Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nutr Hosp ; 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38726608

RESUMEN

The effectiveness of an elemental diet (ED) for preventing adverse events (AEs) during chemotherapy for patients with esophageal cancer (EC) remains unclear. The aim of this meta-analysis was to comprehensively assess the efficacy of ED for preventing AE in EC patients during chemotherapy. Medline (via PubMed), Embase, the Cochrane Library, and Web of Science were searched to retrieve prospective and randomized studies published before April 12, 2023. The odds ratio (OR) of each AE was calculated using Review Manger 5.4.1. The risk of bias was assessed, and a random effect model-based meta-analysis was used to analyze the available data. Four prospective and randomized studies involving 237 patients were identified after a systematic search. Regarding gastrointestinal toxicities, the findings indicated a trend toward a decrease in the risk of mucositis (OM) (OR = 0.54, 95 % CI: 0.25-1.14), constipation (OR = 0.87, 95 % CI: 0.49-1.53), and anorexia (OR = 0.99, 95 % CI: 0.32-3.05), as well as an increasing trend in the risk of diarrhea (OR = 1.48, 95 % CI: 0.79-2.79), among patients treated with ED. However, none of these reached statistical significance. For hematological toxicities, the risk of all-grade neutropenia (OR = 0.28, 95 % CI: 0.14-0.57), grade ≥ 2 leucopenia (OR = 0.43, 95 % CI: 0.22-0.84), grade ≥ 2 neutropenia (OR = 0.34, 95 % CI: 0.17-0.67), and grade ≥ 3 neutropenia (OR = 0.28, 95 % CI: 0.12-0.63) was significantly decreased. There is no firm evidence confirming the preventive effect of an ED against OM or diarrhea. However, an ED may potentially be helpful in preventing neutropenia and leucopenia.

2.
Cancer Prev Res (Phila) ; 16(12): 669-679, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37857481

RESUMEN

Numerous studies and clinical trials have shown that immune checkpoint inhibitors can effectively prevent tumor growth and metastasis in esophageal squamous cell carcinoma (ESCC) patients. In this study, we aimed to evaluate the anti-tumor effects of PD-1 antibody preventive treatment in patients with early stages ESCC as well as patients with high-grade intraepithelial neoplasia (HGIN). We first established an ESCC model using C57BL/6J mice treated with the chemical carcinogen 4- NQO and observed esophageal lesions at different time points. Second, we compared the antitumor efficacy of PD-1 antibody treatment in mice at the ESCC stage and PD-1 antibody preventive treatment in mice at the HGIN stage. The results showed that PD-1 antibody preventive treatment effectively impeded the progression of 4NQO-induced esophageal tumorigenesis. IHC analysis was performed to observe the infiltration of immune cells into the tumor microenvironment. It has been shown that active tissue-resident memory T cells can be induced and resided into the tumor microenvironment for a long period after treatment with PD-1 antibody. Reexposure to the oncogenic environment colonized by CD8+TRM cells can still exert antitumor effects. These results provide new strategies for the treatment of patients with early stage ESCC and HGIN. PREVENTION RELEVANCE: Immune checkpoint inhibitors have shown promising results in multiple tumor species. However, there is currently no clinical application to evaluate their therapeutic value in cancer preventive treatment. Prophylactic use of immune checkpoint inhibitors in the early stages of ESCC may provide long-term benefits to patients.


Asunto(s)
Carcinoma in Situ , Carcinoma de Células Escamosas , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Humanos , Animales , Ratones , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas/patología , Neoplasias Esofágicas/patología , Receptor de Muerte Celular Programada 1 , Células T de Memoria , Inhibidores de Puntos de Control Inmunológico , Ratones Endogámicos C57BL , Carcinoma in Situ/patología , Anticuerpos , Carcinogénesis , Microambiente Tumoral
3.
Biomedicines ; 11(5)2023 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-37239162

RESUMEN

OBJECTIVE: Epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) is a first-line treatment for lung adenocarcinoma with EGFR-sensitive mutations, but acquired resistance to EGFR-TKIs remains a problem in clinical practice. The development of epithelial-mesenchymal transition (EMT) is a critical mechanism that induces acquired resistance to TKIs. Reversing acquired resistance to EGFR-TKIs through targeting the key molecules driving EMT provides an alternative choice for patients. We, therefore, aimed to explore the role of doublecortin-like kinase 1 (DCLK1) as an EMT driver gene in the acquired resistance of lung adenocarcinoma to EGFR-TKIs. METHODS: The IC50 of Gefitinib or Osimertinib in PC9/HCC827 cells was measured using a cell counting kit-8 (CCK8) assay. The expression levels of EMT-related genes in PC9 and HCC827 cells were detected using RT-PCR and Western blot. Cell migration and invasion abilities were assessed via a transwell assay. For the in vivo experiments, PC9 cells were subcutaneously injected into BALB/c nude mice to form tumors. Upon harvesting, tumor tissues were retained for RT-PCR, Western blot, and polychromatic fluorescence staining to detect biomarker changes in the EMT process. RESULTS: Gefitinib-resistant PC9 (PC9/GR) and Osimertinib-resistant HCC827 (HCC827/OR) cells showed remarkable activation of EMT and enhanced migration and invasion abilities compared to TKI-sensitive cells. In addition, DCLK1 expression was markedly increased in EGFR-TKI-resistant lung adenocarcinoma cells. The targeted knockout of DCLK1 effectively reversed the EMT phenotype in TKI-resistant cells and improved EGFR-TKI sensitivity, which was further validated by the in vivo experiments. CONCLUSIONS: DCLK1 facilitates acquired resistance to EGFR-TKI in lung adenocarcinoma by inducting EMT and accelerating the migration and invasion abilities of TKI-resistant cells.

4.
Breast Cancer Res ; 25(1): 43, 2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-37069669

RESUMEN

Triple-negative breast cancer (TNBC) exhibits the poorest outcomes among breast cancer subtypes due to the high heterogeneity and a lasting scarcity of effectual treatments. Targeted therapies based on molecular subtypes of TNBC are critical step toward tailoring treatments to improve clinical outcomes. Gastrointestinal cancer stem cell (CSC) marker DCLK1 was reported to be highly expressed in stem cell-rich subtype of TNBC. Here, we firstly explored the impacts of DCLK1 on tumor cells as well as their immune microenvironment in TNBC and potential therapeutic strategies for TNBC patients with high DCLK1 expression. Our results disclosed that DCLK1 overexpression promoted, while knockout of DCLK1 suppressed the CSC-like traits of TNBC cells and resistance to chemotherapeutics. Besides, DCLK1 supported immune escape by inhibiting intratumoral cytotoxic T cell infiltration in TNBC and hence limited immune checkpoint inhibitors efficacy. Mechanistically, bioinformatics analysis revealed that IL-6/STAT3 signaling was significantly enriched in high DCLK1-expressing patients, and our results further revealed that DCLK1 enhanced IL-6 expression and STAT3 activation in TNBC cells, which finally gave rise to upregulated CSC traits and suppressed CD8+ T-cell activity. Inhibiting IL-6/STAT3 pathway by IL-6R antagonist, Tocilizumab or STAT3 inhibitor, S31-201 could abolish DCLK1-promoted malignant phenotypes of TNBC cells. Finally, DCLK1 was identified to be specifically and highly expressed in the mesenchymal-like subtype of TNBC and targeting DCLK1 could improve chemotherapy efficacy and activate antitumor immunity. Overall, our study revealed the potential clinical benefits of targeting DCLK1 in TNBC treatment.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Interleucina-6/genética , Interleucina-6/metabolismo , Proteínas Serina-Treonina Quinasas , Transducción de Señal , Línea Celular Tumoral , Células Madre Neoplásicas/patología , Microambiente Tumoral , Quinasas Similares a Doblecortina , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/uso terapéutico
6.
Cancer Gene Ther ; 30(1): 192-208, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36151333

RESUMEN

The chromatin-modifying enzyme ATAD2 confers oncogenic competence and proliferative advantage in malignances. We previously identified ATAD2 as a marker and driver of cell proliferation in ovarian cancer (OC); however, the mechanisms whereby ATAD2 is regulated and involved in cell proliferation are still unclear. Here, we disclose that ATAD2 displays a classical G2/M gene signature, functioning to facilitate mitotic progression. ATAD2 ablation caused mitotic arrest and decreased the ability of OC cells to pass through nocodazole-arrested mitosis. ChIP-seq data analyses demonstrated that DREAM and MYBL2-MuvB (MMB), two switchable MuvB-based complexes, bind the CHR elements in the ATAD2 promoter, representing a typical feature and principle mechanism of the periodic regulation of G2/M genes. As a downstream target of MYBL2, ATAD2 deletion significantly impaired MYBL2-driven cell proliferation. Intriguingly, ATAD2 silencing also fed back to destabilize the MYBL2 protein. The significant coexpression of MYBL2 and ATAD2 at both the bulk tissue and single-cell levels highlights the existence of the MYBL2-ATAD2 signaling in OC patients. This signaling is activated during tumorigenesis and correlated with TP53 mutation, and its hyperactivation was found especially in high-grade serous and drug-resistant OCs. Disrupting this signaling by CRISPR/Cas9-mediated ATAD2 ablation inhibited the in vivo growth of OC in a subcutaneous tumor xenograft mouse model, while pharmacologically targeting this signaling with an ATAD2 inhibitor demonstrated high therapeutic efficacy in both drug-sensitive and drug-resistant OC cells. Collectively, we identified a novel MYBL2-ATAD2 proliferative signaling axis and highlighted its potential application in developing new therapeutic strategies, especially for high-grade serous and drug-resistant OCs.


Asunto(s)
Neoplasias Ováricas , Transducción de Señal , Humanos , Ratones , Animales , Femenino , Proliferación Celular/genética , Neoplasias Ováricas/patología , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , Transactivadores/genética , Proteínas de Ciclo Celular/genética , ATPasas Asociadas con Actividades Celulares Diversas/genética , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Proteínas de Unión al ADN/metabolismo
7.
Cancer Lett ; 531: 83-97, 2022 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-35157971

RESUMEN

Lung adenocarcinoma is the most common form of lung cancer, accounting for 60% of non-small cell lung cancer (NSCLC) cases in Asian patients. Importantly, nearly half of these patients have epithelial growth factor receptor (EGFR) mutations. Though EGFR-tyrosine kinase inhibitors (EGFR-TKIs) are recommended as the first-line therapy for NSCLC patients, the development of resistance reduces their efficiency and limits their application. As the complicated and heterogeneous mechanism of acquired resistance among individuals, the efficiency of anti-angiogenesis therapy, immune checkpoint inhibitors, or chemo-radiotherapies is rather less promising. In this research, we investigated the role of the tumor stem cell marker DCLK1 in EGFR-TKI resistance of lung adenocarcinoma. We discovered that DCLK1 was critical in maintaining the stemness of tumor cells through the Wnt/ß-Catenin pathway, which was conducive to the development of EGFR-TKI resistance. Inhibiting DCLK1 activity restored the sensitivity of TKI-resistant tumor cells and organoids. Moreover, our study showed that DCLK1 inhibitor had a synergistic effect in controlling tumor growth when combined with EGFR-TKIs. Overall, our study provides new insights into EGFR-TKI resistant lung adenocarcinoma through inhibition of DCLK1 expression.


Asunto(s)
Adenocarcinoma del Pulmón , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Quinasas Similares a Doblecortina , Resistencia a Antineoplásicos , Receptores ErbB/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento/genética , beta Catenina/genética
8.
Transl Oncol ; 17: 101317, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34998236

RESUMEN

Immunotherapy has recently become a promising cancer therapy with extensive applications of immune checkpoint inhibitors (ICIs). However, pancreatic ductal adenocarcinoma (PDAC) appears to be unresponsive to immunotherapy due to the immunosuppressive microenvironment. Recent studies showed that cancer stem cell marker DCLK1 promoted the initiation and development of PDAC. Nevertheless, the mechanism driving this process remains unclear. Here, by performing gain-of-function investigations in PDAC cell lines, we demonstrate that both DCLK1 long (DCLK1-iso1, DCLK1-AS) and short (DCLK1-iso4, DCLK1-BL) isoforms can efficiently activate EMT leading to tumor migration and invasion. Consistent with experiments in vitro, bioinformatic analysis demonstrates that DCLK1 may act as a driver of EMT activation in PDAC. Further analysis showed that EMT was associated with an immunosuppressive microenvironment, which includes more immunosuppressive cells and chemokines, and patients with a higher EMT score were less sensitive to immune checkpoint inhibitors according to the TIDE (Tumor Immune Dysfunction and Exclusion) algorithm. Multiplexed immunofluorescence results demonstrated the close correlation between DCLK1, EMT and immunosuppression in PDAC patients. The findings were further confirmed in vivo reflected by decreased CD4+, CD8+ T cells and increased M2 macrophages as well as E-cad loss in DCLK1-overexpressing subcutaneous tumors. Importantly, the highly-specific DCLK1 inhibitor (DCLK1-IN-1) was able to effectively block EMT process and restore T-cell activity. Altogether, our data demonstrate that DCLK1 is strongly associated with tumor immune escape in PDAC and inhibiting DCLK1 kinase activity may be a promising therapeutic modality.

10.
Front Oncol ; 11: 769727, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34926275

RESUMEN

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is the most common type of esophageal cancer and the seventh most prevalent cause of cancer-related death worldwide. Tumor microenvironment (TME) has been confirmed to play an crucial role in ESCC progression, prognosis, and the response to immunotherapy. There is a need for predictive biomarkers of TME-related processes to better prognosticate ESCC outcomes. AIM: To identify a novel gene signature linked with the TME to predict the prognosis of ESCC. METHODS: We calculated the immune/stromal scores of 95 ESCC samples from The Cancer Genome Atlas (TCGA) using the ESTIMATE algorithm, and identified differentially expressed genes (DEGs) between high and low immune/stromal score patients. The key prognostic genes were further analyzed by the intersection of protein-protein interaction (PPI) networks and univariate Cox regression analysis. Finally, a risk score model was constructed using multivariate Cox regression analysis. We evaluated the associations between the risk score model and immune infiltration via the CIBERSORT algorithm. Moreover, we validated the signature using the Gene Expression Omnibus (GEO) database. Within the ten gene signature, five rarely reported genes were further validated with quantitative real time polymerase chain reaction (qRT-PCR) using an ESCC tissue cDNA microarray. RESULTS: A total of 133 up-regulated genes were identified as DEGs. Ten prognostic genes were selected based on intersection analysis of univariate COX regression analysis and PPI, and consisted of C1QA, C1QB, C1QC, CD86, C3AR1, CSF1R, ITGB2, LCP2, SPI1, and TYROBP (HR>1, p<0.05). The expression of 9 of these genes in the tumor samples were significantly higher compared to matched adjacent normal tissue based on the GEO database (p<0.05). Next, we assessed the ability of the ten-gene signature to predict the overall survival of ESCC patients, and found that the high-risk group had significantly poorer outcomes compared to the low-risk group using univariate and multivariate analyses in the TCGA and GEO cohorts (HR=2.104, 95% confidence interval:1.343-3.295, p=0.001; HR=1.6915, 95% confidence interval:1.053-2.717, p=0.0297). Additionally, receiver operating characteristic (ROC) curve analysis demonstrated a relatively sensitive and specific profile for the signature (1-, 2-, 3-year AUC=0.672, 0.854, 0.81). To identify the basis for these differences in the TME, we performed correlation analyses and found a significant positive correlation with M1 and M2 macrophages and CD8+ T cells, as well as a strong correlation to M2 macrophage surface markers. A nomogram based on the risk score and select clinicopathologic characteristics was constructed to predict overall survival of ESCC patients. For validation, qRT-PCR of an ESCC patient cDNA microarray was performed, and demonstrated that C1QA, C3AR1, LCP2, SPI1, and TYROBP were up-regulated in tumor samples and predict poor prognosis. CONCLUSION: This study established and validated a novel 10-gene signature linked with M2 macrophages and poor prognosis in ESCC patients. Importantly, we identified C1QA, C3AR1, LCP2, SPI1, and TYROBP as novel M2 macrophage-correlated survival biomarkers. These findings may identify potential targets for therapy in ESCC patients.

11.
Mol Cancer Res ; 19(12): 1980-1991, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34610960

RESUMEN

Cancer stem cell (CSC) marker doublecortin-like kinase 1 (DCLK1) contributes greatly to the malignancy of gastrointestinal cancers, and DCLK1-targeted agents have potential therapeutic value. However, the molecular pathways regulated by DCLK1-S (DCLK1 isoform 4), a shortened splice variant of DCLK1, still remain obscure. Here we found that the expression of DCLK1-S is significantly increased in human esophageal squamous cell carcinoma (ESCC) tissues and associated with malignant progression and poor prognosis. Functional studies indicated that silencing total of DCLK1 mediated by CRISPR/Cas9 inhibited ESCC cell proliferation, migration, and invasion. Conversely, these changes were largely reversed after DCLK1-S rescue or overexpression. More importantly, DCLK1-S significantly enhanced primary tumor formation and metastatic lung colonization in vivo. The Cancer Genome Atlas database and molecular analysis showed that DCLK1-S was closely related to the epithelial-mesenchymal transition (EMT) process in patients with ESCC. Further RNA sequencing and Kyoto Encyclopedia of Genes and Genomes analysis demonstrated that MAPK signaling pathway was significantly enriched. Our in vitro study proclaimed that DCLK1-S induced MMP2 expression in ESCC cells via MAPK/ERK signaling, leading to the activation of EMT. In addition, administration of ERK1/2 blocker SCH772984 attenuated the proliferative and migratory phenotype induced by DCLK1-S. In conclusion, these findings suggest that DCLK1-S may be a key molecule in MAPK/ERK/MMP2 pathway-mediated progression of ESCC, and that it has potential as a biomarker or therapeutic target to improve outcomes in patients with ESCC. IMPLICATIONS: : DCLK1-S induces ESCC progression by activating the MAPK/ERK/MMP2 axis and may serve as a prognostic biomarker or therapeutic target for patients with ESCC.


Asunto(s)
Quinasas Similares a Doblecortina/metabolismo , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago/genética , Sistema de Señalización de MAP Quinasas/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Neoplasias Esofágicas/mortalidad , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/mortalidad , Carcinoma de Células Escamosas de Esófago/patología , Femenino , Humanos , Ratones , Ratones Desnudos , Persona de Mediana Edad , Análisis de Supervivencia
12.
Bioengineered ; 12(1): 7678-7693, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34550868

RESUMEN

Schisandrin A (SchA) has been reported to have good anti-cancer effects. However, its anti-cancer mechanism in breast cancer remains unknown. This study aimed to explore the mechanism of SchA in breast cancer treatment using bio-informatics analysis and in vitro experiments. The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), Gene Cards, and PharmMapper databases were used to screen the candidate targets of SchA against MDA-MB-231 cells selected as the tested cell line through MTT analysis. The functions and pathways of the targets were identified using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis and further analyzed using DAVID 6.8.1 database. Network pharmacology analysis revealed 77 candidate targets, 31 signal pathways, and 208 GO entries (P < 0.05). The targets regulated serine-type endopeptidase and protein tyrosine kinase activities, thereby promoting the migration and inhibiting the apoptosis of MDA-MB-231 cells. Comprehensive analysis of the 'Protein-Protein Interaction' (PPI) and 'Component-Targets-Pathways' (C-T-P) networks constructed using Cytoscape 3.7.1 software revealed four core targets: EGFR, PIK3R1, MMP9 and Caspase 3. Their docking scores with SchA were subsequently investigated through molecular docking. The wound healing, Hoechst 33342/PI, and western blot assays confirmed that SchA significantly down-regulated EGFR, PIK3R1, and MMP9, but up-regulated cleaved-caspase 3, thus inhibiting the migration and promoting the apoptosis of MDA-MB-231 cells. Reckoning the findings of the study, SchA is a potential adjuvant treatment for breast cancer.


Asunto(s)
Neoplasias de la Mama , Biología Computacional , Ciclooctanos , Lignanos , Compuestos Policíclicos , Mapas de Interacción de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama/química , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclooctanos/química , Ciclooctanos/metabolismo , Femenino , Humanos , Lignanos/química , Lignanos/metabolismo , Simulación del Acoplamiento Molecular , Farmacología en Red , Compuestos Policíclicos/química , Compuestos Policíclicos/metabolismo
13.
Altern Ther Health Med ; 27(6): 34-39, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32619205

RESUMEN

CONTEXT: Pulmonary hypertension (PH) is a complication of numerous pulmonary conditions. Previous studies have confirmed that Selaginella doederleini has pharmacological effects against many cancers, and triflavones have been newly isolated as one of its active ingredients, with antioxidant and antitumor activities. The chronic hypoxia model is one of the models most used to study PH pathogenesis and treatment. OBJECTIVE: The present study was designed to investigate the protective effects of triflavones from selaginella doederlleini against PH and on the proliferation and apoptosis of ASMCs in a hypoxia-induced PH model in rats. METHODS: The research team performed an animal study. SETTING: The study took place at the Tongji Medical College at the Huazhong University of Science and Technology in Wuhan, Hubei, PR China. ANIMALS: The animals were 40 specific pathogen free (SPF), male SD rats weighing 200 ± 20 g each. INTERVENTION: The animals were divided into 4 groups, with 10 animals in each group: (1) the control group, (3) the hypoxia group (PH group), (3) the control + triflavones group (Tri group) and (4) the hypoxia + triflavones group (PH + Tri group). The rats in 2 hypoxia groups were exposed to 10% oxygen to induce PH, and the animals in the 2 control groups were exposed to room air, both for 3 consecutive weeks. Animals in the 2 triflavones groups were injected with 200 µL of triflavones-100 mg/mL dissolved in 0.9% normal saline-and the animals in the control and PH groups were injected with 200 µL of 0.9% normal saline. OUTCOME MEASURES: In vitro, the primary aorta smooth muscle cells (ASMCs) were isolated, and the proliferation and apoptosis of the ASMCs were assayed by CCK-8 kit and flow cytometry. The expression levels of the alpha-smooth muscle actin (α-SMA), transforming growth factor beta 1 (TGF-ß1), and phosphoinositide 3-kinases (P13K)/ protein kinase B (Akt) in the ASMCs were also assayed by Western blot. RESULTS: Triflavones effectively decreased mPAP, the ratio of RV/ (LV + S), and the thickness of the arteries of the PH + Tri group. Furthermore, triflavones reversed the increased proliferation and inhibited apoptosis induced by chronic hypoxia for that group. Hypoxia increased TGF-ß1 protein expression and the activation of P13K/Akt, as shown in the PH group, and was abrogated by the triflavones. CONCLUSION: Triflavones are promising protective agents against PH due to their inhibitory effects on vascular remodeling through P13K/Akt signaling.


Asunto(s)
Flavonas/farmacología , Hipoxia , Selaginellaceae , Remodelación Vascular/efectos de los fármacos , Animales , Proliferación Celular , Hipoxia/tratamiento farmacológico , Masculino , Fosfatidilinositol 3-Quinasas , Fitoquímicos/farmacología , Proteínas Proto-Oncogénicas c-akt , Arteria Pulmonar , Ratas , Ratas Sprague-Dawley , Selaginellaceae/química
14.
Clin Exp Pharmacol Physiol ; 47(2): 220-226, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31587339

RESUMEN

Hypertension is a common type of cardiovascular disease that remains a major cause of death in the world. Vascular remodelling is an important complication of hypertension, and vascular smooth muscle cells (VSMCs) play a major role in vascular remodelling. Sauchinone is one of the active lignins which has been found to possess vascular protective effects. However, the functional role of sauchinone in hypertension has not been investigated. The aim of this study was to evaluate the role of sauchinone in the angiotensin II (Ang II)-induced vascular remodelling model in VSMCs. The results showed that treatment of sauchinone inhibited Ang II-induced VSMCs proliferation and migration in VSMCs. Sauchinone treatment suppressed the reactive oxygen species (ROS) production and NADPH oxidase (NOX) activity in Ang II-induced VSMCs. The inhibitory effects of Ang II on expressions of VSMCs phenotype markers including α-smooth muscle actin (α-SMA), calponin, osteopontin were mitigated by sauchinone treatment. Furthermore, sauchinone inhibited Ang II-induced over-activation of TGF-ß1/Smad3 signalling pathway in VSMCs. Taken together, this study identified sauchinone as a potential agent for preventing vascular remodelling in hypertension.


Asunto(s)
Angiotensina II/farmacología , Benzopiranos/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dioxoles/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/fisiología
15.
J Transl Med ; 17(1): 393, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31775789

RESUMEN

BACKGROUND: RH1 is one of the most clinically important blood group antigens in the field of transfusion and in the prevention of fetal incompatibility. The molecular analysis and characterization of serologic weak D phenotypes is essential to ensuring transfusion safety. METHODS: Blood samples from a northeastern Chinese population were randomly screened for a serologic weak D phenotype. The nucleotide sequences of all 10 exons, adjacent flanking intronic regions, and partial 5' and 3' untranslated regions (UTRs) were detected for RHD genes. Predicted deleterious structural changes in missense mutations of serologicl weak D phenotypes were analyzed using SIFT, PROVEAN and PolyPhen2 software. The protein structure of serologic weak D phenotypes was predicted using Swiss-PdbViewer 4.0.1. RESULTS: A serologic weak D phenotype was found in 45 individuals (0.03%) among 132,479 blood donors. Seventeen distinct RHD mutation alleles were detected, with 11 weak D, four partial D and two DEL alleles. Further analyses resulted in the identification of two novel alleles (RHD weak D 1102A and 399C). The prediction of a three-dimensional structure showed that the protein conformation was disrupted in 16 serologic weak D phenotypes. CONCLUSIONS: Two novel and 15 rare RHD alleles were identified. Weak D type 15, DVI Type 3, and RHD1227A were the most prevalent D variant alleles in a northeastern Chinese population. Although the frequencies of the D variant alleles presented herein were low, their phenotypic and genotypic descriptions add to the repertoire of reported RHD alleles. Bioinformatics analysis on RhD protein can give us more interpretation of missense variants of RHD gene.


Asunto(s)
Donantes de Sangre , Biología Computacional , Sistema del Grupo Sanguíneo Rh-Hr/genética , Pruebas Serológicas , Alelos , Sustitución de Aminoácidos/genética , China , Frecuencia de los Genes/genética , Humanos , Proteínas Mutantes/química , Fenotipo , Estructura Terciaria de Proteína , Sistema del Grupo Sanguíneo Rh-Hr/química
16.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 24(5): 518-9, 2007 Oct.
Artículo en Chino | MEDLINE | ID: mdl-17922417

RESUMEN

OBJECTIVE: To identify a novel HLA allele in Chinese population. METHODS: A new HLA-B allele was initially detected by an usual PCR-SSP and PCR-SSOP in routine typing HLA allele. Sequence-based typing (SBT) was used to identify and analysis the difference between the new allele and HLA-B 4409 allele. RESULTS: The HLA-B exon 3 nucleotide sequence of the novel allele was different from all other known alleles. The allele had 3 nucleotides replaced of the closest matching B 4409 allele at nt538(G>C), nt539(A>T) and nt540 (C>G) in exon 3, resulting in an amino acid change from D(GAC) to L(CTG) at codon 180. CONCLUSION: A novel HLA allele was confirmed by the SBT and it was officially designated as HLA-B 4446 by WHO Nomenclature Committee for Factors of the HLA System in September,2005.


Asunto(s)
Alelos , Antígenos HLA-B/clasificación , Antígenos HLA-B/genética , Secuencia de Bases , Humanos , Masculino , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...