Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Chin J Cancer Res ; 36(3): 298-305, 2024 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-38988482

RESUMEN

Objective: Nucleotide excision repair (NER) plays a vital role in maintaining genome stability, and the effect of NER gene polymorphisms on hepatoblastoma susceptibility is still under investigation. This study aimed to evaluate the relationship between NER gene polymorphisms and the risk of hepatoblastoma in Eastern Chinese Han children. Methods: In this five-center case-control study, we enrolled 966 subjects from East China (193 hepatoblastoma patients and 773 healthy controls). The TaqMan method was used to genotype 19 single nucleotide polymorphisms (SNPs) in NER pathway genes, including ERCC1, XPA, XPC, XPD, XPF, and XPG. Then, multivariate logistic regression analysis was performed, and odds ratios (ORs) and 95% confidence intervals (95% CIs) were utilized to assess the strength of associations. Results: Three SNPs were related to hepatoblastoma risk. XPC rs2229090 and XPD rs3810366 significantly contributed to hepatoblastoma risk according to the dominant model (adjusted OR=1.49, 95% CI=1.07-2.08, P=0.019; adjusted OR=1.66, 95% CI=1.12-2.45, P=0.012, respectively). However, XPD rs238406 conferred a significantly decreased risk of hepatoblastoma under the dominant model (adjusted OR=0.68, 95% CI=0.49-0.95; P=0.024). Stratified analysis demonstrated that these significant associations were more prominent in certain subgroups. Moreover, there was evidence of functional implications of these significant SNPs suggested by online expression quantitative trait loci (eQTLs) and splicing quantitative trait loci (sQTLs) analysis. Conclusions: In summary, NER pathway gene polymorphisms (XPC rs2229090, XPD rs3810366, and XPD rs238406) are significantly associated with hepatoblastoma risk, and further research is required to verify these findings.

2.
Mol Carcinog ; 63(8): 1559-1571, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38780126

RESUMEN

The therapeutic effect of anlotinib on neuroblastoma is still not fully understood. This study aims to explore the differentiation therapeutic effects of anlotinib on neuroblastoma and its potential association with the neural development regulatory protein collapsin response mediator protein 5 (CRMP5), both in vivo and in vitro. A patient-derived xenograft (PDX) model was established to observe the therapeutic effect of anlotinib. Neuroblastoma cell lines SK-N-SH and SK-N-AS were cultured to observe the morphological impact of anlotinib. Transwell assay was used to evaluate the cell invasion, and Western blot analysis and immunohistochemistry were employed to detect the expressions of neuronal differentiation-related proteins. Results indicate that anlotinib effectively inhibited tumor growth in the PDX model, modulated the expressions of neuronal differentiation markers. In vitro, anlotinib treatment induced neurite outgrowth in neuroblastoma cells and inhibited their invasive ability, reflecting a change in neuronal marker expression patterns consistent with the PDX model. Similarly, in the SK-N-AS mouse xenograft model, anlotinib demonstrated comparable tumor-suppressing effects and promoted neuronal-like differentiation. Additionally, anlotinib significantly downregulated CRMP5 expression in neuroblastoma both in vivo and in vitro. Overexpression of CRMP5 significantly reversed the differentiation therapy effect of anlotinib, exacerbating the aggressiveness and reducing the differentiation level of neuroblastoma. These findings highlight the potential of anlotinib as an anti-neuroblastoma agent. It may suppress tumor proliferation and invasion by promoting the differentiation of tumor cells towards a neuronal-like state, and this differentiation therapy effect involves the inhibition of CRMP5 signaling.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Indoles , Proteínas del Tejido Nervioso , Neuroblastoma , Quinolinas , Ensayos Antitumor por Modelo de Xenoinjerto , Humanos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/patología , Neuroblastoma/metabolismo , Neuroblastoma/genética , Animales , Ratones , Quinolinas/farmacología , Diferenciación Celular/efectos de los fármacos , Indoles/farmacología , Línea Celular Tumoral , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Ratones Desnudos , Hidrolasas/genética , Hidrolasas/metabolismo , Antineoplásicos/farmacología , Proteínas Asociadas a Microtúbulos
3.
PLoS One ; 19(4): e0300548, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38578740

RESUMEN

Biomechanical cue within the tissue microenvironment is known to play a critical role in regulating cell behaviors and maintaining tissue homeostasis. As hydrostatic pressure often increases in biliary system under pathological states, we investigated the effect of the moderate elevation of the hydrostatic pressure on biliary epithelial cells, especially on the epithelial-mesenchymal transition (EMT). Human intrahepatic biliary epithelial cells were loaded to hydrostatic pressure using a commercial device. We found that loading the cells to 50 mmHg hydrostatic pressure induced obvious morphological changes and significantly upregulated vimentin, ZEB1, and pSmad2/3, fibronectin, and collagen 1α. All changes induced by hydrostatic pressure loading were effectively mitigated by either ROCK inhibitor (Y-27632) or ALK5 inhibitor (SB-431542). Our in vitro experimental data suggests that hydrostatic pressure loading induces EMT of cholangiocytes through RhoA/ROCK and TGF-ß/Smad pathways. Elevated hydrostatic pressure in biliary duct system under pathological states may promote the biliary epithelial cells shifting to profibrotic and mesenchymal characteristics.


Asunto(s)
Transducción de Señal , Factor de Crecimiento Transformador beta , Humanos , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Presión Hidrostática , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
4.
Clin Case Rep ; 12(3): e8589, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38449897

RESUMEN

This was the first article reported a fatal case of chlorfenapyr poisoning in a child, and the typical symptoms before death include high fever, severe sweating, coma, and limb stiffness, and elevation of myocardial enzymes and myoglobin; neurological symptoms tend to appear earlier in children than in adults.

5.
Drug Dev Res ; 85(1): e22140, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38349263

RESUMEN

Everolimus, a known inhibitor of the mammalian target of rapamycin (mTOR), has shown uncertain efficacy in treating hepatoblastoma. This study delves into the potential anti-hepatoblastoma properties of everolimus and its intricate relationship with autophagy and ferroptosis, both in vitro and in vivo. In vivo, tumor tissue from hepatoblastoma patient and human hepatoblastoma cell line HuH-6 were xenografted into nude mice to establish xenograft models for observing the effect of everolimus on tumor growth. In vitro, HuH-6 cells were cultured to evaluate the anti-hepatoblastoma activity of everolimus. Transmission electron microscopy and microtubule-associated proteins 1 light chain 3 (LC3), beclin 1, and p62 protein expressions were employed to investigate autophagy. Additionally, indicators of cell apoptosis, reactive oxygen species (ROS) and proteins associated with ferroptosis were measured to evaluate ferroptosis. The results demonstrate that everolimus treatment effectively induced the formation of autophagosomes in hepatoblastoma cells, upregulated the LC3II/I ratio and beclin 1 expression, and downregulated p62 expression, indicating an enhanced autophagy level both in vitro and in vivo. Furthermore, everolimus treatment induced cell apoptosis, increased ROS level, elevated concentrations of malondialdehyde, 4-hydroxynonenal, and iron content, while reducing the ratio of glutathione/oxidized glutathione, and downregulating the protein expression of glutathione peroxidase 4 and solute carrier family 7 member 11, suggesting its ability to induce ferroptosis in hepatoblastoma cells. Importantly, the induction of ferroptosis by everolimus was significantly reversed in the presence of autophinib, an autophagy inhibitor, indicating the autophagy-dependent of everolimus-induced ferroptosis. Taken together, these findings suggest that everolimus holds promise as an effective anti-hepatoblastoma drug, with its mechanism of action potentially involving the induction of autophagy-dependent ferroptosis in hepatoblastoma cells.


Asunto(s)
Ferroptosis , Hepatoblastoma , Neoplasias Hepáticas , Animales , Ratones , Humanos , Everolimus/farmacología , Hepatoblastoma/tratamiento farmacológico , Beclina-1 , Ratones Desnudos , Especies Reactivas de Oxígeno , Autofagia , Neoplasias Hepáticas/tratamiento farmacológico , Mamíferos
6.
Artículo en Inglés | MEDLINE | ID: mdl-38218563

RESUMEN

Four tyrosine kinase inhibitors, alectinib, apatinib, lenvatinib and anlotinib, have been shown to be effective in the treatment of clinical tumors, but their cardiac risks have also raised concerns. In this study, zebrafish embryos at 6 h post fertilization (hpf) were exposed to the four drugs at concentrations of 0.05-0.2 mg/L until 72 hpf, and then the development of these embryos was quantified, including heart rate, body length, yolk sac area, pericardial area, distance between venous sinus and balloon arteriosus (SV-BA), separation of cardiac myocytes and endocardium, gene expression, vascular development and oxidative stress. At the same exposure concentrations, alectinib and apatinib had little effect on the cardiac development of zebrafish embryos, while lenvatinib and anlotinib could induce significant cardiotoxicity and developmental toxicity, including shortened of body length, delayed absorption of yolk sac, pericardial edema, prolonged SV-BA distance, separation of cardiomyocytes and endocardial cells, and downregulation of key genes for heart development. Heart rate decreased in all four drug treatment groups. In terms of vascular development, alectinib and apatinib did not inhibit the growth of embryonic intersegmental vessels (ISVs) and retinal vessels, while lenvatinib and anlotinib caused serious vascular toxicity, and the inhibition of anlotinib in vascular development was more obvious. Besides, the level of reactive oxygen species (ROS) in the lenvatinib and anlotinib treatment groups was significantly increased. Our results provide reference for comparing the cardiotoxicity of the four drugs.


Asunto(s)
Carbazoles , Cardiotoxicidad , Indoles , Compuestos de Fenilurea , Piperidinas , Piridinas , Quinolinas , Pez Cebra , Animales , Cardiotoxicidad/metabolismo , Embrión no Mamífero
7.
Toxicology ; 503: 153735, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38272385

RESUMEN

Dimethyl fumarate (DMF) is an immunosuppressant commonly used to treat multiple sclerosis and other autoimmune diseases. Despite known side effects such as lymphopenia, the effect of DMF on cardiac development remains unclear. To assess this, we used zebrafish to evaluate the cardiac developmental toxicity of DMF. Our study showed that DMF reduced the survival rate of zebrafish embryos, with those exposed to 1, 1.3, and 1.6 mg/L exhibiting heart rate reduction, shortened body length, delayed yolk sac absorption, pericardial edema, increased distance from sinus venous to bulbus arteriosus, and separation of cardiomyocytes and endocardial cells at 72 hpf. Heart development-related genes showed disorder, apoptosis-related genes were up-regulated, and the oxidative stress response was down-regulated. Treatment with cysteamine ameliorated the heart development defects. Our study demonstrates that DMF induces cardiac developmental toxicity in zebrafish, possibly by down-regulating oxidative stress responses. This study provides a certain research basis for further study of DMF-induced cardiac developmental toxicity, and provides some experimental evidence for future clinical application and study of DMF.


Asunto(s)
Cardiopatías Congénitas , Pez Cebra , Animales , Pez Cebra/fisiología , Dimetilfumarato/toxicidad , Dimetilfumarato/metabolismo , Regulación hacia Abajo , Embrión no Mamífero , Estrés Oxidativo , Cardiotoxicidad/metabolismo
8.
Mol Cancer Ther ; 23(5): 721-732, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38295302

RESUMEN

In this study, we explored the therapeutic potential of everolimus, an mTOR inhibitor, in a patient-derived xenograft (PDX) of rhabdomyosarcoma, the most prevalent malignant pediatric sarcoma. In addition, rhabdoid tumor cell line A-204 and Ewings sarcoma cell line A-673 were cultured to assess the in vitro effect of everolimus. Furthermore, the cell-derived xenograft (CDX) of A-673 was established and treated with everolimus in vivo. IHC and Western blotting were performed to detect the expressions of pertinent proteins. Results showed that everolimus intervention had limited inhibitory effect on PDX tumor growth compared with cyclophosphamide. Nevertheless, everolimus treatment significantly influenced the phosphorylation levels of S6 kinase beta 1 (S6K1) and eIF4E-binding protein 1 (p-4E-BP1), resulting in the inhibition of angiogenesis in vitro and in vivo. Interestingly, everolimus led to an upregulation in the level of IL17A in sarcoma cells. Notably, when secukinumab, a mAb of IL17A, was combined with everolimus, it synergistically enhanced the inhibitory effect of everolimus on sarcoma cell proliferation in vitro and on the growth of PDX or CDX xenograft tumors in vivo. Importantly, this combination therapy did not affect the mTOR signaling. These results indicate that everolimus exerts an antipediatric sarcoma effect by inhibiting mTOR signal. However, everolimus induces sarcoma cells to produce IL17A, which promotes tumor cell survival and counteracts its antipediatric sarcoma effect. The combination of secukinumab effectively eliminates the effects of IL17A, thereby improving the therapeutic efficacy of everolimus in the context of pediatric sarcomas.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Proliferación Celular , Everolimus , Interleucina-17 , Ensayos Antitumor por Modelo de Xenoinjerto , Everolimus/farmacología , Humanos , Animales , Ratones , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Interleucina-17/metabolismo , Interleucina-17/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/patología , Sarcoma/tratamiento farmacológico , Sarcoma/patología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Sinergismo Farmacológico
9.
Ecotoxicol Environ Saf ; 270: 115911, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38181604

RESUMEN

Iprodione is an effective and broad-spectrum fungicide commonly used for early disease control in fruit trees and vegetables. Due to rainfall, iprodione often finds its way into water bodies, posing toxicity risks to non-target organisms and potentially entering the human food chain. However, there is limited information available regarding the developmental toxicity of iprodione specifically on the liver in existing literature. In this study, we employed larval and adult zebrafish as models to investigate the toxicity of iprodione. Our findings revealed that iprodione exposure led to yolk sac edema and increased mortality in zebrafish. Notably, iprodione exhibited specific effects on zebrafish liver development. Additionally, zebrafish exposed to iprodione experienced an overload of reactive oxygen species, resulting in the upregulation of p53 gene expression. This, in turn, triggered hepatocyte apoptosis and disrupted carbohydrate/lipid metabolism as well as energy demand systems. These results demonstrated the substantial impact of iprodione on zebrafish liver development and function. Furthermore, the application of astaxanthin (an antioxidant) and p53 morpholino partially mitigated the liver toxicity caused by iprodione. To summarize, iprodione induces apoptosis through the upregulation of p53 mediated by oxidative stress signals, leading to liver toxicity in zebrafish. Our study highlights that exposure to iprodione can result in hepatotoxicity in zebrafish, and it may potentially pose toxicity risks to other aquatic organisms and even humans.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Enfermedad Hepática Inducida por Sustancias y Drogas , Hidantoínas , Pez Cebra , Animales , Humanos , Pez Cebra/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Estrés Oxidativo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Embrión no Mamífero/metabolismo , Apoptosis
10.
Fish Physiol Biochem ; 50(2): 403-412, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38085449

RESUMEN

BPA is so ubiquitous that 27 million tons of BPA-containing plastic, including mineral water bottles and baby bottles, is produced worldwide each year. The potential toxicity of BPA to humans and aquatic organisms has been the subject of intense research. In this study, a zebrafish model system was used to assess BPA-mediated hepatotoxicity. Zebrafish larvae at 72-144 hpf were exposed to BPA at different concentrations (0,1, 3 and 5mg/L). For example, BPA-treated zebrafish larvae showed increased mortality, delayed uptake of nutrients in yolk sac, shortened body length, smaller liver area, abnormal expression of genes related to liver development, and pathological changes in the liver tissue. Mechanistically, BPA exposure induced excessive oxidative stress in the liver of zebrafish and increased the level of hepatocyte apoptosis in zebrafish larvae, and the antioxidant astaxanthin could rescue the BPA-mediated liver toxicity.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Contaminantes Químicos del Agua , Humanos , Animales , Pez Cebra/genética , Compuestos de Bencidrilo/toxicidad , Fenoles/toxicidad , Contaminantes Químicos del Agua/toxicidad , Estrés Oxidativo , Larva , Apoptosis
11.
Cancer Chemother Pharmacol ; 93(4): 381-391, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38148335

RESUMEN

BACKGROUND: Burkitt's lymphoma, one of the most common subtypes of pediatric malignant lymphoma, is notorious for its swift onset, aggressive proliferation, pronounced invasiveness, and marked malignancy. The therapeutic landscape for Burkitt's lymphoma currently falls short of providing universally effective and tolerable solutions. Andrographolide, a primary active component of Andrographis paniculata, is renowned for its properties of heat-clearing, detoxification, inflammation reduction, and pain relief. It is predominantly used in treating bacterial and viral infections of the upper respiratory tract, as well as dysentery. Various reports highlight the antitumor effects of andrographolide. Yet, its specific impact and the underlying mechanism of action on Burkitt's lymphoma remain an uncharted area of research. METHOD: We employed network pharmacology to pinpoint the targets of andrographolide's action on Burkitt's lymphoma and the associated pathways. We then evaluated the impact of andrographolide on Burkitt's lymphoma using both in vitro and in vivo patient-derived xenograft (PDX) models. Concurrently, we confirmed the molecular targets of andrographolide in Burkitt's lymphoma through immunofluorescence assays. RESULT: Utilizing network pharmacology, we identified 15 relevant targets, 60 interrelationships between these targets, and numerous associated signaling pathways for andrographolide's action on Burkitt's lymphoma. In vitro efficacy tests using High-throughput Drug Sensitivity Testing and in vivo PDX model evaluations revealed that andrographolide effectively curtailed the growth of Burkitt's lymphoma. Moreover, we observed a increased in the expression of JUN (c-Jun) and CASP3 (Caspase 3) proteins in Burkitt's lymphoma cells treated with andrographolide. CONCLUSION: Andrographolide inhibits the growth of Burkitt's lymphoma by inhibiting JUN and CASP3 proteins.


Asunto(s)
Linfoma de Burkitt , Diterpenos , Humanos , Niño , Linfoma de Burkitt/tratamiento farmacológico , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/patología , Caspasa 3
12.
Math Biosci Eng ; 20(11): 19133-19151, 2023 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-38052593

RESUMEN

Malignancies such as bladder urothelial carcinoma, colon adenocarcinoma, liver hepatocellular carcinoma, lung adenocarcinoma and prostate adenocarcinoma significantly impact men's well-being. Accurate cancer classification is vital in determining treatment strategies and improving patient prognosis. This study introduced an innovative method that utilizes gene selection from high-dimensional datasets to enhance the performance of the male tumor classification algorithm. The method assesses the reliability of DNA methylation data to distinguish the five most prevalent types of male cancers from normal tissues by employing DNA methylation 450K data obtained from The Cancer Genome Atlas (TCGA) database. First, the chi-square test is used for dimensionality reduction and second, L1 penalized logistic regression is used for feature selection. Furthermore, the stacking ensemble learning technique was employed to integrate seven common multiclassification models. Experimental results demonstrated that the ensemble learning model utilizing multiple classification models outperformed any base classification model. The proposed ensemble model achieved an astonishing overall accuracy (ACC) of 99.2% in independent testing data. Moreover, it may present novel ideas and pathways for the early detection and treatment of future diseases.


Asunto(s)
Adenocarcinoma , Carcinoma Hepatocelular , Carcinoma de Células Transicionales , Neoplasias del Colon , Neoplasias Hepáticas , Neoplasias Pulmonares , Neoplasias de la Vejiga Urinaria , Humanos , Masculino , Metilación de ADN , Adenocarcinoma/genética , Carcinoma de Células Transicionales/genética , Reproducibilidad de los Resultados , Neoplasias de la Vejiga Urinaria/genética , Neoplasias del Colon/genética , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/genética , Neoplasias Pulmonares/genética , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética
13.
Math Biosci Eng ; 20(11): 19320-19340, 2023 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-38052602

RESUMEN

The RNA secondary structure is like a blueprint that holds the key to unlocking the mysteries of RNA function and 3D structure. It serves as a crucial foundation for investigating the complex world of RNA, making it an indispensable component of research in this exciting field. However, pseudoknots cannot be accurately predicted by conventional prediction methods based on free energy minimization, which results in a performance bottleneck. To this end, we propose a deep learning-based method called TransUFold to train directly on RNA data annotated with structure information. It employs an encoder-decoder network architecture, named Vision Transformer, to extract long-range interactions in RNA sequences and utilizes convolutions with lateral connections to supplement short-range interactions. Then, a post-processing program is designed to constrain the model's output to produce realistic and effective RNA secondary structures, including pseudoknots. After training TransUFold on benchmark datasets, we outperform other methods in test data on the same family. Additionally, we achieve better results on longer sequences up to 1600 nt, demonstrating the outstanding performance of Vision Transformer in extracting long-range interactions in RNA sequences. Finally, our analysis indicates that TransUFold produces effective pseudoknot structures in long sequences. As more high-quality RNA structures become available, deep learning-based prediction methods like Vision Transformer can exhibit better performance.


Asunto(s)
Algoritmos , ARN , Conformación de Ácido Nucleico , ARN/genética , ARN/química , Análisis de Secuencia de ARN/métodos , Secuencia de Bases
14.
Int J Rheum Dis ; 26(12): 2534-2542, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37905746

RESUMEN

OBJECTIVE: This study aims to construct an artificial intelligence (AI) model capable of effectively discriminating between abdominal Henoch-Schönlein purpura (AHSP) and acute appendicitis (AA) in pediatric patients. METHODS: A total of 6965 participants, comprising 2201 individuals with AHSP and 4764 patients with AA, were enrolled in the study. Additionally, 53 laboratory indicators were taken into consideration. Five distinct artificial intelligence (AI) models were developed employing machine learning algorithms, namely XGBoost, AdaBoost, Gaussian Naïve Bayes (GNB), MLPClassifier (MLP), and support vector machine (SVM). The performance of these prediction models was assessed through receiver operating characteristic (ROC) curve analysis, calibration curve assessment, and decision curve analysis (DCA). RESULTS: We identified 32 discriminative indicators (p < .05) between AHSP and AA. Five indicators, namely the lymphocyte ratio (LYMPH ratio), eosinophil ratio (EO ratio), eosinophil count (EO count), neutrophil ratio (NEUT ratio), and C-reactive protein (CRP), exhibited strong performance in distinguishing AHSP from AA (AUC ≥ 0.80). Among the various prediction models, the XGBoost model displayed superior performance evidenced by the highest AUC (XGBoost = 0.895, other models < 0.89), accuracy (XGBoost = 0.824, other models < 0.81), and Kappa value (XGBoost = 0.621, other models < 0.60) in the validation set. After optimization, the XGBoost model demonstrated remarkable diagnostic performance for AHSP and AA (AUC > 0.95). Both the calibration curve and decision curve analysis suggested the promising clinical utility and net benefits of the XGBoost model. CONCLUSION: The AI-based machine learning model exhibits high prediction accuracy and can differentiate AHSP and AA from a data-driven perspective.


Asunto(s)
Apendicitis , Vasculitis por IgA , Humanos , Niño , Inteligencia Artificial , Vasculitis por IgA/diagnóstico , Apendicitis/diagnóstico , Apendicitis/etiología , Teorema de Bayes , Aprendizaje Automático , Proteínas Sanguíneas , Chaperonas Moleculares
15.
Photodiagnosis Photodyn Ther ; 44: 103846, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37838234

RESUMEN

In recent years, it has been reported that indocyanine green can be used for intraoperative navigation in Kasai surgery. However, there are no reports of its application in surgery for rare type II cystic biliary atresia. We report a girl presented with jaundice and light-colored stools. Laboratory tests showed impaired liver function with elevated serum bilirubin and bile acid levels. The abdominal ultrasound and MRCP suggested a common hepatic duct cyst. A diagnosis of choledochal cyst was suspected and biliary atresia could not be excluded. Conservative treatment was unsatisfactory. Laparoscopic exploration with indocyanine green fluorescence was performed on the 38th day of her life, and intraoperative diagnosis of type II CBA was made because the common hepatic duct cyst and its downstream anatomical structures did not show fluorescence. The postoperative bilirubin and bile acid levels decreased significantly and she was discharged two weeks after surgery. This result suggests that indocyanine green can be safely used in laparoscopic surgery for type II CBA, which not only helps in the differential diagnosis of CBA and choledochal cyst, but also confirms bile flow in real time.


Asunto(s)
Atresia Biliar , Quiste del Colédoco , Laparoscopía , Fotoquimioterapia , Humanos , Femenino , Atresia Biliar/diagnóstico por imagen , Atresia Biliar/cirugía , Verde de Indocianina , Quiste del Colédoco/diagnóstico por imagen , Quiste del Colédoco/cirugía , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes , Laparoscopía/métodos , Imagen Óptica , Bilirrubina , Ácidos y Sales Biliares
16.
Oncogene ; 42(44): 3260-3273, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37740007

RESUMEN

Pancreatic cancer cells undergo intricate metabolic reprogramming to sustain their survival and proliferation. p53 exhibits a dual role in tumor cell ferroptosis. However, the precise role and mechanisms underlying wild-type p53 activation in promoting ferroptosis in pancreatic cancer cells remain obscure. In this study, we applied bioinformatics tools and performed an analysis of clinical tissue sample databases and observed a significantly upregulated expression of solute carrier family 35 member F2 (SLC35F2) in pancreatic cancer tissues. Our clinical investigations indicated that elevated SLC35F expression was related to adverse survival outcomes. Through multi-omics analyses, we discerned that SLC35F2 influences the transcriptome and inhibits ferroptosis in pancreatic cancer cells. Moreover, our findings reveal the pivotal involvement of p53 in mediating SLC35F2-mediated ferroptosis, both in vitro and in vivo. SLC35F2 inhibits ferroptosis by facilitating TRIM59-mediated p53 degradation. Further mechanistic investigations demonstrated that SLC35F2 competitively interacts with the E3 ubiquitin ligase SYVN1 of TRIM59, thereby stabilizing TRIM59 expression and consequentially promoting p53 degradation. Utilizing protein 3D structure analysis and drug screening, we identified irinotecan hydrochloride and lapatinib ditosylate as compounds targeting SLC35F2, augmenting the antitumor effect of imidazole ketone erastin (IKE) in a wild-type p53 patient-derived xenograft (PDX) model. However, in the p53 mutant PDX model, irinotecan hydrochloride and lapatinib ditosylate did not alter the sensitivity of the tumor xenograft model to IKE-triggered ferroptosis. In summary, our work establishes a novel mechanism wherein the SLC35F2-SYVN1-TRIM59 axis critically regulates ferroptosis of pancreatic cancer cells by inhibiting endogenous p53. Thus, SLC35F2 emerges as a promising therapeutic target for treating pancreatic cancer.


Asunto(s)
Ferroptosis , Neoplasias Pancreáticas , Humanos , Ferroptosis/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Irinotecán/farmacología , Lapatinib/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Línea Celular Tumoral , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Neoplasias Pancreáticas
17.
Environ Toxicol ; 38(11): 2679-2690, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37551640

RESUMEN

Apatinib, a small-molecule VEGFR2-tyrosine kinase inhibitor, has shown potent anticancer activity in various clinical cancer treatments, but also different adverse reactions. Therefore, it is necessary to study its potential toxicity and working mechanism. We used zebrafish to investigate the effects of apatinib on the development of embryos. Zebrafish exposed to 2.5, 5, and 10 µM apatinib showed adverse effects such as decreased liver area, pericardial oedema, slow yolk absorption, bladder atrophy, and body length shortening. At the same time, it leads to abnormal liver tissue structure, liver function and related gene expression. Furthermore, after exposure to apatinib, oxidative stress levels were significantly elevated but liver developmental toxicity was effectively ameliorated with oxidative stress inhibitor treatment. Apatinib induces down-regulation of key target genes of Wnt signaling pathway in zebrafish, and it is found that Wnt activator can significantly rescue liver developmental defects. These results suggest that apatinib may induce zebrafish hepatotoxicity by inhibiting the Wnt signaling pathway and up-regulating oxidative stress, helping to strengthen our understanding of rational clinical application of apatinib.

18.
Respir Res ; 24(1): 127, 2023 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-37165378

RESUMEN

BACKGROUND: Congenital pulmonary airway malformation (CPAM) is the most frequent pulmonary developmental malformation and the pathophysiology remains poorly understood. This study aimed to identify the characteristic gene expression patterns and the marker genes essential to CPAM. METHODS: Tissues from the cystic area displaying CPAM and the area of normal appearance were obtained during surgery. Bulk RNA sequencing (RNA-seq) and single-cell RNA sequencing (scRNA-seq) were performed for integrating analysis. Iterative weighted gene correlation network analysis (iWGCNA) was used to identify specifically expressed genes to CPAM. RESULTS: In total, 2074 genes were significantly differentially expressed between the CPAM and control areas. Of these differentially expressed genes (DEGs), 1675 genes were up-regulated and 399 genes were down-regulated. Gene ontology analysis revealed these DEGs were specifically enriched in ciliated epithelium and involved in immune response. We also identified several CPAM-related modules by iWGCNA, among them, P15_I4_M3 module was the most influential module for distinguishing CPAMs from controls. By combining the analysis of the expression dataset from RNA-seq and scRNA-seq, SPOCK2, STX11, and ZNF331 were highlighted in CPAM. CONCLUSIONS: Through our analysis of expression datasets from both scRNA-seq and bulk RNA-seq of tissues obtained from patients with CPAM, we identified the characteristic gene expression patterns associated with the condition. Our findings suggest that SPOCK2 could be a potential biomarker gene for the diagnosis and therapeutic target in the development of CPAM, whereas STX11 and ZNF331 might serve as prognostic markers for this condition. Further investigations with larger samples and function studies are necessary to confirm the involvement of these genes in CPAM.


Asunto(s)
Malformación Adenomatoide Quística Congénita del Pulmón , Humanos , Malformación Adenomatoide Quística Congénita del Pulmón/metabolismo , Pulmón/metabolismo , Biomarcadores/metabolismo , Epitelio/metabolismo , ARN/genética , ARN/metabolismo , Proteoglicanos
19.
Front Med (Lausanne) ; 10: 1132630, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37138757

RESUMEN

The manifestation of severe pneumonia is only occasional, and pneumomediastinum is a condition that occurs rarely in Coronavirus disease 2019 (COVID-19) patients, especially in those patients who are infected with the Omicron variant. In addition, whether severe pneumonia or pneumomediastinum often occurs in patients in older age, in poor physical condition, or with underlying diseases remains to be ascertained. To date, severe pneumonia and pneumomediastinum due to Omicron infection had not been reported in a young patient with an excellent physical condition. In this study, we report such a case with the aforementioned manifestations in a robust adolescent infected with Omicron BA.5.2.

20.
Biochem Biophys Res Commun ; 667: 104-110, 2023 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-37210870

RESUMEN

Biomechanical forces are known to regulate the biological behaviors of cells. Although negative pressure has been used for wound healing, it is still unknown about its role in regulating cell plasticity. We investigated whether negative pressure could induce the dedifferentiation of hepatocytes. Using a commercial device, we found that the exposure of primary human hepatocytes to -50 mmHg quickly induced the formation of stress fibers and obviously changed cell morphology in 72 h. Moreover, the exposure of hepatocytes to -50 mmHg significantly upregulated RhoA, ROCK1, and ROCK2 in 1-6 h, and dramatically enhanced the expression of marker molecules on "stemness", such as OCT4, SOX2, KLF4, MYC, NANOG, and CD133 in 6-72 h. However, all these changes in hepatocytes induced by -50 mmHg stimulation were almost abrogated by ROCK inhibitor Y27623. Our data suggest that an appropriate force of negative pressure stimulation can effectively induce the dedifferentiation of hepatocytes via RhoA/ROCK pathway activation.


Asunto(s)
Desdiferenciación Celular , Hepatocitos , Proteína de Unión al GTP rhoA , Humanos , Hepatocitos/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Transducción de Señal , Desdiferenciación Celular/genética , Desdiferenciación Celular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA