Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Oncologist ; 29(1): e25-e37, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-37390841

RESUMEN

BACKGROUND: The association between different phenotypes and genotypes of circulating tumor cells (CTCs) and efficacy of neoadjuvant chemotherapy (NAC) remains uncertain. This study was conducted to evaluate the relationship of FTH1 gene-associated CTCs (F-CTC) with/without epithelial-mesenchymal transition (EMT) markers, or their dynamic changes with the efficacy of NAC in patients with non-metastatic breast cancer. PATIENTS AND METHODS: This study enrolled 120 patients with non-metastatic breast cancer who planned to undergo NAC. The FTH1 gene and EMT markers in CTCs were detected before NAC (T0), after 2 cycles of chemotherapy (T1), and before surgery (T2). The associations of these different types of CTCs with rates of pathological complete response (pCR) and breast-conserving surgery (BCS) were evaluated using the binary logistic regression analysis. RESULTS: F-CTC in peripheral blood ≥1 at T0 was an independent factor for pCR rate in patients with HER2-positive (odds ratio [OR]=0.08, 95% confidence interval [CI], 0.01-0.98, P = .048). The reduction in the number of F-CTC at T2 was an independent factor for BCS rate (OR = 4.54, 95% CI, 1.14-18.08, P = .03). CONCLUSIONS: The number of F-CTC prior to NAC was related to poor response to NAC. Monitoring of F-CTC may help clinicians formulate personalized NAC regimens and implement BCS for patients with non-metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama , Células Neoplásicas Circulantes , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/cirugía , Células Neoplásicas Circulantes/patología , Estudios Prospectivos , Terapia Neoadyuvante , Mastectomía Segmentaria , Ferritinas/uso terapéutico , Oxidorreductasas/uso terapéutico
2.
J Med Virol ; 96(5): e29640, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38699969

RESUMEN

After the termination of zero-COVID-19 policy, the populace in China has experienced both Omicron BA.5 and XBB waves. Considering the poor antibody responses and severe outcomes observed among the elderly following infection, we conducted a longitudinal investigation to examine the epidemiological characteristics and antibody kinetics among 107 boosted elderly participants following the Omicron BA.5 and XBB waves. We observed that 96 participants (89.7%) were infected with Omicron BA.5, while 59 (55.1%) participants were infected with Omicron XBB. Notably, 52 participants (48.6%) experienced dual infections of both Omicron BA.5 and XBB. The proportion of symptomatic cases appeared to decrease following the XBB wave (18.6%) compared to that after the BA.5 wave (59.3%). Omicron BA.5 breakthrough infection induced lower neutralizing antibody titers against XBB.1.5, BA.2.86, and JN.1, while reinfection with Omicron XBB broadened the antibody responses against all measured Omicron subvariants and may alleviate the wild type-vaccination induced immune imprinting. Boosted vaccination type and comorbidities were the significant factors associated with antibody responses. Updated vaccines based on emerging severe acute respiratory syndrome coronavirus 2 variants are needed to control the Coronavirus Disease 2019 pandemic in the elderly.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Inmunización Secundaria , SARS-CoV-2 , Humanos , Anciano , COVID-19/epidemiología , COVID-19/inmunología , COVID-19/virología , Vacunas contra la COVID-19/administración & dosificación , Masculino , Femenino , Estudios Longitudinales , China/epidemiología , SARS-CoV-2/clasificación , SARS-CoV-2/fisiología , Anticuerpos Neutralizantes , Cinética , Anticuerpos Antivirales/sangre , Reinfección/epidemiología
3.
Exp Eye Res ; 223: 109201, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35940240

RESUMEN

The degeneration of the optic nerve narrows the visual field, eventually causing overall vision loss. This study aimed to identify global protein changes in the retina of optic nerve crushing (ONC) mice and to identify key regulators and pathways involved in injury-induced cell death during the progression of optic neurodegeneration. Label-free quantitative proteomics combined with bioinformatic analysis was performed on retinal protein extracts from ONC and sham-operated mice. Among the 1433 proteins detected, 121 proteins were differentially expressed in the retina of ONC mice. Further bioinformatic analysis showed that various metabolic pathways, including glutamate metabolism and γ-aminobutyric acid (GABA) synthesis, were significantly dysregulated in the injured mouse retinas. Glutamate decarboxylase 1 (GAD1) is the enzyme that converts glutamate into GABA, which was significantly up-regulated during ONC injury. Exogenous GAD1 treatment increased retinal ganglion cell (RGC) survival in the ONC-injured retina. In addition, changes in GAD1 expression were also observed in several other ophthalmic diseases. Vascular endothelial growth factor B (VEGF-B) has previously been reported to protect RGCs from apoptosis and positively regulated the expression of GAD1 in the retina. Notably, combination treatment with GAD1 and VEGF-B also provided strong protection against injury-induced RGC apoptosis. These results suggest that GAD1 expression may serve as an intrinsic protective mechanism that is commonly activated during retinal injury. Targeting GAD1 may serve as a potential strategy to treat optic neurodegenerative diseases.


Asunto(s)
Traumatismos del Nervio Óptico , Células Ganglionares de la Retina , Animales , Apoptosis/fisiología , Modelos Animales de Enfermedad , Glutamato Descarboxilasa , Glutamatos/metabolismo , Ratones , Compresión Nerviosa , Traumatismos del Nervio Óptico/metabolismo , Células Ganglionares de la Retina/metabolismo , Factor B de Crecimiento Endotelial Vascular/metabolismo , Ácido gamma-Aminobutírico/metabolismo
4.
Cancer Sci ; 111(11): 4288-4302, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32945042

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is one of the most common causes of hepatocellular carcinoma (HCC), but the underlying mechanisms behind the correlation of NAFLD with HCC are unclear. We aimed to uncover the genes and potential mechanisms that drive this progression. This study uncovered the genes and potential mechanisms through a multiple 'omics integration approach. Quantitative proteomics combined with phenotype-association analysis was performed. To investigate the potential mechanisms, a comprehensive transcriptome/lipidome/phenome-wide association analysis was performed in genetic reference panel BXD mice strains. The quantitative proteomics combined with phenotype-association results showed that VDAC1 was significantly increased in tumor tissues and correlated with NAFLD-related traits. Gene co-expression network analysis indicated that VDAC1 is involved in mitochondria dysfunction in the tumorigenic/tumor progression. The association between VDAC1 and mitochondria dysfunction can be explained by the fact that VDAC1 was associated with mitochondria membrane lipids cardiolipin (CL) composition shift. VDAC1 was correlated with the suppression of mature specie CL(LLLL) and elevation level of nascent CL species. Such profiling shift was supported by the significant positive correlation between VDAC1 and PTPMT1, as well as negative correlation with CL remodeling enzyme Tafazzin (TAZ). This study confirmed that the expression of VADC1 was dysregulated in NAFLD-driven HCC and associated with NAFLD progression. The VDAC1-driven mitochondria dysfunction is associated with cardiolipin composition shift, which causes alteration of mitochondria membrane properties.


Asunto(s)
Carcinoma Hepatocelular/etiología , Neoplasias Hepáticas/etiología , Mitocondrias/genética , Mitocondrias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/genética , Canales Aniónicos Dependientes del Voltaje/genética , Anciano , Animales , Carcinoma Hepatocelular/diagnóstico , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Variación Genética , Humanos , Neoplasias Hepáticas/diagnóstico , Masculino , Ratones , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Proteoma , Proteómica/métodos , Sitios de Carácter Cuantitativo , Transducción de Señal , Canales Aniónicos Dependientes del Voltaje/metabolismo
5.
Chin J Cancer Res ; 32(5): 564-579, 2020 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-33223752

RESUMEN

OBJECTIVE: Although T-cell immunoglobulin and mucin-domain containing molecule-3 (Tim-3) has been recognized as a promising target for cancer immunotherapy, its exact role in breast cancer has not been fully elucidated. METHODS: Tim-3 gene expression in breast cancer and its prognostic significance were analyzed. Associated mechanisms were then explored in vitro by establishing Tim-3-overexpressing breast cancer cells. RESULTS: In a pooled analysis of The Cancer Genome Atlas (TCGA) database, Tim-3 gene expression levels were significantly higher (P<0.001) in breast cancer tissue, compared with normal tissues. Tim-3 was a prognosis indicator in breast cancer patients [relapse-free survival (RFS), P=0.004; overall survival (OS), P=0.099]. Tim-3 overexpression in Tim-3low breast cancer cells promoted aggressiveness of breast cancer cells, as evidenced by enhanced proliferation, migration, invasion, tight junction deterioration and tumor-associated tubal formation. Tim-3 also enhanced cellular resistance to paclitaxel. Furthermore, Tim-3 exerted its function by activating the NF-κB/STAT3 signalling pathway and by regulating gene expression [cyclin D1 (CCND1), C-Myc, matrix metalloproteinase-1(MMP1), TWIST, vascular endothelial growth factor (VEGF) upregulation, concomitant with E-cadherin downregulation). Lastly, Tim-3 downregulated tight junction-associated molecules zona occludens (ZO)-2, ZO-1 and occludin, which may further facilitate tumor progression. CONCLUSIONS: Tim-3 plays an oncogenic role in breast cancer and may represent a potential target for antitumor therapy.

6.
Oncologist ; 24(11): e1044-e1054, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31300482

RESUMEN

BACKGROUND: The molecular phenotype of circulating tumor cells (CTCs) was associated with clinical outcome of patients with breast cancer. CTCs isolated from patients with metastatic breast cancer (MBC) display a unique microRNA (miRNA) expression profile. The aim of this study was to enhance the prognostic accuracy of the CTC phenotype in patients with MBC, by incorporating miRNA into a combined prediction model. SUBJECTS, MATERIALS, AND METHODS: CTCs were detected by CellSearch and enriched by magnetic cell sorting. miRNA deep sequencing and quantitative polymerase chain reaction were used to screen and verify potentially CTC-specific miRNA candidates. Patients with MBC were enrolled from two independent cohorts, and overall survival (OS) and chemotherapy response were analyzed. RESULTS: We screened and identified that miR-106b was an upregulated molecule in patients with MBC with CTC ≥5/7.5 mL (n = 16) compared with patients with CTC = 0/7.5 mL (n = 16) and healthy donors (n = 8). The expression of CTC-specific miR-106b correlated with vimentin and E-cadherin in CTC and acted as an independent factor for predicting OS (hazard ratio 2.157, 95% confidence interval [CI] 1.098-4.239, p = .026). Although CTC-specific miR-106b, E-cadherin, and vimentin showed a prognostic potential independently, the prognostic performance for OS based on the combination of three markers was significantly enhanced in Cohort 1 (area under the curve [AUC] 0.752, 95% CI 0.658-0.847, n = 128) and further validated in Cohort 2 (AUC 0.726, 95% CI 0.595-0.856, n = 91). Besides, a combined model incorporating miR-106b was associated with therapy response. CONCLUSION: The phenotypic assemblies of CTC incorporating miR-106b show enhanced prognostic accuracy of overall survival in patients with MBC. IMPLICATIONS FOR PRACTICE: In order to enhance the prognostic accuracy of the circulating tumor cell (CTC) phenotype in patients with metastatic breast cancer (MBC), this study screened and identified a CTC-specific microRNA (miRNA), miR-106b, as an upregulated molecule based on the comparison of miRNA profile between CTCs, primary tumors, and healthy blood donors. By incorporating miR-106b into a combined prediction model, the prognostic accuracy of the CTC phenotype for patients with MBC was greatly improved in both the training and validation cohorts. This work provides clinical evidence supporting the prognostic potential of CTC-specific miRNA for patients with MBC. These results indicate that developing CTC-specific miRNAs as new biomarkers will help to further optimize personalized therapy.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/secundario , MicroARNs/genética , Células Neoplásicas Circulantes/patología , Adulto , Anciano , Biomarcadores de Tumor/genética , Neoplasias de la Mama/patología , Estudios de Cohortes , Transición Epitelial-Mesenquimal/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Persona de Mediana Edad , Pronóstico , Análisis de Supervivencia , Adulto Joven
7.
Clin Proteomics ; 16: 12, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30988666

RESUMEN

BACKGROUND: Esophageal cancer (EC) is one of the malignant tumors with a poor prognosis. The early stage of EC is asymptomatic, so identification of cancer biomarkers is important for early detection and clinical practice. METHODS: In this study, we compared the protein expression profiles in esophageal squamous cell carcinoma (ESCC) tissues and adjacent normal esophageal tissues from five patients through high-resolution label-free mass spectrometry. Through bioinformatics analysis, we found the differentially expressed proteins of ESCC. To perform the rapid identification of biomarkers, we adopted a high-throughput protein identification technique of Quantitative Dot Blot (QDB). Meanwhile, the QDB results were verified by classical immunohistochemistry. RESULTS: In total 2297 proteins were identified, out of which 308 proteins were differentially expressed between ESCC tissues and normal tissues. By bioinformatics analysis, the four up-regulated proteins (PTMA, PAK2, PPP1CA, HMGB2) and the five down-regulated proteins (Caveolin, Integrin beta-1, Collagen alpha-2(VI), Leiomodin-1 and Vinculin) were selected and validated in ESCC by Western Blot. Furthermore, we performed the QDB and IHC analysis in 64 patients and 117 patients, respectively. The PTMA expression was up-regulated gradually along the progression of ESCC, and the PTMA expression ratio between tumor and adjacent normal tissue was significantly increased along with the progression. Therefore, we suggest that PTMA might be a potential candidate biomarker for ESCC. CONCLUSION: In this study, label-free quantitative proteomics combined with QDB revealed that PTMA expression was up-regulated in ESCC tissues, and PTMA might be a potential candidate for ESCC. Since Western Blot cannot achieve rapid and high-throughput screening of mass spectrometry results, the emergence of QDB meets this demand and provides an effective method for the identification of biomarkers.

8.
Biochem Biophys Res Commun ; 499(2): 338-344, 2018 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-29574154

RESUMEN

In order to better understand the mechanisms underlying the development of papillary thyroid carcinoma (PTC), and to identify new potential biomarkers, high-resolution label-free mass spectrometry was performed on PTC tissues and adjacent normal thyroid tissues from six patients. In this process, 2788 proteins were identified, out of which 49 proteins presented significant differences between PTC tissues and adjacent normal thyroid tissues. Gene ontology revealed that the majority of these proteins are involved in the catalytic activity and binding. We selected three proteins with differential expressions: PDZ and LIM domain 5 (PDLIM5), PDLIM1 and ALDH1A1; Protein expressions were further verified by RT-PCR and western blot. Among these, expression of PDLIM5 and PDLIM1 was up-regulated, while that of ALDH1A1 was down-regulated in PTC tissues. Next, we confirmed their expression through quantitative dot blot (QDB) technique. We found that knockdown of PDLIM5 expression in the B-CPAP cell line could inhibit the migration, invasion and proliferation of PTC cells. In addition, PDLIM5 knockdown reduced Ras and Phospho-ERK1/2 expression. Thus, we suggested that PDLIM5 promotes PTC via activation of the Ras-ERK pathway. Our research provides new molecular insight into the function of PDLIM5, which may assist in studying the mechanism of PTC. In addition, PDLIM5 could be further explored as a potential candidate for PTC treatment.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Biomarcadores de Tumor/metabolismo , Carcinoma Papilar/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteómica , Neoplasias de la Tiroides/metabolismo , Western Blotting , Carcinoma Papilar/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Técnicas de Silenciamiento del Gen , Humanos , Sistema de Señalización de MAP Quinasas , Invasividad Neoplásica , Proteínas de Neoplasias/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reproducibilidad de los Resultados , Coloración y Etiquetado , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides/patología , Proteínas ras/metabolismo
9.
Tumour Biol ; 39(6): 1010428317699755, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28618940

RESUMEN

Wnt1-inducible signaling pathway protein-1 is a cysteine-rich protein that belongs to the CCN family, which has been implicated in mediating the occurrence and progression through distinct molecular mechanisms in several tumor types. However, the association of Wnt1-inducible signaling pathway protein-1 with gastric cancer and the related molecular mechanisms remain to be elucidated. Therefore, this study aimed to clarify the biological role of Wnt1-inducible signaling pathway protein-1 in the proliferation, migration, and invasion in gastric cancer cells and further investigated the associated molecular mechanism on these biological functions. We first detected the expression level of Wnt1-inducible signaling pathway protein-1 in gastric cancer, and the reverse transcription polymerase chain reaction have shown that Wnt1-inducible signaling pathway protein-1 expression levels were upregulated in gastric cancer tissues. The expression of Wnt1-inducible signaling pathway protein-1 in gastric cancer cell lines was also detected by quantitative real-time polymerase chain reaction and Western blotting. Furthermore, two gastric cancer cell lines with high expression of Wnt1-inducible signaling pathway protein-1 were selected to explore the biological function of Wnt1-inducible signaling pathway protein-1 in gastric cancer. Function assays indicated that knockdown of Wnt1-inducible signaling pathway protein-1 suppressed cell proliferation, migration, and invasion in BGC-823 and AGS gastric cancer cells. Further investigation of mechanisms suggested that cyclinD1 was identified as one of Wnt1-inducible signaling pathway protein-1 related genes to accelerate proliferation in gastric cancer cells. In addition, one pathway of Wnt1-inducible signaling pathway protein-1 induced migration and invasion was mainly through the enhancement of epithelial-to-mesenchymal transition progression. Taken together, our findings presented the first evidence that Wnt1-inducible signaling pathway protein-1 was upregulated in gastric cancer and acted as an oncogene by promoting proliferation, migration, and invasion in gastric cancer cells.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Proteínas CCN de Señalización Intercelular/biosíntesis , Proliferación Celular/genética , Proteínas Proto-Oncogénicas/biosíntesis , Neoplasias Gástricas/genética , Biomarcadores de Tumor/genética , Proteínas CCN de Señalización Intercelular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteínas Proto-Oncogénicas/genética , Transducción de Señal , Neoplasias Gástricas/patología
10.
FASEB J ; 30(2): 578-89, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26432781

RESUMEN

The actin cytoskeleton is composed of a highly dynamic network of filamentous proteins, yet the molecular mechanism that regulates its organization and remodeling remains elusive. In this study, Na(+)/H(+) exchanger regulatory factor (NHERF)-1 loss-of-function and gain-of-function experiments reveal that polymerized actin cytoskeleton (F-actin) in HeLa cells is disorganized by NHERF1, whereas actin protein expression levels exhibit no detectable change. To elucidate the molecular mechanism underlying actin cytoskeleton disorganization by NHERF1, a combined 2-dimensional electrophoresis-matrix-assisted laser desorption/ionization-time of flight mass spectrometry approach was used to screen for proteins regulated by NHERF1 in HeLa cells. α-Actinin-4, an actin cross-linking protein, was identified. Glutathione S-transferase pull-down and coimmunoprecipitation studies showed the α-actinin-4 carboxyl-terminal region specifically interacted with the NHERF1 postsynaptic density 95/disc-large/zona occludens-1 domain. The NHERF1/α-actinin-4 interaction increased α-actinin-4 ubiquitination and decreased its expression levels, resulting in actin cytoskeleton disassembly. Our study identified α-actinin-4 as a novel NHERF1 interaction partner and provided new insights into the regulatory mechanism of the actin cytoskeleton by NHERF1.


Asunto(s)
Actinina/metabolismo , Citoesqueleto/fisiología , Fosfoproteínas/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Actinina/genética , Animales , Células COS , Chlorocebus aethiops , Regulación hacia Abajo , Regulación de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Fosfoproteínas/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Intercambiadores de Sodio-Hidrógeno/genética , Ubiquitina/metabolismo
11.
Nanomedicine ; 13(2): 641-657, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27521693

RESUMEN

Disulfiram (DS), an anti-alcoholism drug, shows very strong cytotoxicity in many cancer types. However its clinical application in cancer treatment is limited by the very short half-life in the bloodstream. In this study, we developed a poly lactic-co-glycolic acid (PLGA)-encapsulated DS protecting DS from the degradation in the bloodstream. The newly developed DS-PLGA was characterized. The DS-PLGA has very satisfactory encapsulation efficiency, drug-loading content and controlled release rate in vitro. PLGA encapsulation extended the half-life of DS from shorter than 2minutes to 7hours in serum. In combination with copper, DS-PLGA significantly inhibited the liver cancer stem cell population. CI-isobologram showed a remarkable synergistic cytotoxicity between DS-PLGA and 5-FU or sorafenib. It also demonstrated very promising anticancer efficacy and antimetastatic effect in liver cancer mouse model. Both DS and PLGA are FDA approved products for clinical application. Our study may lead to repositioning of DS into liver cancer treatment.


Asunto(s)
Inhibidores del Acetaldehído Deshidrogenasa/administración & dosificación , Disulfiram/administración & dosificación , Neoplasias Hepáticas/tratamiento farmacológico , Nanopartículas , Animales , Portadores de Fármacos , Glicoles , Humanos , Ácido Láctico , Ácido Poliglicólico
12.
Anticancer Drugs ; 27(1): 24-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26457548

RESUMEN

Neocarzinostatin (NCS) consists of an enediyne chromophore and an apoprotein (NCP). Lidamycin (LDM) is composed of another active enediyne chromophore (AE) and an acidic protein (LDP). Although the structures of NCP and LDP are very similar, LDM has been shown to have an increased tumor-suppressive activity than that of NCS. The aim of this study was to construct a chimeric protein (CMP) that consists of both the terminus residue of NCP and an LDP pocket-forming residue that can bind AE. This CMP will have a structure similar to NCS and an antitumor activity similar to LDM. The assembling efficiency of LDP, CMP, and NCP was 73.9, 1.5, and 1.1%, respectively. The cytotoxicity was consistent with their assembling efficiency of AE in proteins. When CMP-AE and NCP-AE were administered at equivalent AE doses of LDM, the inhibition rate of CMP-AE was the same as LDM and significantly higher than that of NCP-AE. Our study implied that the binding activity between LDP and AE was very specific. The terminus residue of LDP could affect the specifically binding activity. The pocket-forming residue could confer a protective function to the chromophore. Further investigation of its bioactivity might serve as a new drug design strategy and drug-delivery carrier in targeted cancer therapy.


Asunto(s)
Aminoglicósidos/química , Antineoplásicos/química , Apoproteínas/química , Enediinos/química , Proteínas Recombinantes de Fusión/química , Cinostatina/química , Aminoglicósidos/genética , Animales , Antineoplásicos/farmacología , Apoproteínas/genética , Apoproteínas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología
13.
J Enzyme Inhib Med Chem ; 31(3): 495-502, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26382011

RESUMEN

Novel N-aroyl-α,ß-unsaturated piperidones, series 1, series 2 and series 3 (featuring 2-bromo-4,5-dimethoxybenzylidene, 4-dimethylaminobenzylidene and 4-trifluoromethylbenzylidene, respectively), were synthesized as candidate cytotoxins. Most of the compounds displayed potent cytotoxicity against the human neoplastic cell lines SK-BR-3, PG-BE1, NCI-H460, MIA PaCa-2 and SW1990 in vitro, and approximately 64% of the IC50 values were lower than 5 µM. Among those tested, compound 1b of series 1, 3a, 3d and 3e of series 3 proved to be the most active. Importantly, 1b displayed marked inhibitory effects on tumor growth in vivo and had no apparent toxicity to mice; this was evaluated by a nude mouse PG-BE1 xenograft model. In addition, the fluorescent properties of compounds series 1-3 were investigated. The interesting fluorescence exhibited by these compounds could be useful for their visualization in tumor cells, permitting further studies on these α,ß-unsaturated piperidones as candidates for novel fluorescent antitumor agents.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Fluorescencia , Piperidonas/farmacología , Animales , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Ratones Desnudos , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Piperidonas/síntesis química , Piperidonas/química , Relación Estructura-Actividad
14.
Anticancer Drugs ; 26(1): 64-73, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25122444

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) mainly induces apoptosis through the extrinsic death receptor-induced pathway by ligation with death receptor 4 (DR4) and death receptor 5 (DR5). On the basis of the antitumor activity to cancer cells and no cytotoxity to normal cells of TRAIL and the target of the epidermal growth factor receptor (EGFR) ligand peptide, the study constructed a new bispecific fusion protein and a new bifunctional enediyne-energized fusion protein and investigated their antitumor efficacy. Bispecific fusion protein Ec-LDP-TRAIL showed potent binding activity to cancer cell lines with a high expression of EGFR or DR4/DR5 such as A431 and H460 cells, whereas poor binding activity to NIH/3T3 cells with low expressing EGFR and DR4/DR5. Ec-LDP-TRAIL also showed more potent cytotoxicity to A431 and H460 cells than Ec-LDP, which could result from the TRAIL-inducing apoptosis. Results of an in-vivo efficacy study showed that Ec-LDP-TRAIL at a dose of 10 mg/kg decreased the growth of epidermoid carcinoma A431 xenografts by 80.19% (P < 0.01) on day 26. Immunohistochemical detection of nuclear antigen factor Ki-67 suggested that Ec-LDP-TRAIL effectively induced cell necrosis and inhibited cell proliferation of tumor. From IC50 values, bispecific and bifunctional energized fusion protein Ec-LDP-TRAIL-AE was more potent and selective in its cytotoxicity against different carcinoma cell lines than corresponding lidamycin in vitro and induction of the cleavage of poly(ADP-ribose)polymerase was observed in A431 cells treated with Ec-LDP-TRAIL-AE and lidamycin, respectively. Ec-LDP-TRAIL-AE also significantly inhibited the growth of A431 xenografts in a nude mouse model. These properties suggested that Ec-LDP-TRAIL and Ec-LDP-TRAIL-AE may be promising candidates for targeted cancer therapy.


Asunto(s)
Aminoglicósidos/genética , Antineoplásicos/farmacología , Receptores ErbB/metabolismo , Oligopéptidos/genética , Proteínas Recombinantes de Fusión/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Enediinos , Xenoinjertos , Humanos , Ligandos , Ratones , Ratones Desnudos , Células 3T3 NIH , Necrosis , Trasplante de Neoplasias , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/uso terapéutico
15.
J Nanosci Nanotechnol ; 15(5): 3773-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26505004

RESUMEN

PEGylated hollow mesoporous silica nanoparticles (HMSN-PEG) were successfully fabricated by only one simple step through hydrothermal treatment in Na2CO3 solution. HMSN-PEG nanoparticles were transformed from conventional PEG-modified mesoporous silica nanoparticles (MSN-PEG). The as-synthesized HMSN-PEG nanoparticles exhibited higher loading capacity of anticancer drug (Doxorubicin) and better sustained release property than MSN and MSN-PEG particles. In vitro cell viability of HMSN-PEG nanoparticles to Hep-G2 cells was evaluated. HMSN-PEG nanoparticles have little in vitro cytotoxicity up to a concentration of 500 µg/ml. Furthermore, the DOX-loaded HMSN-PEG nanoparticles exhibited higher cytotoxicity than the DOX-loaded MSN and MSN-PEG nanoparticles against Hep-G2 cells. Therefore, the HMSN-PEG nanoparticle that generated in this PEG protecting etching strategy is a promising nanocarrier toward its potential application for cancer therapy.


Asunto(s)
Portadores de Fármacos/química , Nanopartículas/química , Dióxido de Silicio/química , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/química , Doxorrubicina/farmacología , Portadores de Fármacos/toxicidad , Células Hep G2 , Humanos , Nanopartículas/toxicidad , Porosidad , Dióxido de Silicio/toxicidad
16.
Breast Cancer Res Treat ; 145(1): 45-59, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24710637

RESUMEN

αB-crystallin (CRYAB) is present at a high frequency in poor prognosis basal-like breast tumours, which are largely absent of oestrogen, progesterone receptors and HER2 known as triple-negative breast cancer (TNBC). CRYAB functions as a molecular chaperone to bind to and correct intracellular misfolded/unfolded proteins such as vascular endothelial growth factor (VEGF), preventing non-specific protein aggregations under the influence of the tumour microenvironment stress and/or anti-cancer treatments including bevacizumab therapy. Directly targeting CRYAB can sensitize tumour cells to chemotherapeutic agents and decrease tumour aggressiveness. However, growing evidence shows that CRYAB is a critical adaptive response element after ischemic heart disease and stroke, implying that directly targeting CRYAB might cause serious unwanted side effects. Here, we used structure-based molecular docking of CRYAB and identified a potent small molecular inhibitor, NCI-41356, which can strongly block the interaction between CRYAB and VEGF165 without affecting CRYAB levels. The disruption of the interaction between CRYAB and VEGF165 elicits in vitro anti-tumour cell proliferation and invasive effects through the down-regulation of VEGF signalling in the breast cancer cells. The observed in vitro anti-tumour angiogenesis of endothelial cells might be attributed to the down-regulation of paracrine VEGF signalling in the breast cancer cells after treatment with NCI-41356. Intraperitoneal injection of NCI-41356 greatly inhibits the tumour growth and vasculature development in in vivo human breast cancer xenograft models. Our findings provide 'proof-of-concept' for the development of highly specific structure-based alternative targeted therapy for the prevention and/or treatment of TNBC.


Asunto(s)
Antineoplásicos/farmacología , Ácido Glutámico/análogos & derivados , Neoplasias de la Mama Triple Negativas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cadena B de alfa-Cristalina/antagonistas & inhibidores , Animales , Antineoplásicos/química , Línea Celular Tumoral , Descubrimiento de Drogas , Ensayo de Inmunoadsorción Enzimática , Femenino , Ácido Glutámico/química , Ácido Glutámico/farmacología , Humanos , Ratones , Ratones Desnudos , Simulación del Acoplamiento Molecular , Neovascularización Patológica/metabolismo , Estructura Cuaternaria de Proteína , Ensayos Antitumor por Modelo de Xenoinjerto , Cadena B de alfa-Cristalina/química
17.
PeerJ ; 12: e17163, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38766480

RESUMEN

Background: The evidence on the effects of extreme meteorological conditions and high air pollution levels on incidence of hand, foot and mouth disease (HFMD) is limited. Moreover, results of the available studies are inconsistent. Further investigations are imperative to elucidate the specific issue. Methods: Data on the daily cases of HFMD, meteorological factors and air pollution were obtained from 2017 to 2022 in Jining City. We employed distributed lag nonlinear model (DLNM) incorporated with Poisson regression to explore the impacts of extreme meteorological conditions and air pollution on HFMD incidence. Results: We found that there were nonlinear relationships between temperature, wind speed, PM2.5, SO2, O3 and HFMD. The cumulative risk of extreme high temperature was higher at the 95th percentile (P95th) than at the 90th percentile(P90th), and the RR values for both reached their maximum at 10-day lag (P95th RR = 1.880 (1.261-2.804), P90th RR = 1.787 (1.244-2.569)), the hazardous effect of extreme low temperatures on HFMD is faster than that of extreme high temperatures. The cumulative effect of extreme low wind speeds reached its maximum at 14-day lag (P95th RR = 1.702 (1.389-2.085), P90th RR = 1.498(1.283-1.750)). The cumulative effect of PM2.5 concentration at the P90th was largest at 14-day lag (RR = 1.637 (1.069-2.506)), and the cumulative effect at the P95th was largest at 10-day lag (RR = 1.569 (1.021-2.411)). High SO2 concentration at the P95th at 14-day lag was associated with higher risk for HFMD (RR: 1.425 (1.001-2.030)). Conclusion: Our findings suggest that high temperature, low wind speed, and high concentrations of PM2.5 and SO2 are associated with an increased risk of HFMD. This study not only adds insights to the understanding of the impact of extreme meteorological conditions and high levels of air pollutants on HFMD incidence but also holds practical significance for the development and enhancement of an early warning system for HFMD.


Asunto(s)
Contaminantes Atmosféricos , Contaminación del Aire , Enfermedad de Boca, Mano y Pie , Enfermedad de Boca, Mano y Pie/epidemiología , China/epidemiología , Humanos , Incidencia , Contaminantes Atmosféricos/efectos adversos , Contaminantes Atmosféricos/análisis , Contaminación del Aire/efectos adversos , Contaminación del Aire/análisis , Material Particulado/efectos adversos , Material Particulado/análisis , Preescolar , Femenino , Viento , Masculino , Lactante , Dióxido de Azufre/análisis , Dióxido de Azufre/efectos adversos , Conceptos Meteorológicos , Clima Extremo , Niño
18.
Adv Sci (Weinh) ; 11(6): e2307271, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38072640

RESUMEN

Chemotherapy is widely used to treat colorectal cancer (CRC). Despite its substantial benefits, the development of drug resistance and adverse effects remain challenging. This study aimed to elucidate a novel role of glucagon in anti-cancer therapy. In a series of in vitro experiments, glucagon inhibited cell migration and tube formation in both endothelial and tumor cells. In vivo studies demonstrated decreased tumor blood vessels and fewer pseudo-vessels in mice treated with glucagon. The combination of glucagon and chemotherapy exhibited enhanced tumor inhibition. Mechanistic studies demonstrated that glucagon increased the permeability of blood vessels, leading to a pronounced disruption of vessel morphology. Signaling pathway analysis identified a VEGF/VEGFR-dependent mechanism whereby glucagon attenuated angiogenesis through its receptor. Clinical data analysis revealed a positive correlation between elevated glucagon expression and chemotherapy response. This is the first study to reveal a role for glucagon in inhibiting angiogenesis and vascular mimicry. Additionally, the delivery of glucagon-encapsulated PEGylated liposomes to tumor-bearing mice amplified the inhibition of angiogenesis and vascular mimicry, consequently reinforcing chemotherapy efficacy. Collectively, the findings demonstrate the role of glucagon in inhibiting tumor vessel network and suggest the potential utility of glucagon as a promising predictive marker for patients with CRC receiving chemotherapy.


Asunto(s)
Neoplasias Colorrectales , Glucagón , Humanos , Animales , Ratones , Glucagón/farmacología , Glucagón/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neoplasias Colorrectales/patología , Transducción de Señal , Línea Celular Tumoral
19.
BMC Cancer ; 13: 479, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24128285

RESUMEN

BACKGROUND: Endostatin (ES) inhibits endothelial cell proliferation, migration, invasion, and tube formation. It also shows antiangiogenesis and antitumor activities in several animal models. Endostatin specifically targets tumor vasculature to block tumor growth. Lidamycin (LDM), which consists of an active enediyne chromophore (AE) and a non-covalently bound apo-protein (LDP), is a member of chromoprotein family of antitumor antibiotics with extremely potent cytotoxicity to cancer cells. Therefore, we reasoned that endostatin-lidamycin (ES-LDM) fusion proteins upon energizing with enediyne chromophore may obtain the combined capability targeting tumor vasculature and tumor cell by respective ES and LDM moiety. METHODS: In this study, we designed and obtained two new endostatin-based fusion proteins, endostatin-LDP (ES-LDP) and LDP-endostatin (LDP-ES). In vitro, the antiangiogenic effect of fusion proteins was determined by the wound healing assay and tube formation assay and the cytotoxicity of their enediyne-energized analogs was evaluated by CCK-8 assay. Tissue microarray was used to analyze the binding affinity of LDP, ES or ES-LDP with specimens of human lung tissue and lung tumor. The in vivo efficacy of the fusion proteins was evaluated with human lung carcinoma PG-BE1 xenograft and the experimental metastasis model of 4T1-luc breast cancer. RESULTS: ES-LDP and LDP-ES disrupted the formation of endothelial tube structures and inhibited endothelial cell migration. Evidently, ES-LDP accumulated in the tumor and suppressed tumor growth and metastasis. ES-LDP and ES show higher binding capability than LDP to lung carcinoma; in addition, ES-LDP and ES share similar binding capability. Furthermore, the enediyne-energized fusion protein ES-LDP-AE demonstrated significant efficacy against lung carcinoma xenograft in athymic mice. CONCLUSIONS: The ES-based fusion protein therapy provides some fundamental information for further drug development. Targeting both tumor vasculature and tumor cells by endostatin-based fusion proteins and their enediyne-energized analogs probably provides a promising modality in cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Endostatinas/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Endostatinas/farmacocinética , Endostatinas/uso terapéutico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Femenino , Humanos , Pulmón/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/uso terapéutico , Análisis de Matrices Tisulares , Distribución Tisular , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Mol Med Rep ; 28(4)2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37681454

RESUMEN

Vascular endothelial growth factor B (VEGFB) plays a crucial role in glucolipid metabolism and is highly associated with type 2 diabetes mellitus (T2DM). The role of VEGFB in the insulin secretion of ß cells remains unverified. Thus, the present study aimed to discuss the effect of VEGFB on regulating insulin secretion in T2DM development, and its underlying mechanism. A high­fat diet and streptozocin (STZ) were used for inducing T2DM in mice model, and VEGFB gene in islet cells of T2DM mice was knocked out by CRISPR Cas9 and overexpressed by adeno­Associated Virus (AAV) injection. The effect of VEGFB and its underlying mechanism was assessed by light microscopy, electron microscopy and fluorescence confocal microscopy, enzyme­linked immunosorbent assay, mass spectrometer and western blot analysis. The decrement of insulin secretion in islet ß cell of T2DM mice were aggravated and blood glucose remained at a high level after VEGFB knockout (KO). However, glucose tolerance and insulin sensitivity of T2DM mice were improved after the AAV­VEGFB186 injection. VEGFB KO or overexpression can inhibit or activate PLCγ/IP3R in a VEGFR1­dependent manner. Then, the change of PLCγ/IP3R caused by VEGFB/VEGFR1 will alter the expression of key factors on the Ca2+/CaMK2 signaling pathway such as PPP3CA. Moreover, VEGFB can cause altered insulin secretion by changing the calcium concentration in ß cells of T2DM mice. These findings indicated that VEGFB activated the Ca2+/CaMK2 pathway via VEGFR1­PLCγ and IP3R pathway to regulate insulin secretion, which provides new insight into the regulatory mechanism of abnormal insulin secretion in T2DM.


Asunto(s)
Traumatismos Craneocerebrales , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Animales , Ratones , Secreción de Insulina , Factor B de Crecimiento Endotelial Vascular , Transducción de Señal , Dependovirus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA