We have previously demonstrated that brain expressed X-linked 1 (BEX1) was silenced in secondary imatinib-resistant K562 cells and that re-expression of BEX1 can restore imatinib sensitivity resulting in the induction of apoptosis. However, the mechanism by which BEX1 executes its pro-apoptotic function remains unknown. We identified B-cell lymphoma 2 (BCL-2) as a BEX1-interacting protein using a yeast two-hybrid screen. The interaction between BEX1 and BCL-2 was subsequently confirmed by co-immunoprecipitation assays. Like BCL-2, BEX1 was localized to the mitochondria. The region between 33K and 64Q on BEX1 is important for its localization to the mitochondria and its ability to interact with BCL-2. Additionally, we found that this region is essential for BEX1-regulated imatinib-induced apoptosis. Furthermore, we demonstrated that the interaction between BCL-2 and BEX1 promotes imatinib-induced apoptosis by suppressing the formation of anti-apoptotic BCL-2/BCL-2-associated X protein (BAX) heterodimers. Our results revealed an interaction between BEX1 and BCL-2 and a novel mechanism of imatinib resistance mediated by the BEX1/BCL-2 pathway.


Assuntos
Apoptose/genética , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Humanos , Mesilato de Imatinib/uso terapêutico , Imunoprecipitação , Células K562 , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas do Tecido Nervoso/genética , Ligação Proteica , Mapas de Interação de Proteínas/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética
13.
Nan Fang Yi Ke Da Xue Xue Bao ; 32(1): 66-70, 2012 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-22366007

RESUMO

OBJECTIVE: To construct a recombinant lentiviral vector that stably express hepatocyte nuclear factor 6 (HNF6) in colorectal cancer cell line and examine its effects on the invasive ability of SW620 cells. METHODS: The lentiviral vector pLeno-DCE-HA-HNF6 was constructed and transfected into 293T cells. The supernatant containing the lentivirus particles was harvested to determine the virus titer. A stable cell line was established by infecting SW620 cells with the lentivirus particles, and the transfection efficiency was examined by fluorescence microscopy and flow cytometry. The invasion ability of the transfected SW620-HNF6 cells was assessed by wound healing and transwell assays. RESULTS: The recombinant lentiviral vector was correctly constructed and verified by sequencing. SW620-HNF6 cell line with stable HNF6 expression was established successfully, and the transfection efficiency reached 82.3%. Western blotting and quantitative PCR demonstrated significantly upregulated HNF6 expression in SW620-HNF6 cells, which showed obviously suppressed invasive ability in wound healing and transwell assays. CONCLUSION: We have successfully established a colorectal cancer cell line SW620-HNF6 stably expressing HNF6, which shows a lowered migration activity in vitro.


Assuntos
Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Fator 6 Nuclear de Hepatócito/biossíntese , Lentivirus/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Vetores Genéticos/genética , Fator 6 Nuclear de Hepatócito/genética , Humanos , Lentivirus/genética , Invasividade Neoplásica , Metástase Neoplásica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA